IBM vs. Google and the Race to Quantum Supremacy – Citizen Truth

Though IBM contests Googles claim of quantum supremacy, it concedes that it passed an important milestone. For the science of computing, that is all that matters.

Googles quantum supremacy claim has now been disputed by its close competitor IBM. Not because Googles Sycamore quantum computers calculations are wrong, but because Google had underestimated what IBMs Summit, the most powerful supercomputer in the world, could do. Meanwhile, Googles paper, which had accidentally been leaked by a NASA researcher, has now been published in the prestigious science journal Nature. Googles claims are official now, and therefore can be examined in the way any new science claim should be examined: skeptically until all the doubts are addressed.

Previously, I have covered what quantum computing is, and in this article, I will move on to the key issue of quantum supremacy, the claim that IBM has challenged and what it really means. IBM concedes that Google has achieved an important milestone, but does not accept that it has achieved quantum supremacy.

IBM refuted Googles claim around the same time as Googles Nature paper was published. Google had claimed that IBMs supercomputer, Summit, would take 10,000 years to solve the problem Googles Sycamore had solved in a mere 200 seconds. IBM showed that Summit, with clever programming and using its huge disk space, could actually solve the problem in only 2.5 days. Sycamore still beat Summit on this specific problem by solving it 1,100 times faster, but not 157 million times faster, as Google had claimed. According to IBM, this does not establish quantum supremacy as that requires solving a problem a conventional computer cannot solve in a reasonable amount of time. Two and a half days is reasonable, thereforeaccording to IBMquantum supremacy is yet to be attained.

The original definition of quantum supremacy was given by John Preskill, on which he now has second thoughts. Recently he wrote, supremacy, through its association with white supremacy, evokes a repugnant political stance. The other reason is that the word exacerbates the already overhyped reporting on the status of quantum technology.

Regarding IBMs claim that quantum supremacy has not yet been achieved, Scott Aaronson, a leading quantum computing scientist, wrote that though Google should have foreseen what IBM has done, it does not invalidate Googles claim. The key issue is not that Summit had a special way to solve the specific quantum problem Google had chosen, but that Summit cannot scale: if Googles Sycamore goes from 53 to 60 qubits, IBM will require 33 Summits; if to 70 Qubits, a supercomputer the size of a city!

Why does Summit have to increase at this rate to match Sycamores extra qubits? To demonstrate quantum supremacy, Google chose the simulation of quantum circuits, which is similar to generating a sequence of truly random numbers. Classical computers can produce numbers that appear to be random, but it is a matter of time before they will repeat the sequence.

The resourcesdisk space, memory, computing powerclassical computers require to solve this problem, in a reasonable time, increase exponentially with the size of the problem. For quantum computers, adding qubits linearlymeaning, simply adding more qubitsincreases computing capacity exponentially. Therefore, just 7 extra qubits of Sycamore means IBM needs to increase the size of Summit 33 times. A 17-qubit increase of Sycamore needs Summit to increase by thousands of times. This is the key difference between Summit and Sycamore. For each extra qubit, a conventional computer will have to scale its resources exponentially, and this is a losing game for the conventional computer.

We have to give Google the victory here, not because IBM is wrong, but because the principle of quantum supremacy, that a quantum computer can work as designed, solve a specific problem, and beat a conventional computer in computational time has been established. The actual demonstrationa more precise definition of reasonable time and its physical demonstrationis only of academic value. If 53 qubits can solve the problem, but with IBMs Summit still in the race, even if much slower, it is just a matter of time before it is well and truly beaten.

Of course, there are other ways that this particular test could fail. A new algorithm can be discovered that solves this problem faster, starting a fresh race. But the principle here is not a specific race but the way quantum computing will scale in solving a certain class of problems that classical or conventional computers cannot.

For problems that do not increase exponentially with size, the classical computers work better, are way cheaper, and do not require near absolute zero temperatures that quantum computers require. In other words, classical computers will coexist with quantum computers and not follow typewriters and calculators to the technology graveyards.

The key issue in creating viable quantum computers should not be confused with a race between classical computers and the new kid on the block. If we see the race as between two classes of computers only in terms of solving a specific problem, we are missing the big picture. It is simply that for classical computers, the solution time for a certain class of problems increases exponentially with the size of the problem, and beyond a certain size, we just cant solve them in any reasonable time. Quantum computers have the potential to solve such large problems requiring exponential computing power. This opens a way to solve these classes of problems other than the iffy route of finding new algorithms.

Are there such problems, and will they yield worthwhile technological applications? The Google problem, computing the future states of quantum circuits, was not chosen for any practical application. It was simply chosen to showcase quantum supremacy, defined as a quantum computer solving a problem that a classical computer cannot solve in a reasonable time.

Recently, a Chinese team led by Pan Jianwei has published a paper that shows another problema Boson sampling experiment with 20 photonscan also be a pathway to show quantum supremacy. Both these problems are constructed not to showcase real-world applications, but simply to show that quantum computing works and can potentially solve real-world problems.

What are the classes of problems that quantum computers can solve? The first are those for which the late Nobel laureate Richard Feynman had postulated quantum computers as a simulation of the quantum world. Why do we need such simulations, after all, we live in the macro-world in which quantum effects are not visible? Though such effects may not visible to us, they are indeed all around us and affect us in different ways.

A number of such phenomena arise out of the interaction of the quantum world with the macro-world. It is now clear that using classical computers we cannot simulate, for instance, protein folding, as it involves the quantum world intersecting with the macro-world. A quantum computer could simulate the probability of how many possible ways such proteins could fold and the likely shapes they could take. This would allow us to build not only new materials but also medicines known as biologics. Biologics are large molecules used for treating cancer and auto-immune diseases. They work due to not only their composition but also their shapes. If we could work out their shapes, we could identify new proteinsor new biological drug targets; or complex new chemicals for developing new materials. The other examples are solving real-life combinatorial problems such as searching large databases, cracking cryptographic problems, improved medical imaging, etc.

The business worldIBM, Google, Microsoftis gung-ho on the possible use of quantum computers for such applications, and that is why they are all investing in it big time. Nature reported that in 2017 and 2018, at least $450 million was invested by venture capital in quantum computing, more than four times more than the preceding two years. Nation-states, notably the United States and China, are also investing billions of dollars each year.

But what if quantum computers do not lead to commercial benefitsshould we then abandon them? What if they are useful only for simulating quantum mechanics and understanding that world better? Did we build the Hadron Colliderinvesting $13.25 billion, and with an annual running cost of $1 billiononly because we expected discoveries that will have commercial value? Or, should society invest in knowing the fundamental properties of space and time including that of the quantum world? Even if quantum computers only give us a window to the quantum world, the benefits would be knowledge.

What is the price of this knowledge?

This article was produced in partnership by Newsclick and Globetrotter, a project of the Independent Media Institute.

Read more here:

IBM vs. Google and the Race to Quantum Supremacy - Citizen Truth

Microprotein ID’d Affecting Protein Folding and Cell Stress Linked to Diseases Like Huntington’s, Study Finds – Huntington’s Disease News

PIGBOS a newly discovered mitochondrial microprotein involved in a cellular stress-response mechanism called unfolded protein response (UPR) might be a treatment target for neurodegenerative diseases likeHuntingtons, a study suggests.

The study, Regulation of the ER stress response by a mitochondrial microprotein, was published in the journal Nature Communications.

Maintenance of protein balance including the production, shaping (folding), and degradation of proteins is essential for a cells function and survival.

Dysfunction in protein balance has been associated with the build-up of toxic protein aggregates and the development of neurodegenerative diseases, including Alzheimers, Parkinsons, and Huntingtons disease.

The endoplasmic reticulum (ER) is a key cellular structure in the production, folding, modification, and transport of proteins. Excessive amounts of unfolded or misfolded proteins (proteins with abnormal 3D structures) in the ER results in ER stress, and the activation of the unfolded protein response (UPR) stress response mechanism, which acts to mitigate damage caused by this protein build-up.

UPR promotes the reduction of protein production and an increase in protein folding and degradation of unfolded proteins in the ER. If this fails to restore cellular balance and prolongs the activation of UPR, cell death is induced.

UPR dysfunction contributes to accumulation of key disease-related proteins, and thus plays an essential role in the [development] of many neurodegenerative disorders, including Alzheimers disease, Parkinsons disease, and Huntingtons disease, the researchers wrote.

During UPR, mitochondria the cells powerhouses are known to provide energy for protein folding in the ER and to activate cell death pathways if the cellular balance is not restored. However, how mitochondria and the ER communicate in this context remains unclear.

Researchers at the Salk Institute for Biological Studies, in California, discovered a mitochondrial microprotein, called PIGBOS, that regulates UPR at the sites of contact between mitochondria and the ER.

While the average human protein contains around 300 amino acids (the building blocks of proteins), microproteins have less than 100 amino acids. Microproteins were only recently found to be functional and important in the regulation of several cellular processes.

By conducting protein-binding experiments, the team found that the 54-aminoacidmicroprotein PIGBOS, present in the outer membrane of mitochondria, interacts with a protein called CLCC1 at the ER-mitochondria contact sites.

CLCC1 whose low levels werepreviously associated with increased UPR and neurodegeneration is found at the portion of the ER that contacts the mitochondria, called mitochondria-associated ER membrane.

Further analyses showed that inducing ER stress in cells genetically modified to lack CLCC1 or PIGBOS increased the levels of UPR-related proteins, while the opposite effect was observed in cells overproducing PIGBOS. Lower levels of PIGBOS were also associated with greater cell death.

Researchers noted that these findings suggest that loss of PIGBOS increases cellular sensitivity to ER stress, which in turn increases [cell death] and links PIGBOS levels to the ability of cells to survive stress, emphasizing that modulating PIGBOS levels can in turn modulate cellular sensitivity towards ER stress.

Results also showed that PIGBOSs UPR regulation is dependent on its interaction with CLCC1, and that modulating the number of ER-mitochondria contacts regulates the levels of PIGBOS-CLCC1 interactions.

These data identified PIGBOS as a [previously] unknown mitochondrial regulator of UPR, and the only known microprotein linked to the regulation of cell stress or inter-organelle signaling, the team emphasized.

These findings may help in developing treatment approaches targeting ER stress and cell death.

Given the importance of UPR in biology and disease, future studies on PIGBOSs role in UPR should afford additional insights and may provide methods for regulating this pathway for therapeutic applications, the researchers concluded.

Total Posts: 79

Ana holds a PhD in Immunology from the University of Lisbon and worked as a postdoctoral researcher at Instituto de Medicina Molecular (iMM) in Lisbon, Portugal. She graduated with a BSc in Genetics from the University of Newcastle and received a Masters in Biomolecular Archaeology from the University of Manchester, England. After leaving the lab to pursue a career in Science Communication, she served as the Director of Science Communication at iMM.

Excerpt from:

Microprotein ID'd Affecting Protein Folding and Cell Stress Linked to Diseases Like Huntington's, Study Finds - Huntington's Disease News

The Science Behind Foldit | Foldit

Foldit is a revolutionary crowdsourcing computer game enabling you to contribute to important scientific research. This page describes the science behind Foldit and how your playing can help.

What is a protein? Proteins are the workhorses in every cell of every living thing. Your body is made up of trillions of cells, of all different kinds: muscle cells, brain cells, blood cells, and more. Inside those cells, proteins are allowing your body to do what it does: break down food to power your muscles, send signals through your brain that control the body, and transport nutrients through your blood. Proteins come in thousands of different varieties, but they all have a lot in common. For instance, they're made of the same stuff: every protein consists of a long chain of joined-together amino acids.

What are amino acids? Amino acids are small molecules made up of atoms of carbon, oxygen, nitrogen, sulfur, and hydrogen. To make a protein, the amino acids are joined in an unbranched chain, like a line of people holding hands. Just as the line of people has their legs and feet "hanging" off the chain, each amino acid has a small group of atoms (called a sidechain) sticking off the main chain (backbone) that connects them all together. There are 20 different kinds of amino acids, which differ from one another based on what atoms are in their sidechains. These 20 amino acids fall into different groups based on their chemical properties: acidic or alkaline, hydrophilic (water-loving) or hydrophobic (greasy).

What shape will a protein fold into? Even though proteins are just a long chain of amino acids, they don't like to stay stretched out in a straight line. The protein folds up to make a compact blob, but as it does, it keeps some amino acids near the center of the blob, and others outside; and it keeps some pairs of amino acids close together and others far apart. Every kind of protein folds up into a very specific shape -- the same shape every time. Most proteins do this all by themselves, although some need extra help to fold into the right shape. The unique shape of a particular protein is the most stable state it can adopt. Picture a ball at the top of a hill -- the ball will always roll down to the bottom. If you try to put the ball back on top it will still roll down to the bottom of the hill because that is where it is most stable.

Why is shape important? This structure specifies the function of the protein. For example, a protein that breaks down glucose so the cell can use the energy stored in the sugar will have a shape that recognizes the glucose and binds to it (like a lock and key) and chemically reactive amino acids that will react with the glucose and break it down to release the energy.

What do proteins do? Proteins are involved in almost all of the processes going on inside your body: they break down food to power your muscles, send signals through your brain that control the body, and transport nutrients through your blood. Many proteins act as enzymes, meaning they catalyze (speed up) chemical reactions that wouldn't take place otherwise. But other proteins power muscle contractions, or act as chemical messages inside the body, or hundreds of other things. Here's a small sample of what proteins do:

Proteins are present in all living things, even plants, bacteria, and viruses. Some organisms have proteins that give them their special characteristics:

You can find more information on the rules of protein folding in our FAQ.

What big problems is this game tackling?

How does my game playing contribute to curing diseases?

With all the things proteins do to keep our bodies functioning and healthy, they can be involved in disease in many different ways. The more we know about how certain proteins fold, the better new proteins we can design to combat the disease-related proteins and cure the diseases. Below, we list three diseases that represent different ways that proteins can be involved in disease.

What other good stuff am I contributing to by playing?

Proteins are found in all living things, including plants. Certain types of plants are grown and converted to biofuel, but the conversion process is not as fast and efficient as it could be. A critical step in turning plants into fuel is breaking down the plant material, which is currently done by microbial enzymes (proteins) called "cellulases". Perhaps we can find new proteins to do it better.

Can humans really help computers fold proteins?

Were collecting data to find out if humans' pattern-recognition and puzzle-solving abilities make them more efficient than existing computer programs at pattern-folding tasks. If this turns out to be true, we can then teach human strategies to computers and fold proteins faster than ever!

You can find more information about the goals of the project in our FAQ.

Brian Koepnick, Jeff Flatten, Tamir Husain, Alex Ford, Daniel-Adriano Silva, Matthew J. Bick, Aaron Bauer, Gaohua Liu, Yojiro Ishida, Alexander Boykov, Roger D. Estep, Susan Kleinfelter, Toke Nrgrd-Solano, Linda Wei, Foldit Players, Gaetano T. Montelione, Frank DiMaio, Zoran Popovi, Firas Khatib, Seth Cooper and David Baker. De novo protein design by citizen scientists Nature (2019). [link]

Thomas Muender, Sadaab Ali Gulani, Lauren Westendorf, Clarissa Verish, Rainer Malaka, Orit Shaer and Seth Cooper.Comparison of mouse and multi-touch for protein structure manipulation in a citizen science game interface.Journal of Science Communication (2019). [link]

Lorna Dsilva, Shubhi Mittal, Brian Koepnick, Jeff Flatten, Seth Cooper and Scott Horowitz.Creating custom Foldit puzzles for teaching biochemistry.Biochemistry and Molecular Biology Education (2019). [link]

Seth Cooper, Amy L. R. Sterling, Robert Kleffner, William M. Silversmith and Justin B. Siegel.Repurposing citizen science games as software tools for professional scientists.Proceedings of the 13th International Conference on the Foundations of Digital Games (2018). [link]

Robert Kleffner, Jeff Flatten, Andrew Leaver-Fay, David Baker, Justin B. Siegel, Firas Khatib and Seth Cooper. Foldit Standalone: a video game-derived protein structure manipulation interface using Rosetta. Bioinformatics (2017). [link]

Jacqueline Gaston and Seth Cooper. To three or not to three: improving human computation game onboarding with a three-star system. Proceedings of the SIGCHI Conference on Human Factors in Computing Systems (2017). [link]

Scott Horowitz, Brian Koepnick, Raoul Martin, Agnes Tymieniecki, Amanda A. Winburn, Seth Cooper, Jeff Flatten, David S. Rogawski, Nicole M. Koropatkin, Tsinatkeab T. Hailu, Neha Jain, Philipp Koldewey, Logan S. Ahlstrom, Matthew R. Chapman, Andrew P. Sikkema, Meredith A. Skiba, Finn P. Maloney, Felix R. M. Beinlich, Foldit Players, University of Michigan students, Zoran Popovi, David Baker, Firas Khatib and James C. A. Bardwell. Determining crystal structures through crowdsourcing and coursework. Nature Communications 7, Article number: 12549 (2016). [link]

Dun-Yu Hsiao, Min Sun, Christy Ballweber, Seth Cooper and Zoran Popovi. Proactive sensing for improving hand pose estimation. Proceedings of the SIGCHI Conference on Human Factors in Computing Systems (2016). [link]

Dun-Yu Hsiao, Seth Cooper, Christy Ballweber and Zoran Popovi. User behavior transformation through dynamic input mappings. Proceedings of the 9th International Conference on the Foundations of Digital Games (2014). [link]

George A. Khoury, Adam Liwo, Firas Khatib, Hongyi Zhou, Gaurav Chopra, Jaume Bacardit, Leandro O. Bortot, Rodrigo A. Faccioli, Xin Deng, Yi He, Pawel Krupa, Jilong Li, Magdalena A. Mozolewska, Adam K. Sieradzan, James Smadbeck, Tomasz Wirecki, Seth Cooper, Jeff Flatten, Kefan Xu, David Baker, Jianlin Cheng, Alexandre C. B. Delbem, Christodoulos A. Floudas, Chen Keasar, Michael Levitt, Zoran Popovi, Harold A. Scheraga, Jeffrey Skolnick, Silvia N. Crivelli and Foldit Players. WeFold: a coopetition for protein structure prediction. Proteins (2014). [link]

Christopher B. Eiben, Justin B. Siegel, Jacob B. Bale, Seth Cooper, Firas Khatib, Betty W. Shen, Foldit Players, Barry L. Stoddard, Zoran Popovi and David Baker. Increased Diels-Alderase activity through backbone remodeling guided by Foldit players. Nature Biotechnology (2012). [link]

Erik Andersen, Eleanor O'Rourke, Yun-En Liu, Richard Snider, Jeff Lowdermilk, David Truong, Seth Cooper and Zoran Popovi. The impact of tutorials on games of varying complexity. Proceedings of the SIGCHI Conference on Human Factors in Computing Systems (2012). [link]

Firas Khatib, Seth Cooper, Michael D. Tyka, Kefan Xu, Ilya Makedon, Zoran Popovi, David Baker and Foldit Players. Algorithm discovery by protein folding game players. Proceedings of the National Academy of Sciences of the United States of America (2011). [link]

Miroslaw Gilski, Maciej Kazmierczyk, Szymon Krzywda, Helena Zbransk, Seth Cooper, Zoran Popovi, Firas Khatib, Frank DiMaio, James Thompson, David Baker, Iva Pichov and Mariusz Jaskolskia. High-resolution structure of a retroviral protease folded as a monomer. Acta Crystallographica (2011). [link]

Firas Khatib, Frank DiMaio, Foldit Contenders Group, Foldit Void Crushers Group, Seth Cooper, Maciej Kazmierczyk, Miroslaw Gilski, Szymon Krzywda, Helena Zbransk, Iva Pichov, James Thompson, Zoran Popovi, Mariusz Jaskolski and David Baker. Crystal structure of a monomeric retroviral protease solved by protein folding game players. Nature Structural and Molecular Biology (2011). [link]

Seth Cooper, Firas Khatib, Ilya Makedon, Hao Lu, Janos Barbero, David Baker, James Fogarty, Zoran Popovi and Foldit Players. Analysis of social gameplay macros in the Foldit cookbook. Proceedings of the 6th International Conference on the Foundations of Digital Games (2011). [link]

Seth Cooper, Firas Khatib, Adrien Treuille, Janos Barbero, Jeehyung Lee, Michael Beenen, Andrew Leaver-Fay, David Baker, Zoran Popovi and Foldit Players. Predicting protein structures with a multiplayer online game. Nature (2010). [link]

Seth Cooper, Adrien Treuille, Janos Barbero, Andrew Leaver-Fay, Kathleen Tuite, Firas Khatib, Alex Cho Snyder, Michael Beenen, David Salesin, David Baker, Zoran Popovi and Foldit players. The challenge of designing scientific discovery games. Proceedings of the 5th International Conference on the Foundations of Digital Games (2010). [link]

Foldit has been in dozens of publications over the years - to list them all would take a page of their own. For a sampling, please see our Center for Game Science page.

Check out the Rosetta@Home Screensaver to see how computers fold proteins using distributed computing.

Thank you for using Foldit in your classroom! We have put together a set of instructions to assist you in setting up your students to play Foldit.

You can find the researchers and supporters associated with this study on the game's credits page.

Continued here:

The Science Behind Foldit | Foldit

UCI vision scientist Krzysztof Palczewski elected to National Academy of Medicine – UCI News

Irvine, Calif., Oct. 21, 2019 Krzysztof Palczewski, the Irving H. Leopold Chair in Ophthalmology and a professor of physiology & biophysics at the University of California, Irvine, has been elected to the National Academy of Medicine, one of the highest distinctions accorded to professionals in the medical sciences, healthcare and public health. He is one of 100 new U.S.-based members announced today.

The National Academy of Medicine recognizes leaders in diverse fields including health and medicine; the natural, social and behavioral sciences; and beyond. Through its domestic and global initiatives, the academy works to address critical issues in health, medicine and related policy and inspire positive action across sectors.

Congratulations to Dr. Palczewskion this exceptional achievement which illustrates the academic excellence of UCI faculty,said Enrique Lavernia, UCI provost and executive vice chancellor. With the election of Dr. Palczewski to the National Academy of Medicine, UCI is now home to42 members of the National Academies of Sciences, Engineering and Medicine, 35 members of the American Academy of Arts & Sciences, nine members of the National Academy of Inventors, and four members of the National Academy of Education.

I feel deeply honored by the National Academy of Medicine election, Palczewski said. Such recognition reflects on our colleagues, collaborators and trainees who contributed to impactful research on eye diseases. Clearly, this distinction further encourages us to give our very best efforts in the next stage of our research: developing therapeutics against blinding diseases.

The internationally renowned chemist, pharmacologist and vision scientist has made critical additions to the understanding of the molecular basis of age-related macular degeneration and inherited retinal degeneration, illuminating the path toward the creation of new vision treatments.

Palczewski has studied the pharmacology of vision for more than 30 years, and his work has had a tremendous impact on efforts to restore vision in people suffering from retinitis pigmentosa and other congenital mutations that result in blindness.

He is best known for discovering the structure, folding and binding properties of rhodopsin, a light-sensitive photoreceptor protein. His findings profoundly increased comprehension of the molecular basis of vision and the structure of photoreceptor cells in the retina. They also contributed to the ability to originate new molecular therapies for age-related macular degeneration and other retinopathies.

Palczewski came to UCI last year from Case Western Reserve University in Cleveland to establish the Center for Translational Vision Research at the Gavin Herbert Eye Institute, which is part of the UCI School of Medicine. There, he collaborates with a team of noted vision scientists to maximize opportunities to translate insights from basic science investigations into clinical treatments.

He holds 29 issued and nine pending patents and has received several prestigious accolades, including the 2015 Bressler Prize in Vision Science and the inaugural 2014 Beckman-Argyros Award in Vision Research.

In addition, Palczewski is the only person to have won both the Cogan Award (1996) for the most promising young vision scientist and the Friedenwald Award (2014) for continuously outstanding ophthalmology research from the Association for Research in Vision and Ophthalmology. His work has been cited more than 46,000 times, with an h-index impact factor of 115, according to Google Scholar.

Palczewski earned a Ph.D. in biochemistry at the Wrocaw University of Science and Technology in Poland.

About the University of California, Irvine: Founded in 1965, UCI is the youngest member of the prestigious Association of American Universities. The campus has produced three Nobel laureates and is known for its academic achievement, premier research, innovation and anteater mascot. Led by Chancellor Howard Gillman, UCI has more than 36,000 students and offers 222 degree programs. Its located in one of the worlds safest and most economically vibrant communities and is Orange Countys second-largest employer, contributing $5 billion annually to the local economy. For more on UCI, visit http://www.uci.edu.

Media access: Radio programs/stations may, for a fee, use an on-campus ISDN line to interview UCI faculty and experts, subject to availability and university approval. For more UCI news, visit news.uci.edu. Additional resources for journalists may be found at communications.uci.edu/for-journalists.

More here:

UCI vision scientist Krzysztof Palczewski elected to National Academy of Medicine - UCI News

Discover: Science is often wrong and that’s actually a really good thing – Sudbury.com

Im a geneticist. I study the connection between information and biology essentially what makes a fly a fly, and a human a human. Interestingly, were not that different. Its a fantastic job and I know, more or less, how lucky I am to have it.

Ive been a professional geneticist since the early 1990s. Im reasonably good at this, and my research group has done some really good work over the years. But one of the challenges of the job is coming to grips with the idea that much of what we think we know is, in fact, wrong.

Sometimes, were just off a little, and the whole point of a set of experiments is simply trying to do a little better, to get a little closer to the answer. At some point, though, in some aspect of what we do, its likely that were just flat out wrong. And thats okay. The trick is being open-minded enough, hopefully, to see that someday, and then to make the change.

One of the amazing things about being a modern geneticist is that, generally speaking, people have some idea of what I do: work on DNA (deoxyribonucleic acid). When I ask a group of school kids what a gene is, the most common answer is DNA. And this is true, with some interesting exceptions. Genes are DNA and DNA is the information in biology.

For almost 100 years, biologists were certain that the information in biology was found in proteins and not DNA, and there were geneticists who went to the grave certain of this. How they got it wrong is an interesting story.

Genetics, microscopy (actually creating the first microscopes), and biochemistry were all developing together in the late 1800s. Not surprisingly, one of the earliest questions that fascinated biologists was how information was carried from generation to generation. Offspring look like their parents, but why? Why your second daughter looks like the postman is a question that came up later.

Early cell biologists were using the new microscopes to peer into the cell in ways that simply hadnt been possible previously. They were finding thread-like structures in the interior of cells that passed from generation to generation, were similar within a species, but different between them. We now know these threads as chromosomes. Could these hold the information that scientists were looking for?

Advances in biochemistry paralleled those in microscopy and early geneticists determined that chromosomes were primarily made up of two types of molecules: proteins and DNA. Both are long polymers (chains) made up of repeated monomers (links in the chains). It seemed very reasonable that these chains could contain the information of biological complexity.

By analogy, think of a word as just a string of letters, a sentence as a chain of words, and a paragraph as a chain of sentences. We can think of chromosomes, then, as chapters, and all of our genetic information what we now call our genome (all our genetic material) as these chapters that make up a novel. The question to those early geneticists, then, was: Which string made up the novel? Was it protein or DNA?

You and I know the answer: DNA. Early geneticists, however, got it wrong and then passionately defended this wrong stance for eight decades. Why? The answer is simple. Protein is complicated. DNA is simple. Life is complicated. The alphabet of life, then, should be complicated and protein fits that.

Proteins are made up of 20 amino acids there are 20 different kinds of links in the protein chain. DNA is made up of only four nucleotides there are only four different links in the DNA chain. Given the choice between a complicated alphabet and a simple one, the reasonable choice was the complicated one, namely protein. But, biology doesnt always follow the obvious path and the genetic material was, and is, DNA.

It took decades of experiments to disprove conventional wisdom and convince most people that biological information was in DNA. For some, it took James Watson and Francis Crick (http://www.pbs.org/wgbh/aso/databank/entries/do53dn.html), using data misappropriated from Rosalind Franklin https://www.nature.com/scitable/topicpage/rosalind-franklin-a-crucial-contribution-6538012/), deciphering the structure of DNA in 1953 to drive the nail in the protein coffin. It just seemed to obvious that protein, with all its complexity, would be the molecule that coded for complexity.

These were some of the most accomplished and thoughtful scientists of their day, but they got it wrong. And thats okay if we learn from their mistakes.

It is too easy to dismiss this example as the foolishness of the past. We wouldnt make this kind of mistake today, would we? I cant answer that, but let me give you another example that suggests we would, and Ill argue at the end that we almost certainly are.

Im an American, and one of the challenges of moving to Canada was having to adapt to overcooked burgers (my mother still cant accept that she cant get her burger medium when she visits). This culinary challenge is driven by a phenomenon that one of the more interesting recent cases of scientists having it wrong and refusing to see that.

In the late 1980s, cows started wasting away and, in the late stages of what was slowly recognized as a disease, acting in such bizarre manner that their disease, bovine spongiform encephalitis, became known as Mad Cow Disease. Strikingly, the brains of the cows were full of holes (hence spongiform) and the holes were caked with plaques of proteins clumped together.

Really strikingly, the proteins were ones that are found in healthy brains, but now in an unnatural shape. Proteins are long chains, but they function because they have complex 3D shapes think origami. Proteins fold and fold into specific shapes. But, these proteins found in sick cow brains had a shape not normally seen in nature; they were misfolded.

Sometime after, people started dying from the same symptoms and a connection was made between eating infected cows and contracting the disease (cows could also contract the disease, but likely through saliva or direct contact, and not cannibalism). Researchers also determined the culprit was consumption only of neural tissue, brain and spinal tissue, the very tissue that showed the physical effects of infection (and this is important).

One of the challenges of explaining the disease was the time-course from infection to disease to death; it was long and slow. Diseases, we knew, were transmitted by viruses and bacteria, but no scientist could isolate one that would explain this disease. Further, no one knew of other viruses or bacteria whose infection would take this long to lead to death. For various reasons, people leaned toward assuming a viral cause, and careers and reputations were built on finding the slow virus.

In the late 1980s, a pair of British researchers suggested that perhaps the shape, the folding, of the proteins in the plaques was key. Could the misfolding be causing the clumping that led to the plaques? This proposal was soon championed by Stanely Prusiner, a young scientist early in his career.

The idea was simple. The misfolded protein was itself both the result and the cause of the infection. Misfolded protein clumped forming plaques that killed the brain tissue they also caused correctly folded versions of the proteins to misfold. The concept was straightforward, but completely heretical. Disease, we knew, did not work that way. Diseases are transmitted by viruses or bacteria, but the information is transmitted as DNA (and, rarely, RNA, a closely related molecule). Disease is not transmitted in protein folding (although in 1963 Kurt Vonnegut had predicted such a model for world-destroying ice formation in his amazing book Cats Cradle).

For holding this protein-based view of infection, Prusiner was literally and metaphorically shouted out of the room. Then he showed, experimentally and elegantly, that misfolded proteins, which he called prions, were the cause of these diseases, of both symptoms and infection.

For this accomplishment, he was awarded the 1997 Nobel Prize in Medicine. He, and others, were right. Science, with a big S, was wrong. And thats okay. We now know that prions are responsible for a series of diseases in humans and other animals, including Chronic Wasting Disease, the spread of which poses a serious threat to deer and elk here in Ontario.

Circling back, the overcooked burger phenomenon is because of these proteins. If you heat the prions sufficiently, they lose their unnatural shape all shape actually and the beef is safe to eat. A well-done burger will guarantee no infectious prions, while a medium one will not. We dont have this issue in the U.S. because cows south of the border are less likely to have been infected with the prions than their northern counterparts (or at least Americans are willing to pretend this is the case).

Where does this leave us? To me, the take-home message is that we need to remain skeptical, but curious. Examine the world around you with curious eyes, and be ready to challenge and question your assumptions.

Also, dont ignore the massive things in front of your eyes simply because they dont fit your understanding of, or wishes for, the world around you. Climate change, for example, is real and will likely make this a more difficult world for our children. Ive spent a lot of time in my career putting together models of how the biological world works, but I know pieces of these models are wrong.

I can almost guarantee you that I have something as fundamentally wrong as those early geneticists stuck on protein as the genetic material of cells or the prion-deniers; I just dont know what it is. Yet.

And, this situation is okay. The important thing isnt to be right. Instead, it is to be open to seeing when you are wrong.

Dr. Thomas Merritt is the Canada Research Chair in Genomics and Bioinformatics at Laurentian University.

See original here:

Discover: Science is often wrong and that's actually a really good thing - Sudbury.com

Antibiotics with novel mechanism of action discovered – Drug Target Review

A new family of synthetic antibiotics that possess broad anti-Gram-negative antimicrobial activity has been discovered.

Researchers have reported the discovery and characterisation of a new family of synthetic antibiotics that possess broad-spectrum anti-Gram-negative antimicrobial activity.

The research teams were headed by the University of Zurich (UZH) and Polyphor AG, both Switzerland.

The new antibiotics interact with essential outer membrane proteins in Gram-negative bacteria, said John Robinson from the UZH Department of Chemistry, who co-led the study. According to our results, the antibiotics bind to complex fat-like substances called lipopolysaccharides and to BamA, an essential protein of the outer membrane of Gram-negative bacteria.

E. coli cells treated with a novel chimeric peptidomimetic antibiotic. Cells in blue are alive while green cells are already killed by the peptidomimetic. As the antibiotic destroys the integrity of the bacterial membranes, the scientists observed explosive cell lysis (cells indicated by arrows), which leads in the release of DNA (diffuse green) (credit: Matthias Urfer, UZH).

BamA is the main component of the so-called -barrel folding complex (BAM), which is essential for outer membrane synthesis. After targeting this essential outer membrane protein, the antibiotics destroy the integrity of the bacterial membranes and the cells burst.

The outer membrane of Gram-negative bacteria protects the cells from toxic environmental factors, such as antibiotics. It is also responsible for the uptake and export of nutrients and signalling molecules. Despite its critical importance, so far no clinical antibiotics target these key proteins required for outer membrane biogenesis, Robinson continued.

The plan now is to progress one compound into human clinical trials. POL7306, a first lead molecule of the novel antibiotics class, is now in pre-clinical development, added Daniel Obrecht, chief scientific officer at Polyphor and co-head of the work.

Continued here:

Antibiotics with novel mechanism of action discovered - Drug Target Review

The top AI lighthouse projects to watch in biopharma – FierceBiotech

So-called lighthouse projectsare typically defined as smallefforts focused on deliverables ina narrow area, designed to establish a pathway for larger enterprises down the line. But as the biopharma industrys digital transformation continues apace, with the rapid acceptance of tools such as artificial intelligence and machine learning, certain projects promise to yield impacts on a much wider scale.

At the same time, AI expertise is being quickly diffused throughout the industry through collaborations, with numerous tech firms offering different approaches and services as drug and device makers shop around for those that best suit their needs.

In just the past few years, biopharma has built a large, interconnected network of partnerships as it aims to transform and digitize its processes to maximize their valueacross everything from molecule design to clinical trial planning, as well as in supply chain, quality control and sales strategies.

In a survey released earlier this month by Optum of 500 healthcare industry leaders and professionals, the number of respondents who said their organizations had an AI implementation strategy in place had increased by nearly 88% compared to the year before.

Additionally, the survey found that organizations plan to spend about $40 million apiece on average on AI-related projects over the next five yearsand half of respondents said they expect to see positive returns on AI investments in three years or less.

Its encouraging to see executives growing trust in, and adoption of, AI to make data more actionable in making the health system work better for everyone, Optum President and COO Dan Schumacher said in a statement, although higher levels of trust in AI were seen in regards to administrative applications compared to clinical ones, and the automation of business processes was ranked higher as a priority.

RELATED: Novartis to put AI on every employee's desk through Microsoft partnership

Still, this sets the stage for potentially rapid adoptions in R&D and care delivery as new methods are validated and become available.

Forming collaborations and partnerships will be paramount, as many companies lack the expertise needed to make the transformation on their own in the coming years. Among medtech companies specifically, a report from Deloitte predicts that the current illness-focused system will be completely overhauled by 2040 and replaced by a proactive one that integrates data to personalize a continuum of care spanning before and after a procedure.

One of the ripest sectors for advancement is in digital pathology and assisting diagnosis. Many research projects aim to use machine learning processes to spot the patterns of diseases or conditions in images or scans. This can help ease the burden on hospital departments by screening chest X-rays, MRI scans, tissue slides or pictures of the eye.

In April 2018, the FDA approved the first medical device in the U.S. to use artificial intelligence to detect cases of diabetic retinopathy, the most common cause of vision loss among people with diabetes. Using digitalimages uploaded from a retinal camera, the AIdubbed IDx-DRcan detect those with more than a mild case of the disease and refer them to a healthcare professional.

RELATED: AstraZeneca enlists artificial intelligence for sales rep coaching

The benefits of automating healthcare and research processes are similar in other areas of medicinenot just in mining data for insightbut also in sharing those insights.

Take the MELLODDY project, for example: short for Machine Learning Ledger Orchestration for Drug Discovery, the initiative hopes to share preclinical data among a network of Big Pharma companies and research partners using a blockchain-based infrastructure to protect confidentiality and proprietary information.

Meanwhile, the newest center of the NIH is working to build a universal translator for medical data, with the goal of bringing together researchers from different fields across the healthcare enterpriseon top of redefining our current definitions of disease based on the findings.

Machine learning can also spot patterns in a flood of data from multiple sources, where certain changes, no matter how small, can herald the onset of Alzheimers disease and dementia. Evidation Health, along with Apple and Eli Lilly, is looking to develop digital biomarkers for neurodegenerative disease by tracking peoples daily routines, device usage and changes in speech.

Using all the information provided by patients from different angles, and then feeding that data back into care delivery to potentially improve outcomes, is where machine learning can excel, and Verb Surgicalthe joint venture between Verily and Johnson & Johnsonaims to use those tenets to drive a new generation of surgery.

RELATED: FDA delivers regulatory guidance on AI software and clinical decision-making aids

Elsewherein the Google/Alphabet sphere, DeepMind has developed an AI program to take on one of the most challenging problems in medical science: predicting protein folding, a mathematical problem that reaches a spectacular number of possibilities. The field once relied on human intuition to solve the puzzle. Now were teaching machines to follow similar instincts.

But the biggest changes will come when these methods, tools and knowledge can be made widely available, and the public-private ATOM consortium is looking to do just that. Spun out of the U.S. governments cancer moonshot efforts, the initiative by GlaxoSmithKline, UC San Francisco and federal research laboratories hosts a series of research projects aimed at accelerating preclinical development to a timeline under one year.

This small handful of projects hopes to have an outsized impactand serve as a beacon for the industry as a whole. Read on below. Conor Hale (email | Twitter)

Read more here:

The top AI lighthouse projects to watch in biopharma - FierceBiotech

RNA Folding Insights Lead to New Therapeutics and Synthetic Biology Technologies – Technology Networks

A Northwestern Engineering research team led by Professor Julius Lucks has uncovered a new understanding of how RNA molecules act as cellular 'biosensors' to monitor and respond to changes in the environment by controlling gene expression. The findings could impact the design of future RNA-specific therapeutics as well as new synthetic biology tools that measure the presence of toxins in the environment.

RNA molecules play a pivotal role in storing and propagating genetic information like DNA, as well as performing functions critical to living systems like proteins. At the core of its function is its ability to undergo origami-style folding into intricate shapes inside the cell.

Using high-throughput next-generation sequencing technology developed in his lab that chemically images the dynamic shapes RNAs fold into, Lucks found similarities in the folding tendencies among a family of RNA molecules, called riboswitches. Riboswitches act as natural biosensors to monitor the internal and external state of cells. When a riboswitch binds to a molecule, it changes its shape, causing a change in gene expression.

"These riboswitches have evolved to fold into very specific shapes so they can recognize other compounds, change their shape when they bind to them, and ultimately induce a change in gene expression," said Lucks, associate chair and professor of chemical and biological engineering at the McCormick School of Engineering. "There's been little studied about how exactly they can fold and adjust those shapes, especially since they do so before the RNAs are fully made. We learned that there is an evolutionary pressure on RNAs to not only fold into the final structure, but to have a pathway to do so similarly and efficiently."

A paper outlining the work, titled "A Ligand Gated Strand Displacement Mechanism for ZTP Riboswitch Transcription Control," was published on October 21 in the journalNature Chemical Biology. The study was also featured in the journal's "News & Views" section.

Lucks served as the paper's corresponding author, while Eric Strobel, a Beckman Postdoctoral Fellow in Lucks's group, served as the study's lead author. PhD students Katherine Berman and Luyi Cheng, and visiting predoctoral scholar Paul Carlson, all from the Lucks Lab, also contributed to the research.

The study builds on past research in which Lucks and his team developed a platform that provides super high-resolution representations of RNA shape changing as the RNAs are synthesized.

Finding Folding SimilaritiesPreviously, Lucks and his team used their high-resolution system to study how a riboswitch sensed the fluoride ion. In theNature Chemical Biologypaper, he applied the system to a riboswitch responsible for sensing a natural cellular alarmone molecule called ZTP, which Lucks said functions as an "alarm trigger" in cells.

Despite structural and functional differences between the riboswitches and their respective target compounds, Lucks discovered that in both instances the riboswitches followed the same folding pathway -- the series of shapes the RNA molecule progresses through as it is synthesized.

Using high-throughput next-generation sequencing technology, Professor Julius Lucks found similarities in the folding tendencies among a family of RNA molecules, called riboswitches.

"Once RNAs are made, they immediately fold into a shape that recognizes the molecule. If the molecule is there, the shape locks in and preserves the structure," Lucks said. "If the molecule isn't present, the RNA unravels itself. We found that happened in both instances.

"Whether you're trying to make an origami crane or frog, the first several steps are pretty much the same," he added. "While these RNAs look different, they're amazingly similar when you break them down into their sequence of folding instructions. Finding links to these common features lays the groundwork for coding these principles as design elements for when we want to harness them for our own uses."

Those uses could include future drug delivery strategies. While many therapeutics are designed to treat diseases caused by protein misfolding, such as Alzheimer's or Parkinson's, Lucks believes his lab's work could inform efforts to treat diseases believed to be triggered at the RNA level, including spinal muscular atrophy, a neuromuscular disorder caused by the mis-splicing of the SMN gene.

"You may not only want to target the final structure of an RNA molecule, because they all fold in some sort of structure, but also the folding process to get into that structure," he said.

The findings also represent a positive step toward harnessing RNA's capability as a natural biosensor. Working with Northwestern's Center for Synthetic Biology and Center for Water Research, Lucks and his lab are pursuing how riboswitches could be used within low-cost synthetic biology platforms to detect toxins in the environment, impacting areas like crop health and water quality.

"As we learn more about the architecture behind how RNAs work, we'll seek to understand how to make them work better," Lucks said. "Nature may have evolved to make them do one thing, but we want them to work for us faster or more sensitively. We're still learning how to do that, but we're nearing that level of detail where we can truly design around these principles."

Reference: Strobel et al. 2019.A ligand-gated strand displacement mechanism for ZTP riboswitch transcription control. Nature Chemical Biology.DOI: https://doi.org/10.1038/s41589-019-0382-7.

This article has been republished from the following materials. Note: material may have been edited for length and content. For further information, please contact the cited source.

Excerpt from:

RNA Folding Insights Lead to New Therapeutics and Synthetic Biology Technologies - Technology Networks

The Hidden Inactive Ingredient: Biological Products in Recombinant Pharmaceuticals – P&T Community

INTRODUCTION

Pharmaceutical products have long been prepared with various inactive ingredients to enhance pharmacokinetics, stability, and pharmaceutical elegance. In an age where the use of recombinant technologies is rapidly increasing and consumers are more aware of inactive ingredients in food and cosmetic products, there remains a paucity of centralized information to help medical professionals counsel their patients about recombinant technologies. Although patient-specific factors such as race, age, and weight are considered when prescribing and dispensing medications, oftentimes religion and dietary habits are left out of the conversation. The increasing array of biopharmaceuticals on the market and in production creates an opportunity for healthcare providers and patients to enhance their knowledge of these issues. The purpose of this review is to create a centralized resource for healthcare providers that describes some common dietary restrictions of select faith and ethnic groups and to identify FDA-approved medications of biologic origin.

A biopharmaceutical is any active agent biological in nature and manufactured using living organisms.1 Since the 1980s, scientists have been harnessing the power of single-cell organisms to create unique proteins that can be used to treat a wide range of disease states ranging from diabetes to hematologic disorders. The recombinant process enhances scientists ability to modify and manipulate protein structure and function to produce a targeted agent for therapeutic purposes. Now, almost 40 years later, recombinant technologies are on the rise using a wider range of bacterial, fungal, and animal cell lines.1,2

Original biopharmaceuticals were replacement proteins such as insulin, hormones, and antibodies. Advancements in the field have widened the scope of indications to hematologic and oncologic disorders. Recombinant products for use in oncology have been expanding over the past two decades, with a focus on monoclonal antibody production. The area of targeted drug delivery within oncology is a rapidly growing field, with promise for new agents or enhancements to existing agents that can improve antitumor cytotoxicity while reducing adverse effects. As this technology continues to advance, scientists are experimenting with alternative ways to produce biopharmaceuticals by way of transgenic organisms and animal cloning.3

The process of creating a recombinant product involves transferring the specific genes responsible for creating the protein into a bacteria, yeast, or animal cell. Historically, Escherichia coli was the prominent cell line that was used, and even today is the cell line used to manufacturer medications such as insulin and filgrastim. Over time, scientists explored the use of yeast cells, namely Saccharomyces cerevisiae, and other mammalian cells such as Chinese hamster ovary (CHO) cells.

The use of mammalian cell lines has increased in recent years because of the cell systems ability to produce glycosylated proteins.2 Glycosylation is one of the most complex post-translational modifications that occurs to a protein but it offers certain pharmaceutical advantages, such as increased plasma half-life.4 Many antibodies that were formed as a result of recombinant technology combine fragments from two different species. Often, these are combined with human fragments to form chimeric antibodies. Most commonly, murine antibody fragments have been fused with human fragments to produce an antibody that is more human-like. This differs from a humanized antibody, which may contain elements of nonhuman origin but maintains essentially the same amino acid sequence that can be found in the human antibody.5 Irrespective of drug development, a pharmaceutical manufacturers choice of cell line is balanced by the complexity of protein folding and post-translational modifications with a cell line that is easy to maintain and simple to culture.

The nomenclature of recombinant products is regulated by the World Health Organization (WHO)s International Nonproprietary Names (INN). Names awarded an INN are selected by WHO on the advice of experts from the organizations Expert Advisory Panel on the International Pharmacopoeia and Pharmaceutical Preparations.

The product names can assist health practitioners in understanding how the products are manufactured based on the pre-stems and stems of the names.6 It is beyond the scope of our review to discuss all the nomenclature, but monoclonal antibody medications are a good example of this practice. Previously, two infixes denoted the target of the drug as well as the source. In 2015, the INN together with the United States Adopted Name (USAN) Council agreed that to simplify drug naming, the source infix would be dropped.5 The name now consists of a prefix, a target infix, and the suffix -mab (Table 1).

Often the most convenient source of information on how a drug is manufactured is the package insert. However, information about the recombinant process and the cell line from which the pharmaceutical product is made frequently isnt relayed to the patient during counseling. To compile a compendium of medications from animal origin, we undertook an extensive review of all Food and Drug Administration (FDA)-approved medications using a data set of 15,797 entries based on the National Drug Code (NDC) from the FDA website, on December 2, 2016.7 Duplicate entries based on nonproprietary names were deleted. We also initially excluded entries whose marketing category included cosmetic, over-the-counter (OTC) drug monograph, unapproved homeopathic, or unapproved medical gas, and entries whose product type was a standardized or non-standardized allergenic extract. The remaining 666 records we reviewed manually, and we also reviewed package inserts for possible inclusion. Table 2 is organized by the type of cell line used in the manufacturing process, listed alphabetically by generic name.

Additional ingredients involved in the preparation of pharmaceutical products also may be derived from animal products. Gelatin is a substance of bovine or porcine origin that is commonly used for commercially made capsules.8 A WHO memo from 2001 addresses a ruling that allows Muslims to accept gelatin because it has been transformed from its original source,8,9 although not all Muslims accept the ruling. Specialized compounding centers may offer vegetable-based capsules for those who refuse gelatin-based products.

Lactose is a common sugar that is found as a filler in many capsules and tablets and is derived from cows milk. In addition, lactulose can be synthetically produced in large quantities in the laboratory. Lanolin is a fat extracted from sheeps wool that is used as an ingredient and base in many ointments and topical medications. It is important to consider all ingredients within a pharmaceutical preparation, including all the inactive ingredients.8

It is well known within the medical community that Jehovahs Witnesses patients abstain from blood products. This issue often comes to a head when a patient is critically ill and in need of a transfusion. Many hospitals have developed policies and standard procedures for approaching these situations to respect the autonomy of the patient while continuing to offer appropriate and excellent medical care. Although this restriction is widely known, providers might not be so familiar with other, less common restrictions.

Even within a faith or ethnic group there can be differences about dietary restrictions, but having a general sense of what these restrictions are will enable providers to provide a patient-centered care approach. A lack of communication might lead to distrust and decreased patient satisfaction if a patient subsequently learned how a particular medication is processed and handled.

The decision to adhere to strict dietary restrictions is an individual choice. Through conversations with the patient, a provider can identify the personal dietary beliefs that might affect their therapy selection. Certain religious organizations have thus made formal statements to relieve their followers of strict dietary considerations as they relate to medicine and health. For example, Muslims are permitted to ingest gelatin capsules because the product is believed to be transformed in property and in character. Shariah law also allows for the use of insulin products in case of necessity.9 Table 3 lists common religious and faith-based groups that follow specific dietary restrictions.8,1215 It is important to note that within these general classifications, there are also various practices arising from individual beliefs.

There is no consensus on how to deal with patient refusal based on faith or ethnic beliefs. Because adherence to diet that is based on religion is personal to the patient, it requires a personal response from the provider. Oftentimes, a patients refusal to use a specific product can be met with misunderstanding and frustration by the provider. Sometimes a conversation about the risks and benefits can persuade the patient to adhere to a medication while at other times, frank refusal can lead to patient morbidity and mortality. In certain situations, it would be appropriate to routinely solicit information regarding religious or personal prohibitions in order to identify potential conflicts with the treatment plan. Maintaining open communication with the patient can prevent such conflicts. In the authors experience, patients also may be misinformed about specific pharmaceuticals as a result of their own Internet searches; this can make them hesitant to trust the healthcare team, who can provide them with well-researched information. When confronted with these situations, the goal of healthcare professionals should be to provide their patients with treatment options and sound medical advice while simultaneously respecting patients autonomy when they make their decisions.

Consider the following example of an individual who abstains from pork products. Patients receiving dialysis must have their hemodialysis catheter flushed and locked with an anticoagulant solution to maintain the catheters patency. Normally, heparin locks are the anticoagulant of choice for this indication, but patients who strictly adhere to pork restrictions may refuse these. Heparin in central venous lines is more effective than normal saline at reducing the amount of line occlusions.10 Sodium citrate might be another option but it will present additional challenges because of procurement and availability.11 Additional options and their risks and benefits can be discussed with the patient to ensure a solution is found that meets everyones needs.

As technological advances enhance the ability of researchers and healthcare professionals to provide medications that target specific receptors, the challenge will be to provide consistent and relevant information to the patient. Providers may encounter the need to discuss a patients personal beliefs and opinions of medication-manufacturing processes while they are administering health care. A regularly updated, centralized resource that is easily accessible to providers at the point of treatment is necessary to help inform patients about their healthcare decisions. Although patients decisions and beliefs can be misunderstood by and prove frustrating for providers, this knowledge will help to prepare and facilitate a productive discussion regarding patients desired outcomes.

Disclosure: The authors declare no conflicts or financial interest in any product or service mentioned in the manuscript, including grants, equipment, medications, employment, gifts, and honoraria.

Read the original:

The Hidden Inactive Ingredient: Biological Products in Recombinant Pharmaceuticals - P&T Community

Fava Is All About Balance – East Bay Express

Some chefs dream of white tablecloth, three-Michelin-star glory. But Sylvie Osborne-Calierno and Jeremy Scheiblauer, a duo of chefs and best friends, had other plans in mind.

"We always knew we wanted a tiny little spot," Osborne-Calierno said. So when they heard that the 42-year-old Juice Bar Collective on Vine Street was closing, they jumped at the chance to carve out their own tiny slice of North Berkeley.

It's not that Osborne-Calierno and Scheiblauer lacked the pedigrees to open a larger restaurant. The friends met cooking at Chez Panisse 12 years ago, and both went on to open other restaurants Osborne-Calierno with Toto, a brunch restaurant in Barcelona, Spain, and Scheiblauer with Heyday, a lunch spot in San Francisco's Financial district. Both ultimately returned to Chez Panisse, where they were working until Fava's opening.

Alice Waters' guiding principles are evident in the food that comes out of Fava's kitchen: the insistence upon fresh, top-quality ingredients, simply prepared without relying on fancy equipment. But Fava drops the French cooking techniques, the four-course format, and the triple-digit price tag in favor of a shoebox-sized, counter-service only restaurant, equipped with compostable plates and a few outdoor folding tables and chairs.

With such a small space, the duo had to pare down Fava's menu to just the essentials: a few salads, sandwiches, and a daily soup, plus fresh juices and sesame-chocolate chip cookies.

"It really gives you constraints that sometimes maybe feel a little tight," Osborne-Calierno said. "But at the same time, it really forces you to really hone down and get to the real soul of what you're trying to do."

Fava's style isn't easy to distill into a few words or labels, but the menu, which happens to be entirely housemade and organic, is clearly Mediterranean and Middle Eastern influenced. Latin American influences also make their way onto the menu, albeit more subtly. "We don't really feel pegged to any one culture," Osborne-Calierno said. "We just make things that taste good."

The lamb flatbread is one of the restaurant's best sellers, and for good reason. The bread itself, which lies somewhere between naan and pita, is made in-house every day with whole wheat flour. The last bit of cooking gets done on a plancha, giving it the same effect as freshly made bread. The bread was delightfully airy, chewy, and elastic, with a touch of crispness around the edges and a nutty, toasty flavor. On top of the flatbread was a smear of harissa, which lent bright color and a punch of cumin. Layered on top of that were tender, juicy slices of lamb shoulder braised in a tomatoey broth "grandma-style," Osborne-Calierno calls it, as opposed to French cooking techniques that incorporate mirepoix and wine. Dollops of creamy, rich yogurt topped the lamb, followed by crunchy red cabbage tossed with a touch of vinegar. The crowning glory was a copious handful of mint, dill, and cilantro.

At first, I was taken aback by how many herbs were on top of the flatbread, and since you're eating it open-faced, you're likely to end up with an entire sprig of mint sticking out of your mouth at some point. But the whole thing was surprisingly well-balanced, and it struck me how the herbs and vegetables shone just as brightly as the lamb. The herbs also kept the sandwich light and refreshing, which was a minor miracle considering that lamb is such a fatty, richly flavored protein. "We don't believe in there being too [many] herbs," Osborne-Calierno laughed. "We like to treat herbs like they're a lettuce, like they're a vegetable."

Fava's menu is primarily vegetable-focused every other item on the menu when I visited was vegetarian. I particularly enjoyed the escarole salad. The slight bitterness of the crisp, lightly dressed leaves was perfectly balanced by slices of juicy melon, while housemade chile almonds added a little earthiness and spiciness. Bulgur wheat served double duty, not only by making the salad more filling, but also by brightening it up with plenty of lemon juice. The most striking part was the leaves of purple basil on top, which added an unexpected pop of color and flavor.

"I like the purple basil ... it kinda looks crazy," said Osborne-Calierno. "[We] kinda try to be a little crazy, spice up North Berkeley a little bit."

Also on offer was a hummus bowl with roasted eggplant, topped with cherry tomatoes, French feta, red cabbage, and purslane. The hummus was exceptionally smooth with plenty of tahini flavor, while the golden tomatoes were firm and perfectly ripe. I appreciated the use of French feta here, which provided a punch of salt with a creamier, milder flavor than its Greek and Bulgarian counterparts. Like all the other dishes I tried, it was also a beautiful dish to look at, with the green purslane and purple cabbage dominating the dish with little golden tomatoes shining like gems amid a background of hummus.

For maximum Instagrammability, try one of the brightly colored yet not-too-sweet housemade juices to enhance the look of your meal. I particularly loved the mango lassi, which was creamy and tangy with plenty of real mango flavor. The hibiscus lemonade and green juice, on the other hand, were refreshing and tart.

A rotating selection of soups and sides rounds out the menu. I wasn't a big fan of the side of roasted sweet potatoes. Though they were perfectly roasted and caramelized, they needed a little salt for balance.

The soups, though, are worth ordering. On one of my visits, it was a warm tomato soup with firm chickpeas and a dollop of zesty, creamy lime yogurt; on another, it was a red lentil soup topped with a drizzle of lemon oil for an unexpected citrusy kick.

Don't leave Fava without one (or more) of the sesame chocolate chip cookies. The sesame provided a nutty flavor and subtle richness to the cookie, while giving it a chewier-than-usual texture. Best of all, the cookies used dark chocolate wafers rather than chocolate chips, which provided luscious pools of fruity, bitter dark chocolate.

No, it's not health food. But like everything else at Fava, it's all about balance. Asked whether Fava will open for dinner one day, the duo said they'd like to stick to lunch hours and possibly breakfast one day so they can balance cooking with having an actual life outside of the kitchen, which is a rarity in the restaurant industry.

Osborne-Calierno is the mother of 5-year-old twins. When Scheiblauer isn't cooking in the kitchen, he's busy playing synths in his post-punk, new wave band called Vandal Moon. Sure, the duo is ushering a youthful, casual vibe with plenty of fresh flavors into North Berkeley but unlike generations before them, they won't sacrifice having a balanced lifestyle.

"I don't think moms should have to choose between 'I'm either a mom or I'm a chef,'" Osborne-Calierno said. "We are all these things at once, and that's really important."

Go here to see the original:

Fava Is All About Balance - East Bay Express

Amazon fish wears nature’s ‘bullet-proof vest’ to thwart piranhas – Reuters

WASHINGTON (Reuters) - One of the worlds largest freshwater fish is protected by the natural equivalent of a bullet-proof vest, helping it thrive in the dangerous waters of the Amazon River basin with flexible armor-like scales able to withstand ferocious piranha attacks.

FILE PHOTO: Diver Alex Reeson checks on the Arapaima or Pirarucu fishes in the tropical section of Europe's biggest freshwater aquarium Aquatis, due to open October 21, in Lausanne, Switzerland October 17, 2017. REUTERS/Denis Balibouse/File Photo

Researchers from the University of California, San Diego and University of California, Berkeley on Wednesday described the unique structure and impressive properties of the dermal armor of the fish, called Arapaima gigas. They said their findings can help guide development of better body armor for people as well as applications in aerospace design.

The fish, also known as pirarucu, gets up to 10 feet (3 meters) long and weighs up to 440 pounds (200 kg). Arapaima, a fish that can breathe air and survive up to a day outside of the water, inhabits rivers in Brazil, Guyana and Peru infested with piranhas, known for razor-sharp teeth, incredible bite strength and deadly feeding frenzies.

There long has been an evolutionary arms race in which predators develop ways to kill - like sharp teeth - and other animals develop ways to stay alive like the dermal armor seen in various fish, dinosaurs and mammals over time. Arapaimas scales, the researchers said, possess all the best attributes of a bullet-proof vest, but the elements are better integrated into one solid piece combining imperviousness and flexibility.

Such lightweight and tough materials like fish scales are the sexy topics that materials scientists are pursuing, said Wen Yang, a UCSD materials scientist who helped lead the study published in the journal Matter.

It is true that the natural armor is similar to artificial body armor because of the similar scale overlapping system. However, the natural armor such as these fish scales is tough and much lighter, without impeding body flexibility and locomotion, Yang added. Remember that the fish scales were developed through hundreds of millions of years. They are very advanced.

The researchers conducted laboratory tests of the scales.

The scales, they found, have a hardmineralized outer layer to resist penetration that is bound to a tough-but-flexible inner layer by collagen themain structural protein in skin and other connective tissues in the body.

This structure means the scales can become deformed when bitten by a piranha but are not torn, broken or pierced, protecting the fish from injury.

We were able to see how the collagen fibers deform without a catastrophic failure including the mechanisms of twisting, folding, sliding, stretching, delamination, Yang said.

The study was backed by the U.S. Air Force Office of Scientific Research.

Reporting by Will Dunham; Editing by Sandra Maler

Read more:

Amazon fish wears nature's 'bullet-proof vest' to thwart piranhas - Reuters

9 must-have Instant Pot accessories for healthy eating – CNET

Making bread in an Instant Pot is easy.

The Instant Pot is one of our must-have kitchen devices and with the right Instant Pot accessories the miracle machine can be a great tool for health and wellness, too. As it's a combination pressure cooker, slow cooker, steamer, rice cooker, warmer and more, you can make thousands of satiating mealsin your programmableInstant Pot (plus using fewer dirty dishes). From flavorful meat stews and fall-off-the-bone short ribs to summer shrimp boils and nutrient-packed soups, most healthy Instant Pot recipes are done in minutes and require minimal prep.

Read more on CNET:What's the best Instant Pot to buy? Instant Pot Duo vs. Lux vs. Ultra

One reason an Instant Pot is perfect for preparing dishes forhealthy eatingis how fast it cooks tender meats, perfect beans, creamy Greek yogurt, eggs and other proteins, integral to a balanced diet. With the help of a few smart, healthy Instant Pot accessories like cups for cooking eggs, steamed vegetable baskets and a few cookbooks with healthy Instant Pot recipes in mind, you can stay committed to your Whole30 or keto planor maintain a more balanced diet with loads of protein and nutrient-packed vegetables.

Now playing: Watch this: 8 essential Instant Pot tips

2:11

Below are nine of the best Instant Pot accessories to have around for eating healthy all week (and year) long.

Disclosure:These products and services are independently chosen by our editors. CNET may get a share of the revenue if you buy anything featured on our site.

This Instant Pot steamer basket will make cooking veggies or boiling eggs even easier, and any dietician will tell you steaming adds the least amount of fat to foods while best maintaining their nutritional integrity. This steamer basket Instant Pot accessory is available in three different sizes (3-quart, 6-quart and 8-quart) so there's a size that will fit perfectly for any Instant Pot model. The stainless steel steamer basket also comes with a silicone handle, to keep it from getting too hot and burning your hands!

Serving a similar function as the basket above, this BPA-free silicone version has the added bonus of folding up flat for easy storage so is a great choice for smaller spaces. It also cleans up more easily than metal since it's naturally nonstick. It can be used with 6-quart and 8-quart Instant Pots.

The Instant Pot does come with a metal rack, but this BPA-free silicone version is far easier to clean since it's nonstick. It elevates your food from the liquid in the pot, while the vented bottom promotes better airflow for even cooking and also allows any rendered fat to drain. Use with 6-quartand 8-quart Instant Pots. When not in use, it works as a heat-safe trivet too.

This healthy Instant Pot cookbook contains easy recipes for dishes you'll love, all of which were developed specifically for a low-carb, high-protein keto diet. An Instant Pot is one of the best tools for a high-protein, low-carb lifestyle, cooking savory dishes like fall-off-the-bone red meats, quick chicken and fish and fresh vegetables in a fraction of the time it takes in an oven.

Read more:15 healthy Instant Pot recipes for breakfast, lunch and dinner

Make perfect soft- or hard-boiled eggs in your multicooker with this handy Instant Pot egg steamer rack. It holds nine eggs, making this steam tool ideal for healthy meal prep. Chop up some hard-boiled eggs for your salad or add a soft-boiled egg on top of avocado toast for an easy, healthy weekday breakfast.

If you love Starbucks'sous vide egg bites, you need these silicone Instant Pot egg molds that let you make your own at home. Packed with protein, they're a meal prepper's dream. Customize them with any additions you want, make a double batch on Sunday and grab them for healthy breakfasts and lunch all week long. This silicone mold set for two comes with lids, spoons and a steamer rack, but if you only want one, you can buy asingle egg bite mold with lid by itself.

Read more:Why you should own an Instant Pot: 5 reasons, plus recipes and tips

Whip up some light, healthy meals in your Instant Pot with this great healthy multicooker cookbook, which includes 75 simple recipes all suited for the miracle machine. Each healthy Instant Pot recipe includes a gluten-free option and they are all free of refined sugars. The sweet and savory dishes range from an Instant Pot eggplant parmesan bake to flourless banana oat bread to one-pot chocolate cake and frosting -- yum.

Read more:10 foods you should not cook in your Instant Pot

As if the Instant Pot wasn't convenient enough, these stackable stainless steel Instant Pot inserts are for cooking multiple things at once, like a protein and sides. Each stainless steel inner pot can also serve as layer pans for cooking lasagna or cheesecake on cheat day, but we recommend them as one of the best Instant Pot accessories for anyone looking to save time and make more than one dish at a time in an Instant Pot. Try multiple proteins to keep meal prep interesting, easy and fast!

Read more:13 Instant Pot recipes we keep coming back to

Read more:The hunt for the best air fryer

You can make your own yogurt easily at home with this nifty Instant Pot yogurt maker accessory. We love that it's easy to use and easy to clean. This is a perfect tool for anyone making yogurt or smoothies to power up in the morning. Try creating different flavors and varieties such as Greek yogurt. Incorporate unique add-ins to shake up your healthy breakfast routine.

Read the original:

9 must-have Instant Pot accessories for healthy eating - CNET

Researchers Find Fish Wearing Natural ‘Bullet-Proof Vest’ to Thwart Piranhas in Amazon – News18

One of the worlds largest freshwater fish is protected by the natural equivalent of a bullet-proof vest, helping it thrive in the dangerous waters of the Amazon River basin with flexible armor-like scales able to withstand ferocious piranha attacks.

Researchers from the University of California, San Diego and University of California, Berkeley on Wednesday described the unique structure and impressive properties of the dermal armor of the fish, called Arapaima gigas. They said their findings can help guide development of better body armor for people as well as applications in aerospace design.

The fish, also known as pirarucu, gets up to 10 feet (3 meters) long and weighs up to 440 pounds (200 kg). Arapaima, a fish that can breathe air and survive up to a day outside of the water, inhabits rivers in Brazil, Guyana and Peru infested with piranhas, known for razor-sharp teeth, incredible bite strength and deadly feeding frenzies.

There long has been an evolutionary arms race in which predators develop ways to kill - like sharp teeth - and other animals develop ways to stay alive like the dermal armor seen in various fish, dinosaurs and mammals over time. Arapaimas scales, the researchers said, possess all the best attributes of a bullet-proof vest, but the elements are better integrated into one solid piece combining imperviousness and flexibility.

Such lightweight and tough materials like fish scales are the sexy topics that materials scientists are pursuing, said Wen Yang, a UCSD materials scientist who helped lead the study published in the journal Matter.

It is true that the natural armor is similar to artificial body armor because of the similar scale overlapping system. However, the natural armor such as these fish scales is tough and much lighter, without impeding body flexibility and locomotion, Yang added. Remember that the fish scales were developed through hundreds of millions of years. They are very advanced.

The researchers conducted laboratory tests of the scales.

The scales, they found, have a hard mineralized outer layer to resist penetration that is bound to a tough-but-flexible inner layer by collagen the main structural protein in skin and other connective tissues in the body.

This structure means the scales can become deformed when bitten by a piranha but are not torn, broken or pierced, protecting the fish from injury.

We were able to see how the collagen fibers deform without a catastrophic failure including the mechanisms of twisting, folding, sliding, stretching, delamination, Yang said.

The study was backed by the U.S. Air Force Office of Scientific Research.

Get the best of News18 delivered to your inbox - subscribe to News18 Daybreak. Follow News18.com on Twitter, Instagram, Facebook, Telegram, TikTok and on YouTube, and stay in the know with what's happening in the world around you in real time.

Continued here:

Researchers Find Fish Wearing Natural 'Bullet-Proof Vest' to Thwart Piranhas in Amazon - News18

Structural Biochemistry/Proteins/Protein Folding …

Protein folding is a process in which a polypeptide folds into a specific, stable, functional, three-dimensional structure. It is the process by which a protein structure assumes its functional shape or conformation

Proteins are formed from long chains of amino acids; they exist in an array of different structures which often dictate their functions. Proteins follow energetically favorable pathways to form stable, orderly, structures; this is known as the proteins native structure. Most proteins can only perform their various functions when they are folded. The proteins folding pathway, or mechanism, is the typical sequence of structural changes the protein undergoes in order to reach its native structure. Protein folding takes place in a highly crowded, complex, molecular environment within the cell, and often requires the assistance of molecular chaperones, in order to avoid aggregation or misfolding. Proteins are comprised of amino acids with various types of side chains, which may be hydrophobic, hydrophilic, or electrically charged. The characteristics of these side chains affect what shape the protein will form because they will interact differently intramolecularly and with the surrounding environment, favoring certain conformations and structures over others. Scientists believe that the instructions for folding a protein are encoded in the sequence. Researchers and scientists can easily determine the sequence of a protein, but have not cracked the code that governs folding (Structures of Life 8).

Early scientists who studied proteomics and its structure speculated that proteins had templates that resulted in their native conformations. This theory resulted in a search for how proteins fold to attain their complex structure. It is now well known that under physiological conditions, proteins normally spontaneously fold into their native conformations. As a result, a protein's primary structure is valuable since it determines the three-dimensional structure of a protein. Normally, most biological structures do not have the need for external templates to help with their formation and are thus called self-assembling.

Protein renaturation known since the 1930s. However, it was not until 1957 when Christian Anfinsen performed an experiment on bovine pancreatic RNase A that protein renaturation was quantified. RNase A is a single chain protein consisting of 124 residues. In 8M urea solution of 2-mercaptoethanol, the RNase A is completely unfolded and has its four disulfide bonds cleaved through reduction. Through dialysis of urea and introducing the solution to O2 at pH 8, the enzymatically active protein is physically incapable of being recognized from RNase A. As a result, this experiment demonstrated that the protein spontaneously renatured.

One criteria for the renaturation of RNase A is for its four disulfide bonds to reform. The likelihood of one of the eight Cys residues from RNase A reforming a disulfide bond with its native residue compared to the other seven Cys residues is 1/7. Furthermore, the next one of remaining six Cys residues randomly forming the next disulfide bond is 1/5 and etc. As a result, the probability of RNase A reforming four native disulfide links at random is (1/7 * 1/5 * 1/3 * 1/1 = 1/105). The result of this probability demonstrates that forming the disulfide bonds from RNase A is not a random activity.

When RNase A is reoxidized utilizing 8M urea, allowing the disulfide bonds to reform when the polypeptide chain is a random coil, then RNase A will only be around 1 percent enzymatically active after urea is removed. However, by using 2-mercaptoethanol, the protein can be made fully active once again when disulfide bond interchange reactions occur and the protein is back to its native state. The native state of the RNase A is thermodynamically stable under physiological conditions, especially since a more stable protein that is more stable than that of the native state requires a larger activation barrier, and is kinetically inaccessible.

By using the enzyme protein disulfide isomerase (PDI), the time it takes for randomized RNase A is minimized to about 2 minutes. This enzyme helps facilitate the disulfide interchange reactions. In order for PDI to be active, its two active site Cys residues needs to be in the -SH form. Furthermore, PDI helps with random cleavage and the reformation of the disulfide bonds of the protein as it attain thermodynamically favorable conformations.

Proteins in a "scrambled" state go through PDI to renature, and their native state does not utilize PDI because native proteins are in their stable conformations. However, proteins that are posttranslationally modified need the disulfide bonds to stabilize their rather unstable native form. One example of this is insulin, a polypeptide hormone. This 51 residue polypeptide has two disulfide bonds that is inactivated by PDI. The following link is an image showing insulin with its two disulfide bonds. Through observation of this phenomenon, scientists were able to find that insulin is made from proinsulin, an 84-residue single chain. This link provides more information on the structure of proinsulin and its progression on becoming insulin. The disulfide bonds of proinsulin need to be intact before conversion of becoming insulin through proteolytic excision of its C chain which is an internal 33-residue segment. However, according to two findings, the C chain is not what dictates the folding of the A and B chains, but instead holds them together to allow formation of the disulfide bonds. For one, with the right renaturing conditions in place, scrambled insulin can become its native form with a 30% yield. This yield can be increased if the A and B chains are cross-linked. Secondly, through analysis of sequences of proinsulin from many species, mutations are permitted at the C chain eight times more than if it were for A and B chains.

There are various interactions that help stabilize structures of native proteins. Specifically, it is important to examine how the interactions that form protein structures are organized. In addition, there are only a small amount of possible polypeptide sequences that allow for a stable conformation. Therefore, it is evident that specific sequences are used through evolution in biological systems.

On average, about sixty percent of proteins contain a high amount of alpha helices, and beta pleated sheets. Through hydrophobic interactions, the protein is able to achieve compact nonpolar cores, but they lack the ability to specify which polypeptides to restrict in particular conformations. As seen in polypeptide segments in the coil form, the amount of hydrogen boding is not lesser than that of alpha helices and beta pleated sheets. This observation demonstrates that the different kinds of conformations of polypeptides are not limited by hydrogen bonding requirements. Ken Dill has suggested that helices and sheets occur as a result of the steric hindrance in condensed polymers. Through experimentation and simulation of conformations with simple flexible chains, it can be determined that the proportion of beta pleated sheets and alpha helices increase as the level of complication of chains is increased. Therefore, it can be concluded that helices and sheets are important in the complex structure of a protein, as they are compact in protein folding. The coupling of different forces such as hydrogen bonding, ion pairing, and van der Waals interactions further aids in the formation of alpha helices and beta sheets.

By investigating protein modification, the role of different classes of amino acid residues in protein folding can be determined. For example, in a particular study the free primary amino groups of RNase A were derivatized with poly-DL-alanine which consist of 8 residue chains. The poly-Ala chains are large in size and are water-soluble, thus allowing the RNase's 11 free amino groups to be joined without interference of the native structure of the protein or its ability to refold. As a result, it can be concluded that the protein's internal residues facilitates its native conformation because the RNase A free amino groups are localized on the exterior. Furthermore, studies have shown that mutations that occur on the surface of residues are common, and less likely to change the protein conformation compared to changes of internal residues that occur. This finding suggests that protein folding is mainly due to the hydrophobic forces.

George Rose demonstrated that protein domains consisted of subdomains, and furthermore have sub-subdomains, and etc. As a result, it is evident that large proteins have domains that are continuous, compact, and physically separable. When a polypeptide segment within a native protein is visualized as a string with many tangles, a plane can be seen when the string is cut into two segments. This process can be repeated when n/2 residues of an n-residue domain is highlighted with a blue and red color. As this process is repeated it can be seen that at all stages, the red and blue areas of the protein do not interpenetrate with one another. The following link shows an X-ray structure of HiPIP (high potential iron protein) and its first n/2 residues on the n-residue protein colored red and blue. Furthermore, the subsequent structures shown in the second and third row show this process of n/2 residue splitting reiterated as shown where the left side of the protein has its first and last halves with red and blue while the rest of the chain colored in gray. Through this example, it is clearly seen that protein structures are organized in a hierarchical way, meaning that the polypeptide chains are seen as sub-domains that are themselves compact structures and interact with adjacent structures. These interactions forms a larger well organized structure largely due to hydrogen bonding interactions and has an important role in understanding how polypeptides fold to form their native structure.

Since the side chains inside globular proteins fit together with much complementary its packing density can be almost like that of organic crystals. As a result, in order to confirm whether or not this phenomenon of high packing density was an important factor in contributing to protein structure, Eaton Lattman along with George Rose attempted to verify if there was an interaction between side chains that was preferred in a globular protein. They analyzed a total of 67 well studied structures of globular proteins, and concluded that there were no preferred interactions. This experiment demonstrated that packing is not what directs the native fold, but instead the native fold is necessary for packing of a globular protein. This notion can be further supported as members of a protein family result in the same fold despite their lack of sequence similarity and distant relationships.

In addition, structural experimental data have shown that there are a variety of ways that a protein's internal residues can become compact together in an efficient manner. In an extensive study done by Brian Matthews based on T4 lysozyme, which is produced by bacteriophage T4, it was found that changes in the residues of the T4 lysozyme only affected local shifts and did not result in any global structure change. The following link gives an X-ray view of T4 lysozyme and a brief biochemical description of the structure. Matthews took over 300 different mutants of the 164 residue T4 lysozyme, and compared them with one another. Also, it was observed that the T4 lysozyme could withstand insertions of about 4 residues while still not having any major structural changes to the overall protein structure nor enzyme activity. Furthermore, by using assay techniques it was demonstrated that only 173 of the mutants in T4 of the 2015 single residue substitutions done had significant amounts of enzymatic activity diminished. Through these experiments, it is evident that protein structures are extremely withstanding.

Levinthal's paradox is a thought experiment, also constituting a self-reference in the theory of protein folding. In 1969, Cyrus Levinthal noted that, because of the very large number of degrees of freedom in an unfolded polypeptide chain, the molecule has an astronomical number of possible conformations. An estimate of 3300 or 10143 was made in one of his papers.

The Levinthal paradox observes that if a protein were folded by sequentially sampling of all possible conformations, it would take an large amount of time to do so, even if the conformations were sampled at a rapid rate . Based upon the observation that proteins fold much faster than this, Levinthal then proposed that a random conformational search does not occur, and the protein must, therefore, fold through a series of meta-stable intermediate states.

In 1969 Cyrus Levinthal calculated that if a protein were to randomly sample every possible conformation as it folded from the unfolded state to the native state it would take an astronomical amount of time, even if the protein reached 100 billion conformations in one second. Observing that proteins fold in a relatively short amount of time, Levinthal proposed that proteins fold in a fixed and directed process. We now know that while protein folding is not a random process there does not seem to be a single fixed protein folding pathway.This observation came to be known as the Levinthal paradox. This paradox clearly reveals that proteins do not fold by trying every possible conformation. Instead, they must follow at least a partly defined folding pathway made up of intermediates between the fully denatured proteins and its native structure.

The way out of the Levinthal Paradox is to recognize cumulative selection. According to Richard Dawkins, he asked how long it would take a monkey poking randomly at a typewriter to reproduce "Methinks it is like a weasel", Hamlet's remark to Polonius. A large number of keystrokes, of the order of 1040 would be required. Yet if we suppose that each correct character was preserved, allowing the monkey to retype only the wrong ones, only a few thousand keystrokes, on average, would be needed. The crucial difference between these scenarios is that the first utilizes a completely random search whereas in the second case, partly correct intermediates are retained. This also reveals that the essence of protein folding is the tendency to retain partly correct intermediates, although the protein-folding problem is much more difficult than the one presented to Shakespeare example above.

In order to correctly understand the protein-folding problem, we must consider certain characteristics of protein. Since proteins are only marginally stable, the free-energy difference between the folded and the unfolded states of a typical 1000-residue protein is 42 kJ mol1 and thus each residue contributes on average only 0.42 kJ mol1 of energy to maintain the folded state. This amount is less than the amount of thermal energy, which is 2.5 kJ mol1 at room temperature. This meagre stabilization energy means that correct intermediates, especially those formed early in folding, can be lost. The interactions that lead to cooperative folding, nonetheless, can stabilize intermediates as structure builds up. Thus, local regions that have significant structural preference, though not necessarily stable on their own, will tend to adopt their favored structures and, as they form, can interact with one other, resulting in increased stabilization. Nucleation-condensation model refers to this conceptual framework in solving the protein-folding challenge.

Proteins folding forms energetically favorable structures stabilized by hydrophobic interactions clumping, hydrogen bonding and Van der Waals forces between amino acids. Protein folding first forms secondary structures, such as alpha helices, beta sheets, and loops. Different amino acids have different tendencies for whether they are going to form Alpha Helices, Beta sheets, or Beta Turns based upon polarity of the amino acid and rotational barriers. For example, the amino acids, valine, threonine, isoleucine, tend to destabilize the alpha helices due to steric hindrance. Thus, they prefer conformational shifts towards Beta sheets rather than alpha helices. The relative frequencies of the amino acids in secondary structures are grouped according to their preferences for alpha helices, beta sheets or turns (Table 1). Table 1: Relative frequencies of amino acid residues in secondary structuresThese structures in turn, fold to form tertiary structures, stabilized by the formation of intramolecular hydrogen bonds. Covalent bonding may also occur during the folding to a tertiary structure, through the formation of disulfide bridges or metal clusters. According to Robert Pains Mechanisms of Protein Folding, molecules also often pass through an intermediate molten globule state formed from a hydrophobic collapse (in which all hydrophobic side-chains suddenly slide inside the protein or clump together) before reaching their native confirmation. However, this means all the main chain NH and CO groups are buried in a non-polar environment, but they prefer an aqueous one, so secondary structures must fit together very well, so that the stabilization through hydrogen bonding and Van der Waals forces interactions overrides their hydrophilic tendencies. The strengths of hydrogen bonds in a protein vary depending on their position in the structure; H-bonds formed in the hydrophobic core contribute more to the stability of the native state than H-bonds exposed to the aqueous environment.

Water-soluble proteins fold into compact structures with non-polar, hydrophobic cores. The inside of protein contains non-polar residues in center (i.e. - leucine, valine, methionine and phenylalanine), while the outside contains primarily polar, charged residues (i.e. - aspatate, glutamate, lysine and arginine). This way the polar, charged molecules can interact with the surrounding water molecules while the hydrophobic molecules are protected from the aqueous surroundings. Minimizing the number of hydrophobic side chains on the outer part of the structure makes the protein structure thermodynamically more favorable because the hydrophobic molecules prefer to be clumped together, when surrounded by an aqueous environment (i.e. hydrophobic effect). Proteins that span biological membranes (i.e. - porin) have an inside out distribution, with respect to the water-soluble native structure, they have hydrophobic residue covered outer surfaces, with water filled centers lined with charged and polar amino acids.

In Folding Scene Investigation: Membrane Proteins, a paper written by Paula J Booth and Paul Curnow, the authors attempt to answer how the folding mechanisms of integral membrane proteins with helical structures work.Studying the folding of membrane proteins has always been difficult as these proteins are generally large and made of more than one subunit. The proteins posses a high degree of conformational flexibilitywhich is necessary for them to perform their function in the cell. Also, these proteins have both hydrophobic surfaces, facing the membrane, and hydrophilic surfaces, facing the aqueous regions on either side of the membrane. The proteins are move laterally and share the elastic properties of the lipid bilayer in which they are embedded. In order to study these proteins, Booth and Curnow believe that one must manipulate the lipid bilayer and combine kinetic and thermodynamic methods of investigation.

Reversible Folding and Linear Free EnergyThe free energy of protein folding is measured by reversible chemical denaturation. The reversible folding of a protein depends on this free energy. For the helix proteins that were being studied, it was proven that a reversible, two-state process is followed. bR (a helical membrane protein called bacteriorhodopsin) reversibly unfolds if SDS (a denaturant which is an anionic detergent) is added to mixed lipid, detergent micells. The two-state reaction involves a partly unfolded SDS state and a folded bR state. By comparing the logs of the unfolding and folding rate, and the SDS mole fraction, a linear plot was generated proving a linear relationship. This plot proved that bR had a very high stability outside of its membraneproving that it was unexpectedly stable. Furthermore, bR was so stable outside of the membrane that it would not unfold during a reasonable period of time without addition of denaturant.

Comparison with Water-Soluble ProteinsBooth and Curnow studied the 3 membrane proteins about which the most information is held: bR, DGK (Escherichia coli diacylglycerol kinase) and KcsA (Sterptococcus lividans potassium channel). These three membrane proteins were compared to water-soluble proteins (which fold by 2 or 3 state kinetics). The overall free energy change of unfolding in the absence of denaturant was the same for water-soluble proteins and membrane proteins of similar size. This proves that it is the balance of weak forces rather than the types of forces that stabilize the protein that determines its stability. It was proven that H-bonds in the membrane proteins were of similar strength to those of the water-soluble proteins, rather than being stronger in membrane proteins as was expected.

Mechanical Strength and Unfolding Under Applied ForceDynamic force microscopy can be used to measure the mechanical response of a particular region of a protein under applied force. The unfolding force in this case depends on the activation barrier. This unfolding has nothing to do with the thermodynamic stability of a protein. For unfolding under applied force, the membrane proteins (especially bR) seem to follow the rules of Hammond behavior. The energy difference between two consecutive states of this reaction is reduced and the states become similar in structure.

Influence of Surrounding MembraneMembrane proteins are influenced greatly by the membranes they are surrounded by. If the lipids incorporate in detergent micells-increasing the stability of the lipid structureboth the protein and its folding are stabilized. Different combinations of different lipids can result in different stabilities or folding of membrane proteins. The size of the membrane can also affect the membrane protein.Different types of lipids cause different membrane properties. A type of lipids called PE lipids have higher spontaneous curvatures than a second type of lipid called a PC lipid. By adding PE lipids to PC lipids the monolayer curvature of the bilayer increases. Increasing the curvature of the lipid bilayer increases the stability of the protein folding.

In mitochondria, the proteins that are made from the ribosomes are directly take in from the cytosol. Mitochondrial proteins are first completely synthesized in the cytosol as mitochondrial precursor proteins, then taken up into the membrane. The Mitochondrial proteins contain specific signal sequence at their N terminus. These signal sequences are often removed after entering the membrane but proteins entering membranes that has outer, inner, inter membrane have internal sequences that play a major movement in the translocation within the inner membrane.

Protein translocation plays a major role in translocating proteins across the mitochondrial membranes. Four major multi-subunit protein complexes are found in the outer and the inner membrane. TOM complexes are found in the outer membrane, and two types of TIM complexes are found integrated within the inner membrane: TIM23 and TIM22. The complexes act as receptors for the mitochondrial precursor proteins.

TOM: imports all nucleus encoded proteins. It primarily starts the transport of the signal sequence into the inter membrane space and inserts the transmembrane proteins into outer membrane space. A Beta barrel complex called the SAM complex is then in charge of properly folding the protein in the outer membrane. TIM23 found in the inner membrane moderates the insertion of soluble proteins into the matrix, and facilitates the insertion of transmembrane proteins into the inner membrane. TIM23, another inner membrane complex facilitates the insertion inner membrane proteins comprised of transporters that move ADP, ATP, and phosphate across the mitochondrial membranes. OXA, yet another inner membrane complex, helps insert inner membrane proteins that were synthesized from the mitochondria itself and the insertion of inner membrane proteins that were first transported into the matrix space.File:Translocation.jpg

The place where the protein chain begins to fold is a topic that is greatly studied. As the nascent chain goes through the exit tunnel of the ribosome and into the cellular environment, when does the chain begin to fold? The idea of cotranslational folding in the ribosomal tunnel will be discussed. The nascent chain of the protein is bound to the peptidyl transferase centre (PTC) at its C terminus and will emerge in a vectorial manner. The tunnel is very narrow and enforces a certain rigidity on the nascent chain, with the addition of each amino acid the conformational space of the protein increases. Co translational folding can be a big help in reducing the possible conformational space by helping the protein to acquire a significant level of native state while still in the ribosomal tunnel. The length of the protein can also give a good estimate of its three dimensional structure. Smaller chains tend to favor beta sheets while longer chains (like those reaching 119 out of 153 residues) tend to favor the alpha helix.

The ribosomal tunnel is more than 80 in length and its width is around 10-20 . Inside the tunnel are auxiliary molecules like the L23, L22, and L4 proteins that interact with the nascent chain help with the folding. The tunnel also has hydrophilic character and helps the nascent chain to travel through it without being hindered. Although rigid, the tunnel is not passive conduit but whether or not it has the ability to promote protein folding is unknown. A recent experiment involving cryoEM has shown that there are folding zones in the tunnel. At the exit port (some 80 from the PTC), the nascent chain has assumed a preferred low order conformation. This enforces the suggestion that the chain can have degrees of folding at certain regions. Although some low order folding can occur, the adoption of the native state occurs outside the tunnel, but not necessarily when the nascent chain has been released. The bound nascent chain (RNC) adopts partially folded structure and in a crowded cellular environment, this can cause the chain to self-associate. This self-association, however, is relieved with the staggered ribosomes lined along the exit tunnel that maximizes the distances between the RNC.

Generation of RNC for studies:

One technique of generating RNC and taking snapshots as it emerges from the tunnel is to arrest translation. A truncated DNA without a termination sequence is used. This allows for the nascent chain to remain bound until desired. To determining the residues of the chain, they can be labeled by carbon-13 or nitrogen-15 and later detected by NMR spectroscopy. Another technique is the PURE method and it contains the minimal components required for translation. This method has been used to study the interaction of the chains and auxiliary molecules like the TF chaperone. This method is coupled with quartz-crystal microbalance technique to analyze the synthesis by mass. An in vivo technique in generating RNC chain can be done by stimulating it in a high cell density. This is initially done in an unlabeled environment, the cells are then transferred to a labeled medium. The RNC is generated by SecM. The RNC is purified by affinity chromatography and detected by SDS-PAGE or immunoblotting.

By generating the RNCs, many experiments can be done to study more about the emerging nascent chain. As mentioned above, the chain emerges from the exit tunnel in a vectorial manner. This enables the chain to sample the native folding and increases the probability of folding to the native state. Along with this vectorial folding, chaperones also help in favorable folding rates and correct folding.

Protein Entering the Mammalian ER:The endoplasmic reticulum (ER) is a main checkpoint for protein maturation to ensure that only correctly folded proteins are secreted and delivered to the site of action. The protein entrance to the ER begins with recognition of a N terminus signal sequence. Specially, this sequence is detected by a signal recognition protein (SRP) causing the ribosome/nascent chain/SRP complex bind to the ER membrane. Then, the complex travels through a proteinaceous pore called Sec61 translocon which allows the polypeptide chain enter the lumen portion of the ER.

Processes in Conflict During Protein Folding:After the protein enters the ER, the proteins break up into an ensemble of folding intermediates. These intermediates take three different routes. They are either folded properly and sent to be exported out of the endoplasmic reticulum (ER) into the cytosol, aggregated or picked out for degradation. These three processes are in competition to properly secrete a protein. In order for a protein to be properly secreted, the competition between folding, aggregation and degradation must be in favor of folding, so that folding occurs faster than the other processes. This balance is termed proteostasis. The balance of proteostasis can be tipped in favor of folding by either using smaller molecules to stabilize the protein (called co-factors) or increasing the concentrations of folding factors. This ability to control proteostasis allows scientists the power to overcome some of the protein folding diseases such as cystic fibrosis.

The proteins that are folded properly are ready for anterograde transport, and secreted through the membrane of the ER into the cytosol by a cargo receptor that recognizes the properly folded protein. The proteins that are incorrectly folded are not secreted and are either targeted for degradation or aggregated. The aggregated proteins are able to re-enter the stage of protein ensembles ready to be folded so that they may try again at being folded properly.

Folding Factors in the Endoplasmic Reticulum:

Biochemical research on folding pathways has provided a comprehensive list of folding factors, or chaperones, involved with protein folding in the ER. Folding factors are categorized based on whether they catalyze certain steps or if they interact with intermediates in the folding pathway. General protein folding factors are typically separated into four different groups: heat shock proteins as chaperones or cochaperones, peptidyl prolyl cis/trans isomerases (PPIases), oxidoreductases, and glycan-binding proteins.

Many folding factors are great in that they are multi-functional. One folding factor can take care of different areas of the folding pathway. Unfortunately, this leads to redundancy due to different classes of proteins carrying out overlapping functions. This functional redundancy complicates the understanding of the specific roles of individual folding factors in aiding maturation of client proteins. Folding factors also prefer to act in concert during the maturation process, which further obscures the individual roles of each factor. Since these roles are not clear, it is difficult to confirm that even if one folding factor deals with a particular reaction in one protein, that same folding factor will carry out the same function in another.

In addition to aiding non-covalent folding and unfolding of proteins, folding factors in the ER sometimes delay interactions with the protein. This allows time for nascent proteins to fold properly and enables folded proteins to backtrack on its folding pathway, which prolongs equilibrium in a less folded state, preventing the protein from being held in a non-native state.

Folding after Endoplasmic Reticulum: Although ER provides only correctly assembled proteins to be secreted, some examples exist in which proteins change conformation in the Golgi bodies and beyond. Typically, newly folded proteins are sensitive and prone to unfolding while in the ER but resistant to unfolding after exit. In an environment without chaperones and other folding enzymes, proteins are compact and relatively resistant to change after exiting the ER. However, this doesnt necessarily mean that protein folding ends because some molecular chaperones like Hsp 70s and Hsp 90s continue to assist in protein conformation throughout the proteins existence.

A strategy for studying the folding of proteins is to unfold the protein molecules in high concentrations of a chemical denaturant like guanidinium chloride. The solution is then diluted rapidly until the denaturant concentration is lowered to a level where the native state is thermodynamically stable again. Afterwards, the structural changes of the protein folds may be observed. In theory, this sounds simple. However, such experiments are complex, since unfolded proteins have random coil states in chemical denaturants. Moreover, analyzing the structural changes taking place in a sample may is difficult, since all of the molecules may have significantly different conformations until the final stages of a reaction. As such, the analysis would have to be performed in a matter of seconds rather than days or weeks that are normally allowed to deduce the structure of a single conformation of a native protein. To avoid this problem, the disulphide bonds can be reduced after the protein is unfolded and reformed under oxidative conditions. The protein can then be identified by standard techniques such as mass spectroscopy to draw conclusions about the structure present at stages of folding where disulfide bonds are formed.

Multiple techniques are used to monitor structural changes during the refolding. For instance, in circular dichorism, UV is used from far away to provide a measurement of the appearance of the secondary structure during folding. UV at a close distance monitors the formation of the close-packed environment for aromatic residues. NMR is also a useful technique for characterizing conformations at the level of individual amino-acid residues. It can also be used to monitor how the development of structures protect amide hydrogens from solvent exchanges.

Circular Dichroism: This type of spectroscopy measures the absorption of circularly polarized light since the structures of protein such as the alpha helix and beta sheets are chiral and can absorb this sort of light. The absorption of light indicates the degree of the proteins foldedness. This technique also measures equilibrium unfolding of protein by measuring change of absorption against denaturant concentration or temperature. The denaturant melt measures the free energy of unfolding while the temperature melt measures the melting point of proteins. This technique is the most general and basic strategy for studying protein folding.

Dual Polarization Interferometry: This technique uses an evanescent wave of a laser beam confined to a waveguide to probe protein layers that have been absorbed to the surface of the waveguide. Laser light is focused on two waveguides, one that senses the beam and has an exposed surface, and one that is used to create a reference beam and to excite the polarization modes of the waveguides. The measurement of the interferogram can help calculate the protein density or fold, the size of the absorbed layer, and to infer structural information about molecular interactions at the subatomic resolution. A two-dimensional pattern is obtained in the far field when the light that has passed through the two waveguides is combined.

Mass Spectrometry: The advantages of using Mass Spectroscopy to study protein folding include the ability to detect molecules with different amounts of deuterium, which allows the heterogeneity of the protein folding reactions to be studied. It can also measure the conformation of folding intermediates bound to molecular chaperones without disrupting the complex. Mass spectrometry can also directly compare refolding properties, since mixtures of proteins can be studied without separation if the two proteins have sufficiently different molecular weights.

High Time Resolution: These are fast time-resolved techniques where a sample of unfolded protein is triggered to fold rapidly. The resulting dynamics are then studied. Ways to accomplish this include fast mixing of solutions, photochemical methods, and laser temperature jump spectroscopy.

Computational Prediction of Protein Tertiary Structure: This is a distinct form of protein structure analysis in that it involves protein folding. These programs can simulate the lengthy folding processes, provide information on statistical potential, and reproduce folding pathways.

Protein misfolding refers to the failure of a protein to achieve its tightly packed native conformation efficiently or the failure to maintain that conformation due to reduction in stability as a result of environmental change or mutation. It has been established that failure of protein folding is a general phenomenon at elevated temperatures and under other stressful circumstances. The two most common results of misfolded proteins are degradation and aggregation. When a polypeptide emerges from the cell, it may fold to the native state, degraded by proteolysis, or form aggregates with other molecules. Proteins are in constant dynamic equilibrium so even if the folding process is complete, unfolding in the cellular environment can occur. Unfolded proteins usually refold back into their native states but if control processes fail, misfolding leads to cellular malfunctioning and consequently diseases. Diseases associated with misfolding cover a wide array of pathological conditions such as cystic fibrosis where mutations in the gene encoding the results in a folding to a conformer whose secretion is prevented by quality-control mechanisms in the cell. About 50% of cancers are associated with mutations of the p53 protein that eventually lead to the loss of cell-cycle control and causing the growth of tumors. Failure of proteins to stay folded can result in aggregation, a common characteristic of a group of genetic, sporadic, and infectious conditions known as amyloidoses. Aggregation usually results in disordered species that can be degraded within the organism but it may also result in highly insoluble fibrils that accumulate in tissue. There are about twenty known diseases resulting from the formation of amyloid material including Alzheimers, Type II diabetes, and Parkinsons disease. Amyloid fibrils are ordered protein aggregates that have an extensive beta sheet structure due to intermolecular hydrogen bonds and have an overall similar appearance to the proteins they are derived from. The formation of the amyloid fibrils are the result of prolonged exposure to at least partially denatured conditions.

Alzheimer's: This neurological degeneration is caused by the accumulation of Plaques and Tangles in the nerve cells of the brain.[1] Plaques, composed of almost entirely a single protein, are aggregation of the protein beta-amyloid between the spaces of the nerve cells and Tangles are aggregation of the protein tau inside the nerve cells. Tangles are common in extensive nerve cell diseases whereas neuritic plaque is more specific to Alzheimer's. Although scientists are unsure what role Plaques and Tangles play in the formation of Alzheimer's, one theory is that these accumulated proteins impede the nerve cell's ability to communicate with each other and makes it difficult for them to survive. Studies have shown that Plaques and Tangles naturally occur as people age, but more formation is observed in people with Alzheimer's. The reasons for this increase is still unknown.

Creutzfeldt-Jakob Disease (Mad Cow Disease): This disease is caused by abnormal proteins called prions which eat away and form hole-like lesions in the brain. Prions (proteinaceous infectious virion) were discovered to be proteins with an altered conformation. Scientists hypothesize that these infectious agents could bind to other similar proteins and induce a change in their conformation as well, propagating new, infectious proteins.[2] Prions are highly resistant to heat, ultraviolet light, and radiation which makes them difficult to be eliminated. In Creutzfeldt-Jakob Disease there is an incubation period for years which is then followed by rapid progression of depression, difficulty walking, dementia and death. Currently there is no effective treatment for prion diseases and all are fatal.[3]

Parkinson's disease:A mutation in the gene which codes for alpha-synuclein is the cause of some rare cases of familial forms of Parkinson's disease. Three point mutations have been identified thus far: A53T, A30P and E46K. Also, duplication and triplication of the gene may be the cause of other lineages of Parkinson's disease.Victims of Parkinson's disease have primary symptoms that result from decreased stimulation of the motor cortex by the basal ganglia, normally caused by the insufficient formation and action of dopamine. Dopamines are produced in the dopaminergic neurons of the brain. People who suffer from this disease have brain cell loss (death of dopaminergic neurons), which may be caused by abnormal accumulation of the protein alpha-synucleinbinding to ubiquitin in the damaged cells. This makes the alpha-synuclein-ubiquitin complex unable to be directed to the proteosome. New research shows that the mistransportation of proteins between endoplasmic reticulum and the Golgi apparatus might be the cause of losing dopaminergic neurons by alpha-synuclein.

Cystic Fibrosis: Francis Collins first identified the hereditary genetic mutation in 1989. The problem occurs in the regulator cystic fibrosis transmembrane conductance regulator (CFTR), which regulates salt levels and prevents bacterial growth, when the dissociation of CFTR is disturbed as a protein regulating the chloride ion transport across the cell membrane.[4] The deleted amino acid doesn't allow bacteria in the lungs to be killed thereby causing chronic lung infections eventually leading to an early death.[5] Scientists have used nuclear magnetic resonance spectroscopy (NMR) to study Cystic Fibrosis and its effects.

Sickle Cell Anemia: Sickle-shaped red blood cells cling to walls in narrow blood vessels obstructing the flow of blood define sickle cell anemia. The shortage of red blood cells in the blood stream in addition to the lack of oxygen-carrying blood causes serious medical problems. The defect in the Hemoglobin gene is detected with the presence of two defective inherited genes. The sickle cell shape is formed as hemoglobin give up their oxygen resulting in stiff red blood cells forming rod-like structures. Some symptoms include: fatigue, shortness of breath, pain to any joint or body organ lasting for varying amounts of time, eye problems potentially leading to blindness, and yellowing of the skin and eyes which is due to the rapid breakdown of red blood cells. Luckily, sickle cell anemia can be detected by a simple blood test via hemoglobin electrophoresis. Even though there is no cure, blood transfusions, oral antibiotics, and hydroxyurea are treatments that reduce pain caused.[6]

Huntington's Disease: Also known as the trinucleotide repeat disorder, Huntington's disease results from glutamine repeats in the Huntingtin protein. Roughly 40 or more copies of C-A-G (glutamine) will result in Huntington's disease as the normal amount is between 10 and 35 copies. During the post-translational modification of mutated Huntingtin protein(mHTT), small fractions of polyglutamine expansions misfold to form inclusion bodies. Inclusion bodies are toxic for brain cell. This alteration of the Huntingtin protein does not have a definite effect except that it affects nerve cell function.[7] This incurable disease affects muscle coordination and some cognitive functions.

Cataracts: Eye lens are made up of proteins called crystallins. Crystallins have a jelly-like texture in a lens cytoplasm. The current leading cause of blindness in the world, cataracts occurs when crystallin molecules form aggregates scattering visible light causing the lens of the eye to become cloudy. UV light and oxidizing agents are thought to contribute to cataracts as they may chemically modify crystallins. In children, it has been observed that the deletion or mutation of B-crystallin facilitates cataracts formation. The likelihood of developing cataracts exponentially increases with age. Pain, Roger H. (2000). Mechanisms of Protein Folding. Oxford University Press. pp.420421. ISBN019963788. http://books.google.com/books?id=DvJygJkNCYkC&pg=PA420&lpg=PA420&dq=cataract+protein+folding&source=bl&ots=lDazpccGH2&sig=aHxuSkC1XNmcOnJYnmW4rZPuUvg&hl=en&ei=Z7rbSv3_OJG-sgOvpOGRBg&sa=X&oi=book_result&ct=result&resnum=2&ved=0CBUQ6AEwAQ#v=onepage&q=cataract%20protein%20folding&f=false. Retrieved 2009-10-18.

Protein misfolding caused by impairment in folding efficiency leads to a reduction in number of the proteins available to conduct its normal role and formation of amyloid fibrils, protein structures that aggregate, resulting in a cross- structure that can generate numerous biological functions. Protein aggregation can come from different processes occurring after translation including the increase in likelihood of degradation through the quality control system of the endoplasmic reticulum (ER), improper protein trafficking, or conversion of specific peptides and proteins from its soluble functional states into their highly organized aggregate fibrils.

Structures

X-ray Crystallography

From X-ray crystallography, three-dimensional crystals of amyloid fibril structures were formed and the structure of the peptide formation and how the molecule is packed together were examined. In one particular fragment, the crystal was found to contain parts of parallel -sheets where each peptide contributes one single -strand. The -strands are stacked and -sheets formed are parallel and side chains Asn2, Gln4 and Asn6 interact with each other in a way that water is kept out of the area in between the two -sheets with the rest of the side chains on the outside are hydrated and further away from the next -sheet.

Solid State Nuclear Magnetic Resonance (SSNMR)

Through solid-state nuclear magnetic resonance (SSNMR) and the help of other methods such as computational energy minimization, electron paramagnetic resonance and site-directed fluorescence labeling and hydrogen-deuterium exchange, mass spectrometry, limited proteolysis and proline-scanning mutagenesis the structure of an amyloid fibril was suggested to be four -sheets separated by approximately 10.

Through NMR with computational energy minimization, a 40-residue form of amyloid peptide at pH 7.4 and 24Celius was determined to contribute one pair of -strand to the core of the fibril which is connected by a protein loop. The amyloid peptides are stacked on each other in a parallel fashion.

From experiments of site-directed spin labeling coupled to electron paramagnetic resonance (SDSL-EPR), the molecule was found to be very structured in the fibrils and in parallel arrangement. SDSL-EPR along with hydrogen-deuterium exchange, mass spectrometry, limited proteolysis and proline-scanning mutagenesis suggests that the structure has high flexibility and exposure to solvent of N-terminal side, but is rigid in the other parts of the structure.

Experiments through SSNMR with fluorescence labeling and hydrogen-deuterium exchange determined that the C-terminals are involved in the core of the fibril structure with each molecule contributing four -strands with strands one and three forming one -sheet and strands two and four forming another -sheet about 10 apart.

Further experimentation approaching the atomic level with SSNMR techniques resulted in very narrow resonance lines in the spectra, showing that the molecules within fibrils hold some uniformity with peptides that display extended -strands with the fibrils.

Conclusion

The structures determined from X-ray crystallography or SSNMR were similar to previously proposed structures from cryo-electron microscopy (EM) formed from insulin. EM, which uses electron density maps, revealed untwisted -sheets in the structure. The similarities of the structures found in these experiments suggest a lot of amyloid fibrils can have similar characteristics such as the side-chain packing, aligning of -strands and separation of the -sheets.[8] Annu. Rev. Biochem. 2006.75:333-366. http://www.annualreviews.org. Retrieved 24 Oct 2011

Formation

The capability to form amyloidal protein structures that are considered to be genetic is from the findings that an increasing number of proteins show no signs of protein related diseases. It has been found that amyloidal proteins can be converted from its own protein that has a function rather than disease- related characteristics in living organisms.

In these protein mutations, different factors that affect the formation of amyloid fibril formation and different chains form amyloid fibrils at different speeds. In different polypeptide molecules, hydrophobicity, hydrophillicity, changes in charge, degree of exposure to solvent, the number of aromatic side chains, surface area, and dipole moment can affect the rate of aggregation of protein. It has been found that the concentration of protein, pH and ionic strength of the solution the protein is in as well as the amino acid sequence it is in determines the aggregation rate from the unstructured, non-homologous protein sequences.

As the hydrophobicity of the side chains increases or decreases can change the tendency for the protein to aggregate.

Charge in a protein can create aggregations through interaction of the polypeptide chain with other macromolecules around it. Also, the low tendency for -sheets to form along with the high tendency for -helixes to form contributes in facilitating amyloid formation.

It was found that the degree in which the protein sequence are exposed to solvent tend to affect the formation of amyloids. Proteins that are exposed to solvent seem to promote aggregation. Even though some other parts of the protein that had a high tendency to aggregate were not involved in the aggregation, they seem to at least be partially unexposed to the solvent but other regions that were exposed to solvent that were not involved in the aggregation had a low tendency to form amyloid fibrils.

It has even been raised that protein sequences have evolved over time to avoid forming clusters of hydrophobic residues by alternating the patterns of hydrophobic and hydrophillic regions to lower the tendency for protein aggregation to occur.[8]

The Affects of Sequence on the Formation of Amyloid Proteins

Amyloid formation arises mostly from the properties of the polypeptide chain that are similar in all peptides and proteins, but sometimes, the sequence affects the relative stabilities of the conformational states of the molecules. In that case, the polypeptide chains with different sequences form amyloid fibrils at various rates. Sequence difference affects the behavior of the protein aggression instead of affecting the stability of the protein fold. Various physicochemical factors affect the formation of amyloid structure by unfolded polypeptide chains.

Hydrophobicity of the side chains affects the aggregation of unfolded polypeptide chains. The amino acid in the regions of the aggregation site can change the ability of aggregation of a sequence when they increase or decrease the hydrophobicity at the site of the mutation or folding site. Over time, sequences have evolved to avoid creating clumps of hydrophobic residues by alternating hydrophobic areas of the protein.

Charge affects the aggregation of amyloid protein folding. A high net charge can have the possibility of impeding self association of the protein. Mutations in decreasing the positive net charge may result in the opposite effect of aggregate formation as increasing the positive net charge. It has been seen found that polypeptide chains can be run by interactions with highly charged macromolecules, displaying the importance of charge of a protein aggregation.

Secondary structures of proteins affect the amyloid aggregation as well. Studies show that a low probability to form -helix structures and a high probability to form -sheet structures are contributive factors to amyloid formation. However, it has been found that -sheet formation is not particularly favored by nature since there are little alternation of hydrophilic and hydrophobic residue sequence patterns to be found.

The characteristics of the amino acid sequences affect the amyloid fibril structure and rate of aggregation. Different mutations, including changes in the number of aromatic side chains, the amount of exposed surface area and dipole moment, have been said to change the aggregation rates of lots of polypeptide chains.

Unfolded regions play vital roles in promoting the aggregation of partially folded proteins. Some regions that were found to be flexible or exposed to solvent were fond of aggregation. Other regions that are not involved in the aggregation were found to not be exposed, but rather half buried even though they have high possibility of aggregating while the exposed regions of the structure that are not involved in the aggregation have a low probability of aggregating amyloid fibrils. The fibrils tend to come together by association of unfolded polypeptide segments rather than by docking the structural elements.

Overall, it has been found that unfolded proteins have lower less hydrophobicity and higher net charge than that of a folded protein. Residues that tend not to form the secondary structure of -sheet structured proteins seem to inhibit the occurrence of amyloid aggregation. Concentration of protein, pH and ionic strength were found to be associated with the amino acid sequence, which affects the rate of aggregation.

[8]

It is understood that the primary structure (the amino acid sequence) of a protein predisposes the protein for a specific three dimensional structure and how it will fold from the unfolded form to the native state. The concentration of salts, the temperature, the nature of the primary solvent, macromolecular crowding, and the presence of chaperones are all factors that affect the mechanism of folding and the ratio of unfolded proteins to those in the native state. More than anything, these environmental factors affect the likelihood of any single protein reaching the correct final structure.

Isolated proteins placed in proper environments (specific solvent, solute concentrations, pH, temperature, etc.) tend to self-fold into the correct native conformation. Altering any of these environmental characteristics can disrupt the structure and/or interfere with the folding mechanism. A pH outside the normal range of a given protein can ionize specific amino acids or interfere with both polar and dipole-dipole intramolecular forces that would otherwise stabilize the structure. Excess heat (cooking) proteins can break hydrogen bonds essential to the secondary structure of proteins.

Extreme environments or the presence of chemical denaturants (such as reducing agents that can break disulfide bonds) can cause proteins to denature and lose its secondary and tertiary structure, forming into a random coil. Under certain conditions fully denatured proteins can return to their native state. Intentional denaturing is used in various methods to analyze biomolecules.

The complex environments within cells often necessitate chaperones and other biomolecules for proteins to properly form the native state.

Protein is an essential part of living thing. The development of human body is needed to be parallel with the development of protein. But protein contains so many mysteries that we did not discovery yet. For example, that is protein folding. Folding is a necessary activity of proteins. They need to fold to continue their biological activity. Folding is also a process that very protein goes through to have a stable conformation. But sometimes this process is happened incorrectly, and the scientist call this problem is protein misfolding. The results of protein folding incorrectly are so many bad diseases happening for human, animals and living things such as Alzheimers disease and Mad Cow disease. Because of this reason, the researches about protein folding and misfolding become very important. During the process of discovering about protein, folding, misfolding and its affects, the scientists have been collecting many successes; the mystery about protein is unraveled gradually. As a scientist, W. A. (Bill) Thomasson records many importance things about protein in the article Unraveling the Mystery of Protein Folding; in this article, he make the points about Alzheimers disease and Mad Cow disease and some affects of protein misfolding beside the successes of science about them.Dr Thomasson begins his article by introduce generally about protein folding and misfolding. First of all, proteins consists the sequences of amino acid. The scientists have discovered 20 amino acids appearing in proteins. The protein structure is known with 2 basic shapes which are _helix and _sheet. Most of proteins probably go through several intermediate states on their way to a stable conformation (Campbell and Reece, 79). Proteins need to fold to continue its activity. The scientists have listed 3 type of protein folding; the protein can be folded, partial folded or misfolded. In the process of folding, the proteins called chaperones are associated with the target protein; however once folding is complete (or even before) the chaperone will leave its current protein molecule and go on to support the folding of another (Thomasson). The author of the article records the very important conclusion of Anfinsen about protein misfolding. In his point of view, the misfolding is occurred in the process of folding when the folding goes wrong. The research of protein misfolding is focus on the temperature sensitive mutation; the scientists observe the bacteriophage P22 with the changing of temperature to cause the mutation. And they conclude that the mutant proteins are less stable than the normal. It means, they give a conclusion is that in the tailspike of bacteriophage the misfolded proteins is less stable than the correctly folded proteins and they are difficult to reach the properly folded state. When the protein misfolding occurs, it results many bad disease. The aggregation can appear along with the appearance of misfolding and it is at the brain to cause Alzheimers disease and Mad Cow disease as many scientists consider. One affect of protein misfolding on human life that is Alzheimers disease. This is a disease of the elderly. According to the research of scientist, this disease is occurred when the amyloid precursor protein is misfolding. This protein is processed into a soluble peptide A. The scientists have not known exactly the reason of this disease yet. But the main reason causing the misfolding is the protein apolipoprotein E (apoE) inside our blood stream. The protein apoE has three forms such as apoE2, apoE3 and apoE4. The affects of each form of apoE on the A is not discovered yet but the scientists consider that the apoE can bind to the A. In the process of misfolding, the -amyloid is formed to make neuritic plaque in the Alzheimers patient. This disease is just happened with the older people because in the amyloid process, a nucleus is formed very slowly. The mutation of this protein is not stable and causes the disease. The studying about apoE is still a secret because some scientists show that one form of this protein is developing the disease but another form is decreasing the development of the disease. Finally, the research about Alzheimers disease is continued in order to affirm the results of protein apoE on A and to find the treatment for this disease successfully. Another affect from the protein misfolding is the Mad Cow disease. This is a very dangerous disease because it can be transmitted from animals to human. This disease causes by the misfolding of prions. The process of misfolding is the self-replicating of the prions. Prions are protein particles containing DNA and RNA. The mutation appear in the process of folding, the prions self-replicate and cause the misfolding of the proteins. They contain DNA and RNA. This is a special situation of the protein; it can be served as its chaperons. Because of the replicating, the prion was multiplied very quickly along with the increasing of normal proteins. This disease shows that the protein folding can be occurred without the genetics such as the experiment on the sheep. Dr. Thomasson continues his article by some more information about the misfolding and the way of the scientist to prove the mystery. He gives the information about the protein p53 and its mutation. It can cause the cancer, it also one type of protein misfolding. The point Dr. Thomasson wants to make that is his idea about the drug that can make the protein misfolding becoming more stable and minimize the misfolding of protein. This idea seems very good but its results are like a mystery as the mystery of protein folding. The research about the protein folding is very important to our lives. The misfolding is one of the main reasons causing so many dangerous disease but we did not have a successful treatment yet. The study of protein folding is more and more successful to help the human to be able to destroy the disease causing by misfolding. The disease caused by protein misfolding has become one problem of human that need to be solved.

Molecular Chaperones are known mainly for assisting the folding of proteins. Chaperones are not just involved in the initial stages of a proteins life. Molecular Chaperones are involved in producing, maintaining, and recycling the structure and units of protein chaperones. Chaperones are present in the cytosol but are also present in cellular compartment such as the membrane bounded mitochondria and endoplasmic reticulum. The role or necessity of chaperones to the proper folding of proteins varies. Many prokaryotes have few chaperones and less redundancy in the types of chaperones and whereas eukaryotes have large families of chaperones containing some redundancy. It is hypothesized that some chaperones are essential to proper protein folding such as the example of the prokaryote which has less variations of a chaperone family available. Other chaperones play less of an essential role such as in eukaryotes where more variations within a family of chaperones exist and gradients of efficiency or affinity are produced. This redundancy or existence of less efficient chaperones may exist in one state but the effectiveness of chaperones is also a function of their environment. The pH, space, temperature, protein aggregation and other external factors may render a chaperone that was once ineffective into a more essential chaperone. These environmental factors show why it is important to simulate cellular in vivo conditions, or native states, in order to grasp the conditions that require use of chaperones. This briefly summarizes the difficulties in analyzing and comparing chaperone function in vivo vs. in vitro.Simulating in vivo, or the environment within the cell, is important not just because of physical factors such as pH or temperature but also because the time in which the chaperone begins to conform the polypeptide. Some chaperones are nearby the ribosome and attach immediately to the polypeptide to prevent misconformation. Other chaperones allow the polypeptide to begin folding by itself and attach later on. Thus the role of each chaperone becomes specific to its vicinity to the polypeptide and time and place in which it assists folding. Recent research has implicated that chaperones within the nucleolus not only catalyze protein folding but also catalyze other functions important to maintain a healthy cell. These nucleolar chaperones are called Nucleolar Multitasking Proteins (NoMP's). Heat shock proteins, for example, not only help other proteins fold but also act during moments of stress to regulate protein homeostatis. Furthermore, there is evidence that chaperones work together in networks to oversee certain functions like dealing with toxins, starvation or infection.

The nucleolar chaperone network is divided into different branches that have specific functions. The network is dynamic and can vary in concentration or location of the network components depending on changes in the physiology and environment of the cell. Heat shock proteins (HSPs), which are classified based on their molecular weights, are integral components of the chaperone network. HSP 70s and 90s maintain proteostasis by ensuring that proteins are properly folded and preventing proteotoxicity, which is the damage of a cell function due to a misfolded protein. HSP70s help to fold recently synthesized proteins, while HSP90s help later in the folding process. The nucleolar network also contains chaperones that are part of ribosome biogenesis, or the synthesis of ribosomes in the cells. Proteins in the HSP70 and DNAJ families, which help to process pre-rRNA, are regularly found in protein complexes that process pre-rRNA in Saccharomyces cerevisiae (a species of yeast). Other HSPs are important in ribosome biogenesis as well, including HSP90 which works together with TAH1 and PIH1 to create small nucleolar ribonucleoproteins. The nucleolar chaperone network provide the organization and assistance needed to complete the biological taks necessary for cell survival, and if it does not function properly there can be many problems. For instance, when cancer cells have increased levels of rRNA synthesis, ribosome biogenesis is increased. Scientists are researching the compound CX-3543, which can stop nucleolin from binding with rDNA and impede RNA synthesis, leading to cell death. It is possible to potentially use drugs designed to target specific branches of the nucleolar chaperone network in malfunctioning cells. Other networks of chaperones include networks that specifically participate in de novo protein folding, meaning they help to fold newly made proteins, and the refolding of proteins that have been damaged. One chaperone network that exists in tumor cell mitochondria contains HSP90 and TRAP1, which protect the mitochondria and prevent cell death, allowing the cancer cells to continue to spread uncontrollably.[9]

HSP 70 is a protein in the Heat Shock Protein family along with HSP 90. It works together with HSP 90 to support protein homeostasis. It binds to newly synthesized proteins early in the folding process. It has three major domains, the N-terminal ATPase domain, the Substrate binding domain, and C-terminal domain. The N-terminal ATPase binds and hydrolyzes ATP, the substrate binding domain hold an affinity for neutral, hydrophobic amino acid residues up to seven residues in length while the c-terminal domain acts as a sort of lid for the substrate binding domain. This lid is open when HSP 70 is ATP bound and closes when hsp 70 is ADP bound. HSP70, or DnaK, are bacterial chaperones and can help in folding by clamping down on a peptide.[10]

GroEL and GroES, or 60kDa and 10kDa, are both bacterial chaperones. Both GroEL and GroES are structured so that they are a stacked ring with an empty center. The protein fits in this hollow center. Conformational changes within the chamber can then change the shape and folding of the protein.[10]

HSP 90 is a protein in the Heat Shock Protein family. This particular protein, however, is different from other chaperones in that HSP90 is limited in the folding aspect of molecular chaperones. Instead, Hsp 90 is vital to study and understand because many cancer cells have been able to take over and utilize the Hsp 90 in order to survive in many virulent surroundings. Therefore, if one were to structurally study and somehow target Hsp90 inhibitors, then there could be a way to stop cancer cells from spreading. Furthermore, many studies have been performed in order to test whether or not the Hsp 90 chaperone cycle is driven by ATP binding and hydrolysis or some other factor. But after much research by Southworth and Agard, there was enough evidence to state that HSP90 protein could conformationally change without nucleotide binding but rather the stabilization of an equilibrium is the factor that will change the Hsp90 to a closed or compact or open state. The three conformations of the Hsp90 were found through x-ray crystallography and also through single electron particle microscopy and by studying the three-state conformational changes in yeast Hsp90, human Hsp90 and bacteria Hsp 90 (HtpG) it was clear that there are distinct conformational changes for specific species. Overall, Hsp90 is a chaperone that is more involved with maintaining homeostasis within a cell rather than the involvement of protein folding. Hsp90 has rising potential in the area of drug development in the future since it plays such an essential role in aiding the survival for cancer cells.

This is the first chaperone to interact with the nascent chain as it exits the ribosome tunnel. Without the nascent chain, the TF cycles on and off but once the nascent chain is present, it binds onto the chain, forming a protecting cavity around. In order to do its function, TF scans for any exposed hydrophobic segment of the nascent chain and it can also re-associate with the chain. Folding is found to be more efficient in the presence of the TF, however, this is done at the expense of speed, it can stay with the chain for more than 30 seconds. The release of the chain is triggered when the hydrophobic portions is buried as the folding progresses toward the native state.

YidC, Alb3, and Oxa1 are proteins that facilitate the insertion of proteins in the plasma membrane. YidC is a protein that has only two polypeptide chains. The formation of its structure has been supported by particular phospholipids. YidC proteins can be found in Gram-negative and Gram-positive bacteria. Oxa1 can be found in the inner membrane of the mitochondria. Alb3 locates in the membrane of the thylakoid inside the chloroplast. Experiments showed that YidC protein actively contributes to the insertion of Pf3 coat protein. In addition, YidC also has direct contact with the hydrophobic segment of Pf3 coat protein. Although Oxa1 can only be found in the mitochondria it can also facilitate the insertion of membrane proteins in the nucleus. The role of YidC and Alb3 seems to be interchangeable because Alb3 can replace YidC in E. coli. Moreover, YidC, Oxa1, and Alb3 all support the insertion of Sec-independent proteins. Oxa1 only supports the insertion of Sec-independent proteins because the mitochondria in yeast cell do not have Sec proteins.

Nucleotide-binding domains that are leucine- rich (NLR) provide a pathogen-sensing mechanism that is present in both plants and animals. They could either be triggered directly or indirectly by a derivation of pathogen molecules via elusive mechanisms. Researches show that molecular chaperones like HSP90, SGT1, and RAR1 are main stabilizing components for NLR proteins. HSP90 can monitor the function of its corresponding clients that apply to NLR proteins in three practical ways: promotion of steady-state of functional threshold, activating stimulus-dependent activity, and raising the capacity to evolve.

Plants contain many NLR genes that considered being polymorphic in the LRR domain in order to be familiar with the highly diversified pathogen effectors. The NLR sensor stability will be the mechanism that will determine the pathogen recognition. The HSP90 system is advantageous for plants because it will couple metastable NLR proteins and stabilize them in a signaling competent condition. This will allow for the masking of mutations that would be detrimental.

It is known that chaperones work together to aid in the folding of protein in order to prevent misfolding. However, the mechanism of how chaperones help in protein folding was not fully understood. Recent studies on Hsp40 and Hsp70 have provided more insights into the mechanism of chaperones and their substrate. The Hsp40 family consists of many Hsp40 with different J-domain. Different J-domain will carry out different Hsp70 ATPase activities when Hsp40 binds to Hsp70. In protein folding, an unfolded polypeptide binds to a Hsp40 co-chaparone. From there, the J-domain of Hsp40 binds to the nucleotide-binding domain (NBD) of Hsp70. A conformation change in the Hsp70 substrate-binding domain occurs when the hydrolysis of ATP to ADP takes place on the HSP70 NBD. This causes Hsp70 to have a higher affinity for the polypeptide substrate and unbind the substrate from Hsp40. When ADP is exchange for ATP, the polypeptide substrate is released from Hsp40. Studies have shown that nucleotide exchange factors make changes to the lobe on the Hsp70 ATPASE domain in way that decreases Hsp70s affinity for ADP. Once the polypeptide is released from Hsp70, it can fold to its native state or it can be refolded by the chaperones if there is a misfolding. If a polypeptide that is bounded to Hsp70 is recognized by E3 ubiquitin ligase CHIP, it will be degraded.[11]

See more here:

Structural Biochemistry/Proteins/Protein Folding ...

Food for the soul: Traditional gyza makers and eaters in Utsunomiya try to keep the dumplings rolling – The Japan Times

UTSUNOMIYA, TOCHIGI PREF. Albert Kuwano Bakonyvari beckons me inside his factorys steam chamber, where hundreds of plump, white, crescent-shaped gyza dumplings are neatly lined on stacks of trays fitted in steel racks, waiting to be sent to the freezer.

Want to try one? I bet you havent eaten gyza at this stage of production, says the tall Hungarian-American with a goatee.

As I bite into the chewy dough, the warm juice from the ground meat and finely chopped cabbage, chives and leeks spill into my mouth as the scent of garlic hits. Unlike some of the gyza Ive had in Tokyo eateries, its surprisingly flavorful even without dabbing it in the typical soy sauce, vinegar and chili oil (ryu) combination, a characteristic I later learn is part of what defines the gyza made in Utsunomiya, Tochigi Prefecture, where some restaurants advise against the overuse of condiments to avoid killing the flavor of the dumplings.

Weve put a lot of time and energy into the dough and I think the texture of our gyza is unique its firm but its delightful to eat at the same time, it doesnt fall apart but still has a softness, Kuwano, 54, says.

For the filling of the gyza, we use domestic ingredients. The meat, leeks and chives are from Tochigi Prefecture. We use garlic from Aomori Prefecture and cabbage from the Kanto area. We try to keep the flavor relatively simple so the taste of the vegetables can shine through.

This is the heart of gyza country, at the gyza capital of Japan that boasts the highest annual household expenditure on the finger-sized dumplings.

A successful branding campaign led by the city and local gyza restaurants and makers, combined with word-of-mouth recommendations and media exposure, has made Utsunomiya synonymous with the comfort food. Its a studied example of machiokoshi (town promotion), so successful, perhaps, that protecting the brand has become an occasional source of contention.

AQ Foods, founded by Hisashi and Ayako Kuwano in 1965 as Eikyu Shokuhin, has been running one of the citys largest wholesale gyza businesses, with its products being distributed to department stores and supermarkets across the nation. While the couple is now in semi-retirement, the business was handed down to their son-in-law, Albert, who arrived in Japan in 1991 and married the Kuwanos only daughter.

AQ now produces more than a dozen varieties of gyza and manj dumplings, with its flagship product being a hand-made jumbo gyza that weighs 50 grams more than double the size of a regular gyza.

You cant make quality gyza of that size using machines. Perhaps 35 grams is the limit, says Koichi Chiba, the manager of one of AQs factories in Utsunomiya, a land-locked city with a population of around 520,000. In an age of automation, Chiba says AQ is a rarity in that it still produces hand-made gyza alongside the machine-made variety.

A former itamae chef and a relative of Ayako Kuwano, Chiba and his brother joined AQ around two decades ago. The two apprenticed under the founding couple, with Chiba specializing in the art of making a balanced, flavorful filling that has just the right amount of salt, while his brother oversees the production of gyza wrappers from scratch.

For the jumbo gyza, we have people wrapping them individually and packing them in containers to be shipped, he says. Gyza this size requires real people folding the pleats to seal them. Its not a skill that can be learned overnight, he says, typically taking three months of training before the technique is mastered.

Gyza is made all over Japan, but the ones made in Utsunomiya are the best and I strive to improve our product everyday so I can say ours is No. 1.

Gyza traces its roots to Chinas jiaozi dumplings, which are thought to have originated during the Tang Dynasty (618-907), according to Hideaki Otsuka, a Tsukuba University researcher and expert on Chinese culture.

Dried jiaozi (mummified jiaozi, so to speak), had been excavated from the ruins of Dunhuang. Scholars were surprised to learn that a food so similar to todays jiaozi, with the typical features of the folded dumpling with pleats, a semicircular shape and pointed edges, was eaten in the Tang Dynasty (618-907), he said in a lecture he delivered at a food culture seminar hosted by soy sauce manufacturer Kikkoman Corp.s Tokyo headquarters.

Otsuka says an Edo Period (1603-1868) cookbook introduced three cooking methods for gyza deep-frying, pan-frying and steaming but at the time it was still seen as a Chinese dish and was not a common food.

Chinese cuisine, including jiaozi, began to be served in the Chinatowns of Yokohama, Kobe and other cities during the Meiji Era (1868-1912), but gyzas status wasnt elevated to that of fast food of choice until after World War II, when the repatriation of many Japanese from mainland China who were used to eating jiaozi sparked a gyza boom. But whereas boiled jiaozi is common in China, its birthplace, pan-fried gyza became the norm in Japan.

The first gyza restaurant in Utsunomiya is believed to have opened near Utsunomiya Station in 1952, followed by the predecessor to famed gyza chain Minmin. Other legendary gyza houses that still serve to this day followed, including Masashi and Koran.

Its unclear why gyza shops concentrated in Utsunomiya, although it may have to do with how the city produces many of the dishs essential ingredients of gyza, including pork, flour, cabbage and chives. In any case, Utsunomiya residents embraced the dumplings since 1987, when the then-Management and Coordination Agency began including foodstuff in its annual survey of consumer habits, the city has been repeatedly ranked as the nations top gyza consumer despite the fact that the results excluded gyza eaten in restaurants. The survey is based on average annual expenditure per household on ready-made gyza bought and taken home from supermarkets and gyza restaurants. That means if the figure included gyza consumed at the numerous gyza restaurants dotting the city, the number will likely be substantially higher.

An Utsunomiya city official saw the stats and came up with a plan to promote gyza as a tourist attraction, says Hoshimi Kawazu, a spokeswoman for the Utsunomiya Gyoza Association, an organization formed in 1993 with 38 gyza restaurants. The group became a cooperative association in 2001, and now boasts more than 90 gyza eateries and manufacturers as members.

Until gyza became Utsunomiyas buzzword, the primary tourist draw was the Oya stone mine, Kawazu says, but the quarry that once supplied the Oya stone used for numerous structures in Japan collapsed in 1989, and the city needed an alternative attraction.

In 1993, a TV Tokyo variety show ran a series of programs on Utsunomiyas love affair with gyza. Coupled with the launch of an annual gyza festival and the erection of the Venus of Gyza statue in 1994, Utsunomiya and its culinary pride were soon pushed into the national spotlight.

Utsunomiyas decades-long battle with rival Hamamatsu, Shizuoka Prefecture, over the title of No. 1 gyza-consuming city has also become an annual media event. Hamamatsu beat Utsunomiya in 2018, with households spending an average 3,501 annually on gyza, compared to 3,241 in Utsunomiya.

Until I moved to Utsunomiya 10 years ago, I wasnt aware how much gyza is a part of peoples lives, Kawazu says. I also think what distinguishes us from other regional revitalization projects is how we work hand in hand with the city office.

As the associations influence grew, however, it also began implementing stronger measures to protect its brand. The Utsunomiya Gyza Association was trademarked in 2001, followed by Utsunomiya gyza being trademarked in 2002. These actions were aimed at clamping down on low-quality gyza calling themselves Utsunomiya gyza despite being produced and sold in other prefectures, Kawazu says.

Legal action ensued over the usage of the brand name, and association members were asked to follow strict orders when utilizing the trademarked brand.

In 2017, the operator of Utsunomiya gyza chain Umaiya which isnt a member reached a settlement with the association after being sued for using Utsunomiya gyza in its logo. Umaiya offered to take down the word ganso, or original, from its product logo and apply for membership.

Then in May, Kuwanos AQ Foods was removed from the gyza association, despite being one of the oldest gyza manufacturers in the city.

We were asked to join the association to promote Utsunomiya gyza and make it a thing, Kuwano says. We had already been selling it all over the country as Utsunomiya gyza, and the founder of the association knew that and was eager for us to join since he wanted to use our connections.

For a long time, the partnership was fruitful, Kuwano says, as Utsunomiya cemented its status as the gyza mecca, drawing hordes of hungry tourists.

Unfortunately, all good things like that end up having people with different priorities, and eventually the association started to control the name of Utsunomiya and a lot of our salespeople who used to sell our products as Utsunomiya gyza were not allowed to do that anymore. The association started to say the name should only be used in Utsunomiya and not around the country, whereas our business is promoting it all across the country.

We just had very different paths that we ultimately took, so yeah, were not part of the association anymore.

Without discussing specific issues, Kawazu of the gyza association says these rules are aimed at controlling quality and to prevent outside makers from using the brand to lure customers.

Utsunomiya gyza is such a broad term that it makes it difficult to trace who the producer is when issues surface if its used indiscriminately.

Dumplings are universal, as seen in their many variations, including the South Asian samosa and momo, and European pierogi and ravioli, says Izumi Kitta, a voice actress and gyza specialist who also serves as a special ambassador for Utsunomiya gyza.

So, in a sense, it was only natural for it to acquire comfort food status in Japan, she says, adding that she believes Utsunomiyas public relations campaign helped raise its national prominence.

Kitta, who is currently studying in the United Kingdom, says there were times she would eat gyza three times a day back in Japan. I think I was eating around 400 meals of gyza in 365 days, she says. Now that she lives in England, she goes out hunting for restaurants serving gyza and hosts gyza parties with friends. It made me want to spread the gospel of gyza across the world, she says.

Gyza has come a long way from its humble beginnings to become firmly embedded in Japans culinary psyche. For Hisashi and Ayako Kuwano, their encounter with gyza was life-changing.

Hisashi, 82, met Ayako, 84, when he was employed as a chef at a Japanese restaurant in Ishinomaki, Miyagi Prefecture. The two soon fell in love and ran off together to Tokyo, where Hisashi hopped around eateries. They ran a food stall, and at one point even cruised around the capital selling fish on a bicycle, but decided to settle down in Tochigi Prefecture, where Hisashi hails from, when their daughter was born.

The couple opened a restaurant in Utsunomiya in 1963 that served popular standard fare such as ramen and chhan fried rice. One of the regulars was a factory manager for Utsunomiya-based ice cream maker Futaba Foods Co. who suggested the restaurant serve frozen gyza dumplings Futaba was making to compensate for the fall in ice cream sales during the winter season.

However, Futaba would halt the production of frozen gyza once spring came, so we decided to make our own to satisfy our customers needs, Ayako says. The couples gyza was a hit, and they eventually began selling them to other restaurants, and later established a factory.

It wasnt all smooth sailing, however, and at one point the business was on the verge of collapsing when the price of cabbage soared.

We fell from the mountaintop to the bottom of the canyon, Ayako says. My husband suggested we get a divorce so I wont have to be burdened by debt, but I couldnt imagine my family being separated. In the end, we somehow managed to crawl back up.

The business persevered, and the number of employees grew. Albert, a California native, married the Kuwanos daughter in 1997 and later took over the gyza business.

I think my father-in-law stumbled upon the perfect product because gyza has every major food group you get your carbs, protein, vegetables and it happens to taste very good, Albert says. Its very satisfying but not expensive, so it kind of hits every point.

In 2012, Kuwano launched a craft beer importing business called AQ Bevolution, and opened the Titans Craft Beer Taproom & Bottle Shop in Tokyos Otsuka neighborhood to showcase the beer the company imports and, guess what? AQ Foods gyza.

We have our gyza and beer at one location, Kuwano says. And, as you may know, gyza and beer go well together.

The laid-back American may be an unlikely heir to a Japanese family-run gyza business, but Ayako and Hisashi appreciate his efforts to keep the company going despite the occasional ups and downs.

Asked whether he had any reservations having a foreign national marry their only daughter, Hisashi shakes his head.

Really? Ive heard you didnt want her to marry a gangster or a foreigner, Albert quips.

Hmm, perhaps, Hisashi says with a grin, his eyes wandering as if to recall a distant memory. However, he doesnt forget to add, Were grateful to have you.

Read the rest here:

Food for the soul: Traditional gyza makers and eaters in Utsunomiya try to keep the dumplings rolling - The Japan Times

Molecular Biology 02: ‘Thermodynamics of protein folding’

These are my notes from lecture 02 of Harvards BCMP 200: Molecular Biology course, delivered by Joe Loparo on September 5, 2014.

Continued from lecture 01. is always 0 or +180. If you plot and you find only a few clusters are well-represented: a range of -helix combinations, a -sheet area, and a third rarer area (called L and populated by left-handed -helices). is ususally found in the trans conformation due to steric hindrance of the consecutive side chains, however, proline because it is anchored to the backbone has a unique twist that enables a cis conformation.

-helices and -sheets are two ways of allowing the NH and C=O groups on the backbone to form hydrogen bonds. -helices contain 3.6 residues per rotation, or in other words, each residue spans 100 of rotation. Consecutive rungs of an -helix turns are separated by 5.4. -helices are almost exclusively right-handed. In a right-handed -helix, you turn counter-clockwise as you go up. In a left-handed -helix you turn clockwise as you go up. Side chains point outward from the helix. If you plot out where each residue falls on the helix based on the 3.6 residues/turn rule, you find that amphipathic, half-buried helices have all the hydrophobic residues on one side and the hydrophilic ones on the other side. A fully buried helix will be all hydrophobic residues and a fully exposed helix will be all hydrophilic residues.

In -sheets, all potential H-bonds are satisfied except for the flanking strands at either end of the sheet. About 20% of -sheets found in nature are mixed parallel and anti-parallel, the other 80% are pure one or the other. -sheets are not flat, but pleated.

A single sheet or helix is not stable in water. Tertiary structure is the packing of these elements, and loops connecting them, onto each other.

There are two fundamental problems in protein folding:

As an example, consider the metalloprotease cleaveage of Notch to create the Notch intracellular domain (NICD), which then translocates to the nucleus and affects transcription. The proteolytic site of Notch is protected by Lin12/Notch repeats which are connected to the EGF repeats that interact with Notchs ligand. The ligand is believed to apply a force that unfolds this region, allowing cleavage. Mutations which destabilize this fold and result in constitutive activation cause tumors.

Thermodynamics can only describe whether a chemical reaction will occur spontaneously or not, not how fast it will occur (see Biochemistry 01).

The energy of a system is its capacity to do work.

U = q + w

Where U is internal energy, q is heat and w is work.

q := heat = C(Tf-Ti)

Where C is the heat capacity and f and i mean final and initial.

w := work = Fxx

Where F is force and x is displacement along the x axis.

If you dissolve urea in water at a 4M solution, it will dissolve spontaneously and the solution will become cold (just like guanidine, as I learned here).

Gibbs free energy is defined as:

G = H - TS

Where G, H, T and S are Gibbs free energy, enthalpy, temperature and entropy respectively.

G = H - TS

If G < 0 the reaction will proceed spontaneously.

In the urea example, H > 0 because energy is required to pull apart the interacting urea molecules, using heat from the water. Yet the reaction still occurs spontaneously because S > 0 by a lot - the urea solution is much more entropic than urea and water separately.

For the reaction A + B C + D, we define:

Keq = ([C]eq[D]eq)/([A]eq[B]eq)

Keq = e-G/RT

ATP is a special molecule: its hydrolysis into ADP is spontaneous at physiological concentrations of the reactants and products, i.e. G < 0 for this reaction:

ATP + H2O ADP + Pi

Le Chateliers principle says you could drive the reaction in reverse, making ATP spontaneously, simply by increasing the concentrations of the procuts. However [Pi] never gets high enough in the cell for ATP to be spontaneously generated from ADP. The unfavorable production of ATP is instead created via a coupled reaction with favorable reactions such as the release of protons across the mitochondrial membrane (see Biochemistry 08).

H := Enthalpy = U + PV

Where U, P and V are internal energy, pressure and volume.

In physiological conditions, changes in pressure and volume are almost always negligible, so H and U are closely coupled. In other words, in most biological systems, the enthalpy is equal to the internal energy.

People have developed molecular dynamics simulations of the fundamental atomic forces that determine a proteins enthalpy (dihedral angles, Van der Waals interactions, electrostatic interactions, etc) and attempt to minimize the energy to determine a proteins fold. But there are so many degrees of freedom that computational expense prohibits running the simulation long enough to find the lowest energy state. Still there are attempts, such as Folding@Home, Foldit, and D.E. Shaws Anton. Anton holds the record for the longest molecular dynamics simulation - it ran for some untold amount of time, calculating the energy a protein would have at every femtosecond or something, in order to simulate 1 millisecond of the proteins movement. Obviously, the time that Anton took to simulate that millisecond was more than a millisecond.

S := Entropy = kbln(W)

Where kb is Boltzmanns constant and W is the number of microstates that give rise to the macrostate of interest.

My favorite explanation of this is that given by Richard Feynman. When I read it, I understood for the first time how physical entropy and information entropy are the same concept:

So we now have to talk about what we mean by disorder and what we mean by order. Suppose we divide the space into little volume elements. If we have black and white molecules, how many ways could we distribute them among the volume elements so that white is on one side and black is on the other? On the other hand, how many ways could we distribute them with no restriction on which goes where? Clearly, there are many more ways to arrange them in the latter case. We measure disorder by the number of ways that the insides can be arranged, so that from the outside it looks the same. The logarithm of that number of ways is the entropy. The number of ways in the separated case is less, so the entropy is less, or the disorder is less.

Richard Feynman, quoted here

In biology, entropy is very often the driving force, for instance for the burial of hydrophobic protein domains. Imagine a water molecule in a tetrahedron. The tetrahedron has four corners, and the water has two hydrogens, so you can place the molecule in 4 choose 2 = 6 orientations. If you add a nonpolar group of a neighboring molecule at one corner of the tetrahedron, only three of the six states remain favorable (by still allowing hydrogen bonding). So Shydrophobic = kbln(3) - kbln(6) < 0, meaning that entropy has decreased.

Consider the mixing of epoxy and hardener into cured epoxy. This reaction has S < 0 because the solid has fewer microstates than the liquids did. Yet the reaction occurs spontaneously at room temperature, so it must be true that H < 0. Heat is therefore released - in fact, the reaction is extremely exothermic. Joe measured the temperature of 5-minute epoxy and it rose from 21C to >40C at the 5 minute mark.

An incorrect and simplistic view of protein folding is as follows. An unfolded protein has high configurational entropy but also high enthalpy because it has few stabilizing interactions. A folded protein has far less entropy, but also far less enthalpy. There is a tradeoff between H and S here. Note that because G = H - TS, increased temperature weights the S term more heavily, meaning that higher temperature favors unfolding.

That entire explanation only considers the energy of the protein and not that of the solvent. In fact, hydrophobic domains of a protein constrain the possible configurations of surrounding water (see explanation above), and so their burial upon folding increases the waters entropy. Moreover, it turns out that the hydrogen bonding of polar residues and the backbone is satisfied both in an unfolded state (by water) and in a folded state (by each other). Therefore enthalpy is zero sum, and protein folding is driven almost entirely by entropy.

Here is a description of a technique called differential scanning calorimetry. You apply equal amounts of heat to two solutions, one with only buffer and the other with buffer and protein, and you measure the temperature in each solution. Eventually the protein reaches its melting temperature Tm, where the protein is 50% folded and 50% unfolded and G = 0. At Tm, the melting of the protein aborbs lots of the applied heat, and so the temperature does not rise as much as it does in the buffer-only solution.

Another technique for measuring protein stability is the force required to unfold it using single molecule atomic force microscopy.

Common denaturants are urea and guanidine hydrochloride. Amazingly, we still do not know how they work. It is thought that they stabilize all constituent parts of the unfolded protein. Guanidine may surround those unfavorable hydrophobic domains of the protein but then expose its own hydrophilic side to water, so that the movement of the water is not constrained.

Continued here:

Molecular Biology 02: 'Thermodynamics of protein folding'

Geroscience and it’s Impact on the Human Healthspan: A podcast with John Newman – GeriPal – A Geriatrics and Palliative Care Blog

Ok, I'll admit it. When I hear the phrase "the biology of aging" I'm mentally preparing myself to only understand about 5% of what the presenter is going to talk about (that's on a good day). While I have words like telomeres, sirtuins, or senolytics memorized for the boards, I've never been able to see how this applies to my clinical practice as it always feels so theoretical. Well, today that changed for me thanks to our podcast interview with John Newman, a "geroscientist" and geriatrician here at UCSF and at the Buck Institute for Research on Aging.

In this podcast, John breaks down what geroscience is and how it impacts how we think about many age-related conditions and diseases. For example, rather than thinking about multimorbidity as the random collection of multiple different clinical problems, we can see it as an expression of the fundamental mechanisms of aging. This means, that rather than treating individuals diseases, targeting aging pathways may be a better way to prevent or ameliorate multimorbidity. We talk with John about this, and current trials underway to test this hypothesis, along with so much more!

If you're interested in taking a deeper dive in the subject, take a look at these papers that John co-authored:

You can also find us onYoutube!

Listen to GeriPal Podcasts on:

Alex: This is Alex Smith.

Eric: Alex, we have someone in our studio audience ... our office studio? Our studio-

Alex: Our office studio? We have John Newman, who's a geriatrician and geroscientist-

Eric: A gero-what?

Alex: A geroscientist-

Eric: A gero-what?

Alex: A geroscientist who has held a joint appointment between UCSF and the Buck Institute for Aging Research. Welcome to the GeriPal PodCast, John.

John: Thanks, guys. Thanks, Alex. Thanks, Eric.

Eric: I'm really interested in figuring out what a geroscientist is. But before we do that, can we have a little song request for Alex?

John: Yeah, what should we sing about? Do you know a little song called Who Wants to Live Forever?

Alex: Ah, more Queen.

Eric: Boy, you can never get enough Queen.

Alex: Maybe our audience can. (singing).

Eric: John Newman does, right? John Newman wants all of us to live forever. At least that's why I am currently, those who are watching this on YouTube, can see I am getting fresh stem cells from my baby farm that I get infused every day, so I can live forever.

John: We're transfusing you as we speak.

Eric: As we speak.

John: As we speak.

Eric: Yeah, I give John hefty amounts of money for those baby transfusions.

John: Hey, that deal's just for you, Eric. Don't go advertising me.

Eric: So John, geroscience. What is this field, and is it about living forever?

John: It is not about living forever. It's about living healthier, longer, and staying independent. So what is geroscience?

Eric: That, I have no idea.

Alex: That's, I don't know.

John: I thought I was here, so you guys would tell me.

Alex: Gero ... Gero-

Eric: Gero- so, older.

Alex: Ger, Jerry, Ben and Jerry's.

John: Aging ... aging science.

Alex: Aging science.

John: Aging science with a flavor of people.

Eric: So what is the difference between you and Alex? You're both researchers. Is Alex a geroscientist?

John: Well, geroscience is a, it's a made-up word which was coined for a new field, and a whole new idea, which is now reality. Which is going to sound a little crazy. Taking what we know about the biological mechanisms that drive aging. The biology of aging.

John: And not only understanding that, which itself sounds a little crazy. But actually turning that into therapies, to help to treat or prevent disease, or help to improve the lives, especially of older adults.

John: Geroscience is the idea of translational geriatrics, taking what we know about the basic science of the processes that drive aging. And turning it into therapies and helping to improve people's lives.

Eric: The processes that ... Okay. As I age, I start developing some chronic medical conditions. They start building up. I have more and more medical conditions. If only I could just focus on making sure I don't develop those, or I treat these well; like diabetes, COPD. Would I prevent aging? Is that the goal here?

John: Well, one place this comes from is, what's that common underlying factor that's putting you at risk for COPD and for diabetes and for Alzheimer's disease and for cardiovascular disease and for strokes? And for osteoarthritis and osteoporosis? And for almost everything that we treat.

Eric: Nacho cheese Doritos?

John: That's a big one! That's a big one.

Eric: But there's more.

John: But what's the common variable for all of those? We call these age-related chronic conditions or age-related diseases.

Eric: Yeah.

John: Because they're all driven by aging. The key thing is that's not an accident or it's not just like a probability thing or it's not just time passing. But it's actually the biological mechanisms that change in our bodies as we get older that make us what we perceive as older. There's a biology there. And that biology puts you at risk for all these different chronic diseases.

John: You could try to treat or prevent all of these individually. But, if you're not changing, if you're not affecting the aging that's driving all of them, there's a limit to how far you can go with that, or how effective it's going to be.

John: You prevent diabetes, but you get cancer. You cure Alzheimer's disease, which would be amazing, but then you have a stroke. But if you intervene on the aging that's behind all of this, then maybe you can slow or delay or prevent all of these together. That's the great hope of geroscience.

Alex: Is delay or preventing aging ... Earlier, you said that it's not so much about living longer, as it is improving healthy years of life. And yet, but what you just said was, "delay or prevent aging." I'm a little lost there. Can you help me out?

John: Well, delay or prevent all of the diseases and conditions that are driven by aging. So, targeting aging as the underlying biology that causes or contributes to diabetes, dementia, cancer, heart disease, and all of that.

John: All of that might wind up helping you live longer; who knows. But that's not the goal.

Alex: Ah.

John: The goal is to be healthier for longer.

Alex: Oh.

John: To spend, so we all can spend more years independent and being able to do the things we want to do in a state of better health.

Alex: It's like the principle of the double effect. For those palliative care listeners, right, the primary ... right? We're relating the subject to you.

Alex: The primary intended target in the principle of double effect in palliative care is often opioids for pain relief. And yet, as a secondary effect, if the patient dies sooner, that's acceptable.

Alex: Your primary intended effect is to treat the disease that are associated with aging; the conditions that are associated with aging. As a secondary effect, if people end up living longer, then that's fine too. But it's not the primary target of geroscience?

John: I love that analogy. Living longer may wind up being a side effect of being healthier longer. But when you talk about ... Aging is a weird subject, right? Because it's this, it's not a disease. It's not a bad thing. There are many many positive elements of aging. I am happier now than I was 20 years. Hopefully I'll be even happier 20 years from now, even if I can't jump as high.

John: Aging is not a disease. It's not a bad thing. And yet we're trying to target, almost to treat it. The goal is if we can slow or reduce the bad aspects of aging, the parts of aging that give rise to chronic disease. And you're living healthier longer, you'll probably live longer, too, in good health.

John: When I go to a room and I ask people, "We're talking about aging as a target for therapies. Who wants to live to be 200?"

John: Not a whole lot of people raise their hands, because most of them are thinking, "I'm not sure how I'm going to feel when I'm 95 or 85 or 75. You extrapolate that out, and what am I going to feel like when I'm 200?"

Alex: Right.

John: That doesn't seem like a great choice. But if you ask people, "What if you could have the health that you have now, or the health that you had when you're 60, or the health you had when you're 50? And just keep that for longer?"

Alex: Uh-huh.

John: Most people would volunteer for that.

Alex: Interesting.

Eric: I just want to make sure that when you ... When I hear "aging," I think probably the common definition is, I'm getting older.

Eric: When you hear the word "aging," what do you mean by aging? Especially as we're targeting aging? I can't target the clock; I guess I could target my clock and just turn it around. What do you mean when you say "aging"?

John: Well, here's the geriatrician's perspective. How do we know what "old" is, what an older adult is, for making a clinical decision, for example?

Eric: Yeah.

John: For thinking about prognosis, for example. There's someone I know who's done a lot of work around prognosis and how to estimate someone's life expectancy.

Alex: I don't know who you're talking about [laughter].

John: Yeah, the name, it's right on the tip of my tongue. So how do we know? Of course, we know that someone's birthday doesn't really help a whole lot with that stuff. There's 85-year-olds who are very fit and active and healthy and young. And there are 85-year-olds who are not very young. What makes that difference?

John: In geriatrics, we think of things like functional assessments. Mobility and ADL function, idea function, frailty, trying to get that certain aspect of what does it mean to be older, to have an advanced stage of aging? Geroscience and aging biology is sort of the molecular reflection of that.

John: If we know that you have two 85-year-olds, and one of them needs help with ADLs, they're going to be at risk for complications from surgery. But not necessarily because they need help with ADLs, but because that reflects their biology. The stage of their aging.

Alex: Right.

John: We're learning more and more about what that biology really is.

Alex: Right.

John: Is it their telomeres, for example? Like Eric said earlier. Or how many senescent cells they have in their body. Or what is their NAD reserve? What is the state of their chronic inflammation? How are their proteins folding? What is their proteostatic resilience?

John: We're getting closer to be able to understand what all of these biological aspects are, so we can look at someone who we think, "Is this person an old 85-year-old or a young 85-year-old?" And know what their biology tells us.

Alex: I don't want to ... I mean, I don't want to belabor this point. But I know some of our listeners are probably skeptics. We have one skeptic, a nurse practitioner on our hospice and palliative care service. Patrice Villars. We mentioned we were doing this podcast.

Alex: And she said, "Is it," something along the lines of, "is it morally responsible to focus on helping people to live longer, given the current burden of climate change that humans are placing on the planet currently, much less if we were to live longer lives?"

John: I love that question. I love ... Aging is a really unusual field to study, because it's, again, it's not a disease. It's something universal that happens to all of us. That does make it a little bit different, and opens up these really interesting questions about not only how to study it, and what our goals should be; but also how to ...

John: If we have therapies that effectively target aging, target mechanisms of aging, who gets them? How do we decide how to use them? How do we decide who should have them and who doesn't get them? Is that different from the way that we decide who gets other treatments? These are really interesting questions.

Eric: In the work that you do, I'm guessing ... because I think this is a really fascinating thing to do, like if you extend the life, even 20 years, the ethical issues that come up. Even from a population density standpoint, can our earth even handle that? Then who gets all these treatments? In the field of geroscience, are there ethicists in that field, too, that are looking at these questions?

John: Yeah. This is a brand-new field, an emerging field. There are ethicists who are particularly thinking about these questions. Not very many, in the same way there's not very many clinician scientists who are helping to develop and study these therapies.

John: But there are people who are just starting to really think about, because these questions; even five years ago, this would have felt like a really academic philosophical kind of discussion. Therapies that target aging. Sure, let's maybe 10 years, 20 years down the line.

John: But the crazy thing is the first clinical trial that takes a drug in this case, that targets a cellular mechanism of aging, was given to older adults to treat a chronic syndrome of aging. That clinical trial, that first clinical trial has already been done.

Eric: What are they doing?

John: This was a drug that targets protein quality control. It's a drug called Rapamycin and its related drugs. We can talk about how this fits into the bigger scheme of mechanisms of aging. But it helps to activate pathways in your cells that clean up misfolded proteins and help your cells to make proteins that are more functional.

John: This drug and a related drug were first given to older adults before flu vaccine, to see if it would improve response to the flu vaccine. And it did. Then the next step, they did a clinical trial where they treated people with these drugs for just a month, and then gave them a flu vaccine, and saw that it improved their response to a flu vaccine.

John: But then over the next six months, they saw how often they got respiratory or other infections. What they actually found was that the people who received this treatment just for a month had about a third fewer infections over the next six months. So it had this really interesting long-lasting effect on their immune function in these older adults. It was helping to ameliorate what we call immunosenescence, the decline in function of our immune systems as we get older.

John: It's a really small, limited thing. One drug, some older people, flu vaccine, it was just looking at infections. Doesn't seem like a big deal, and it's not. Except it was the first randomized controlled trial of a drug like this that targets mechanisms of aging in older adults, to improve syndromes of older adults.

Alex: And it's already happened.

John: And it's already happened.

Eric: I guess that's probably the hard part with this is that I would imagine you can't do a randomized controlled trial with this drug for 20-year-olds and wait 90 years to see what happens to them. And then market that drug after a hundred years.

John: Exactly. I mean, hey, the average R01 lasts for five years.

Read the original:

Geroscience and it's Impact on the Human Healthspan: A podcast with John Newman - GeriPal - A Geriatrics and Palliative Care Blog

Protein Folding – Anfinsen’s Experiment ~ Biology Exams 4 U

How Protein folds? During translation, the linear chain of amino acids formed will be gradually released from the ribosome, and these amino acids should fold properly to make a functional protein, the ultimate nano machines in the cells. Protein folding is undoubtedly the most critical events that determine the ability of that given protein to work properly.

How protein folds? Is it a random process? It shouldnt be, as folding determines the function.

Definition:

Protein folding refers to the set of ordered pathways by which protein folds into their native functional confirmation.

Protein folding is primarily driven by hydrophobic forces.

Anfinsen's Experiment

First step..

The first insight to this question was provided by Christian Anfinsen at the NIH. He was working on the properties of ribonuclease A (a single chain protein of 124 amino acids with 4 di-sulphide bonds). He unfolded (denatured) ribonuclease A using urea and mercaptoethanol (denaturants). The protein lost its function. Then he allowed to renature ribonuclease A by removing denaturants, and found out that ribonuclease A folded spontaneously and become functional. He concluded that Ribonuclease A can self assemble into its 3D functional structure.

Protein Folding inside the Cell

Inside the cell, protein folding is assisted by different proteins collectively called as accessory proteins.

The importance of studying protein folding?

Read the original:

Protein Folding - Anfinsen's Experiment ~ Biology Exams 4 U

Protein Folding – an overview | ScienceDirect Topics

8 INHIBITORS OF HEAT-SHOCK PROTEINS (HSP 90)

Protein folding is catalysed in vivo by isomerases and chaperone proteins. Molecular chaperones are ubiquitous proteins that assist folding, assembly, transport, and degradation of proteins within the cell. The first identified chaperones were heat-shock proteins (HSPs), whose names is derived from the elevated levels produced when cells are grown at higher-than-normal temperatures. HSPs stabilize other proteins during their synthesis and assist in protein folding by binding and releasing unfolded or misfolded proteins using an ATP-independent mechanism. Proteins unable to maintain their proper shape are broken down by the proteasome (see Section 1 of Chapter 10) and eliminated, as shown in Fig. 9.33. These events may be favourable if the proteins are previously mutated and hence dangerous for the survival of the cell, but they become a problem if the proteins are necessary for its normal functioning.

Figure 9.33. Function of heat-shock proteins.

HSP 90 is the best known of HSPs and its activity is coupled to an ATPase cycle that is controlled by several cofactors. It has three major domains, namely a highly conserved N-terminal ATPase domain, a middle domain, and a C-terminal dimerization domain. The crystal structure of HSP 90 bound to ATP has shown how this nucleotide is hydrolysed,135 but the detailed mechanism of protein folding remains unknown.

HSP 90 has emerged as an attractive cancer target because its inhibition blocks a large number of cancer-related signalling pathways since a large number of intra-cellular signalling molecules require association with HSP 90 to achieve their active conformation, correct cellular location, and stability.136 These include steroid hormone receptors, transcription factors like the tumor suppressor protein p53 and kinases like Src-kinase.

The conformational changes that take place in HSP 90 after binding and hydrolysis of ATP regulate the stabilization and maturation of client proteins, including hypoxia-inducible factor-1 (HIF-1), a relevant anticancer target.137 This ATP site is known by X-ray crystallography to be very different from that of kinases, allowing the design of inhibitors with high selectivity with regard to other ATP-binding proteins.

The design and study of selective inhibitors of HSP 90 was initially controversial because this protein is critical for the survival of both normal and sick cells. However, HSP does not have much activity under normal conditions. When the cell is under stress by genetic mutations or environmental changes such as heat or infection HSP 90 activity is increased as an emergency response that stabilizes partially unfolded proteins and helps them to achieve their correct shape. This activity also assists the survival of cancer cells despite an abundance of misfolded and unstable proteins, and this is one of the reasons to study HSP 90 as an anticancer target.

The main strategy employed in the design of HSP 90 inhibitors is based in the synthesis of analogues of the natural antitumor geldanamycin, a benzoquinone derivative belonging to the ansamycin class, although some companies working in this field are designing entirely synthetic molecules not related to this compound.

Geldanamycin was originally believed to be a TK inhibitor, but it was later identified as an ATP-competitive inhibitor of HSP 90. It could not be advanced to the clinical stage because it showed unacceptable hepatotoxicity, probably associated with the presence of the electrophilic methoxybenzoquinone moiety. For this reason, displacement of the 17-methoxy group by nucleophiles led to less toxic analogues such as tanespimycin (17-allylaminogeldanamycin, 17-AAG).138 Another problem associated with geldanamycin is its very low solubility, which was solved with the development of the water-soluble analogue alvespimycin (17-dimethylaminoethylaminogeldanamycin, 17-DMAG).139 Both analogues were better tolerated than the parent natural product and are under clinical trials. In another approach, the problematic quinone moiety of 17-AAG was reduced to the hydroquinone stage. The resulting compound, IPI-504, can be formulated as a soluble salt that is suitable for intravenous or oral formulations. It has shown encouraging results in Phase I trials in patients with gastrointestinal stromal tumors that were resistant to imatinib, although further clinical development is necessary.

See more here:

Protein Folding - an overview | ScienceDirect Topics

Rett Syndrome Tied to Altered Protein Levels in Brain in Early Study – Rett Syndrome News

Lack of a functional MeCP2 protein leads toRett syndrome by altering levels of brain proteins associated with energy metabolism and protein regulation, a study in a mouse model suggests.

These altered protein levels might also predict Rett syndromes progression, the investigators said.

The study, Brain protein changes in Mecp2 mouse mutant models: Effects on disease progression of Mecp2 brain specific gene reactivation, was published in theJournal of Proteomics.

Rett syndrome is caused by mutations in the MECP2gene that result in a missing functional MeCP2 protein, a regulator of gene expression. Despite prior studies in animal models, little research has focused on the effects of MeCP2 deficiency in the levels of other proteins in the brain, as well as in Rett syndromes progression.

Researchers from Italy used a mouse model of Rett to address this gap. They did a proteomic analysis of the brains of mice both before and after they developed symptoms, and compared the data to controls withoutMECP2mutations. (Proteomics is the large-scale study of proteins, conducted to draw more global conclusions than possible if assessing proteins one-by-one.)

Results showed abnormal levels of 20 brain proteins in symptomatic mice with Rett syndrome. Twelve of these proteins were overproduced, while eight were at lower levels compared to non-diseased control mice.

Notably, eight (40%) of these 20 proteins were involved in energy metabolism (the process by which cells get energy), and six (30%) were involved in proteostasis, which refers to cellular processes to ensure proper production and folding of proteins.

Presymptomatic mice showed abnormal levels in 18 proteins; 10 at low levels and 8 at high levels compared to controls. Similar to symptomatic mice, these proteins were primarily involved in energy metabolism and proteostasis.

The team then looked at mice that had been engineered to turn the MECP2 gene on in the brain, which was associated with mild symptoms and a longer life than otherwise expected.

By comparing animals lacking functional MeCP2 to mice with so-called MECP2 gene reactivation, the researchers worked to identify the proteins most directly impacted by missing MeCP2.

They found 12 proteins whose levels were normalized by gene reactivation. Seven of these proteins were at low levels and five at high levels without functional MeCP2 protein. Again, most were associated with energy metabolism and proteostasis, while two proteins were involved in how cells respond to oxidants reactive molecules that can damage DNA and cellular structures that is called redox regulation.

Only two of these 12 proteins, PYL2 and SODC, had been previously associated with Rett syndrome via earlier animal model studies that recorded altered levels in the brain.

Our findings suggest that RTT [Rett syndrome] is characterized by a complex metabolic dysfunction strictly related to energy metabolism, proteostasis processes pathways and redox regulation mechanisms, the researchers wrote.

Alteration in the evidenced cellular processes, brain pathways and molecular mechanisms [suggest] the possibility of the use of proteins as predictive biomarkers, they added.

Marisa holds an MS in Cellular and Molecular Pathology from the University of Pittsburgh, where she studied novel genetic drivers of ovarian cancer. She specializes in cancer biology, immunology, and genetics. Marisa began working with BioNews in 2018, and has written about science and health for SelfHacked and the Genetics Society of America. She also writes/composes musicals and coaches the University of Pittsburgh fencing club.

Read this article:

Rett Syndrome Tied to Altered Protein Levels in Brain in Early Study - Rett Syndrome News