Clinical Trials | Genetic Medicine

COPD RESEARCH STUDY

Biology of the Oral Epithelium of Electronic Cigarette Smokers

Little is known about the oral health risks of smoking electronic cigarettes, an increasingly popular form of inhaled alternative tobacco products. Based on the knowledge that adverse effects are observed at the biologic level long before clinical abnormalities manifest, we propose to evaluate the oral epithelium of chronic e-cigarette smokers and those who have never smoked. This study involves a telephone questionnaire, a screeningvisit and a study visit.

Primary Investigator: Ronald G. Crystal, M.D.

Sponsor: National Institutes of Health (NIH), Weill Cornell Medicine

Study Type: Study Type

Phase I Randomized, Double-Blind, Placebo Control Study for an Anti-Cocaine Vaccine

This dose-ranging, placebo-controlled, double-blind study assesses the safety and preliminary efficacy of an anti-cocaine vaccine called dAd5GNE, designed to prevent cocaine from reaching the brain. The study will take place over a period of 32 weeks from the time of vaccine administration. The subject will be randomly assigned to receive either the active dAd5GNE anti-cocaine study drug or placebo, and will receive a total of six doses. The first dose will be administered at week zero, and five boosters will be administered at four-week intervals, during weeks four, eight, 12, 16 and 20. Subjects will receive $25 per visit (up to $75 per week) for ~32 weeks, in order to compensate for travel (this is a total of ~$2,400 per subject over the course of the entire study).

Primary Investigator: Ronald G. Crystal, M.D.

Sponsor: National Institutes of Health (NIH)

Study Type: Study Type

Collection of Airway, Blood and/or Urine Specimens from Subjects for Research Studies

This clinical trial is a protocol to obtain blood, urine and/or airway specimens, from individuals with or without lung disorders, in order to carry out laboratory studies concerning genetic expression, gene transfer, infection, proteins and human genes, and to store specimens for future genetic studies. Specimens collected may include blood, urine and airway samples (nasal, airway brushing, biopsy, lavage and/or washings) from consenting subjects.

Primary Investigator: Ronald G. Crystal, M.D.

Sponsor: National Institutes of Health (NIH), Cystic Fibrosis Foundation, Boehringer Ingelheim and Weill Cornell Medicine

Study Type: Study Type

Read more from the original source:
Clinical Trials | Genetic Medicine

‘Science is a contact sport’: What one U of T researcher learned in the lab of Nobel laureate William Kaelin – News@UofT

When University of Toronto Professor Michael Ohh was scouting postdoctoral positions at Harvard Medical School in the mid-1990s, a group of Canadians that often gathered to talk science told him to check out the molecular oncology floor at the Dana-Farber Cancer Institute, run by legendary virologist David Livingston.

It turned out to be good advice.

Ohh met with three young investigators Livingston had trained and hired, including the clinician-scientist William Kaelin Jr. The floor was full of bright and driven people, and the electricity in the air was palpable, recalls Ohh, now a professor of laboratory medicine and pathobiologyand of biochemistry at U of T.

Ohh spent the next five years in Kaelins lab and co-authored several papers on how cells respond to changes in oxygen levels research that contributed to Kaelin being awarded the 2019 Nobel Prize in Physiology or Medicine.

The work also set the stage for Ohhs career as an independent scientist in Toronto, and it continues to bear fruit and hold promise for treatments of cancer and other diseases.

One of the most significant things I learned in Bills lab was a rigorous approach to science, says Ohh. He really wanted you to nail the mechanism with a series of elegant experiments and he understood that process is highly competitive with other labs. He often said, Science is a contact sport.

Ohh calls his time in Kaelins lab a tour of duty unbearably stressful at times, but balanced by the excitement of doing fascinating science with talented people.

Kaelins lab was focused on three tumour suppressor genes. Two were well known, but the third was a newly discovered gene called VHL that had been shown to cause von Hippel-Lindau syndrome, a rare hereditary disorder in which patients develop multiple benign and cancerous tumours.

Only a handful of labs in the world studied VHL at the time and virtually nothing was known about its function. We tried a guilt-by-association approach, looking for proteins with which VHL interacted in the hope that these might shed clues to VHL function, says Ohh.

By the late 1990s, they had found several and determined that those proteins are part of a larger complex that likely targets other proteins for destruction.

They showed that without a functional VHL protein, these complexes are unable to degrade another protein called hypoxia-inducible factor (HIF), which in turn spurs tumour cell survival and growth even in low-oxygen microenvironments.

The mechanism at play in von Hippel-Lindau syndrome, in other words, also impacts how all cells sense and adapt to oxygen deprivation. The worlds of hypoxia and VHL collided in that discovery, says Ohh.

Researchers have since developed ways to control how cells sense and adapt to changes in oxygen, and are applying those findings for various conditions. A drug based on this approach is in clinical trial for various cancer types, and the findings also look promising in pre-clinical research for cardiovascular disease and anemia.

Ohhs lab in Toronto has just figured out why certain people with mutations in another HIF gene get cancer and/or polycythemia (an excess of red blood cells). Recent clinical reports have detailed the plight of people with this mutation, and clinicians are now asking Ohhs lab if they can help predict what disease their patients will eventually develop, so they can better monitor their patients for these conditions.

Early detection is critical for outcomes in cancer treatment, so this is useful, highly personalized medicine, says Ohh. And its an extension of the work we did 20 years ago, which is gratifying.

Ohh and his lab are also looking at evolutionary aspects of this molecular pathway. They are studying evolutionary diversions of the pathway among different animals over millions of years to see which genetic sequences and motifs are conserved.

Insights into these diversions could offer ways to understand the precise mechanisms and critical regions of HIF and VHL by which cells better respond to oxygen fluctuations.

Great science isnt the only product of the Kaelin lab. Ohh met his wife Meredith Irwin there when she was a research trainee, and they have been together for two decades. Irwin is a professor of pediatrics, medical biophysics and laboratory medicine and pathobiology at U of T, as well as a clinician-scientist at The Hospital for Sick Children.

Irwin was born and raised in New York, studied at the Massachusetts Institute of Technology and Harvard, and trained at Boston Childrens Hospital and Dana-Farber for more than a decade.

The couple travelled to Boston for Kaelins 60th birthday two years ago. They asked us to give talks, and afterward Bill made a comment about me stealing an American to Canada, says Ohh. We really are very happy here in Toronto.

Go here to read the rest:
'Science is a contact sport': What one U of T researcher learned in the lab of Nobel laureate William Kaelin - News@UofT

The 2007 Nobel Prize in Physiology or Medicine – Advanced …

Advanced Information [pdf]

The 2007 Nobel Prize in physiology or medicine is awarded to Drs Mario R. Capecchi, Martin J. Evans and Oliver Smithies for their discoveries of principles for introducing specific gene modifications in mice by the use of embryonic stem cells. Their work has made it possible to modify specific genes in the germline of mammals and to raise offspring that carry and express the modified gene. The toolbox of experimental genetic methods developed by Capecchi, Evans and Smithies, commonly called the knockout technology, has permitted scientists to determine the role of specific genes in development, physiology, and pathology. It has revolutionized life science and plays a key role in the development of medical therapy.

Martin Evans identified and isolated the embryonic stem cell of the early embryo, the cell from which all cells of the adult organism are derived. He established it in cell culture, modified it genetically, and reintroduced it into foster mothers in order to generate a genetically modified offspring. Mario Capecchi and Oliver Smithies, independently of each other, discovered how homologous recombination between segments of DNA molecules can be used to target genes in the mammalian genome and developed methods to generate genetically modified mice. Such animals have become indispensable in medical research. Furthermore, the knowledge concerning stem cell biology and gene technology obtained during the research that led to the "knockout mouse" has changed our understanding of normal development and disease processes and identified new avenues for medical therapy. Fig. 1 shows the general strategy for gene targeting in mice.

B) These ES cells are injected into blastocysts, which are injected into foster mothers to generate chimeric mice able to transmit the mutant gene to their progeny. To facilitate isolation of the desired progeny, the ES cells and recipient blastocysts are derived from mice with different coat colour alleles. In the figure, gene targeted ES cells and their progeny are shown in red and blastocysts in yellow.

A stem cell is a cell that is capable of extensive proliferation, creating more stem cells (self-renewal) as well as more differentiated cellular progeny. Somatic stem cells are necessary for renewal of the tissues of the adult organism. For instance, hematopoietic stem cells in the bone marrow differentiate into blood cells, i.e. erythrocytes, megakaryocytes/platelets, and the different types of leukocytes. While each somatic stem cell of the adult organism is committed to a certain line of differentiation, the early embryo contains stem cells that are totipotent, i.e. they give rise to all cell types in the developing organism. Therefore, the thought that embryonic stem cells from the blastocyst could be used to create to a living mammalian organism has fascinated scientists for many years.

The concept that differentiated cells and tissues are derived from undifferentiated stem cells ("Stammzellen") was already proposed a hundred years ago [1]. However, their precise properties remained elusive for many decades. Studies of testicular teratomas showed that these tumours contain totipotent cells. In the 1950s, Leroy Stevens at the Jackson Laboratory found that mice of the 129Sv strain have a high frequency of such tumours. He showed that their cells could develop into embryoid bodies, i.e. aggregates of embryonic cells. When transplanted, such aggregates could induce solid tumours with many different cell types [2, 3]. A few years later, Kleinsmith and Pierce demonstrated that such tumours were derived from undifferentiated embryonal carcinoma cells [4].

The development of cell culture techniques permitted investigators to establish cultures of embryonal carcinoma cells (EC cells) from murine testicular teratocarcinomas. Several scientists including Martin Evans at the University of Cambridge reported on such cultures in the early 70s [5-7].

Evans obtained 129Sv mice from Stevens, established a colony of mice, and characterized the teratoma derived cells in culture [8, 9]. These embryonal carcinoma (EC) cells could be grown on feeder layers of irradiated fibroblasts. When the latter were withdrawn, extensive in vitro differentiation occurred. It proceeded through a primitive embryonic endoderm, which clumped into embryoid bodies. Attachment on a solid surface gave rise to all kinds of cell types, including skin, nerve, beating cardiac muscle, etc. This showed that the EC cells differentiated in the same way as the inner cell mass of the mouse embryo [8, 9].

Evans saw the potential in using these EC cells not only for cell culture studies but also for creating chimeric mice. In order to realise this vision, he established a collaboration with Richard Gardner in Oxford, who made injections of EC cells into blastocysts and reimplanted them into foster mice. The offspring was chimeric, with contributions from EC cells in nearly every tissue [10]. Similar findings were made by several other groups at about the same time, [11] [12]. However, chimeric mice carrying EC derived cells developed multiple tumours and could not contribute to the germ line due to karyotypic abnormalities.

It became obvious to Evans that an alternative strategy had to be used if one were to obtain germline transmission derived from cultured embryonic stem cells. With the use of monoclonal antibodies, he characterised cell surface macromolecules of EC cells and their normal counterparts, thus identifying molecular markers of early differentiation [13]. The results suggested that normal cells with a similar phenotype as EC cells could be found and used for experiments. In 1980, Evans teamed up with the embryologist Matt Kaufman to combine cell culture and embryo manipulation. As described by Evans in a later review [14], he had intended to use haploid embryos for cell culture but prepared some diploid ones as controls. Evans writes [14]:

"When I cultured these blastocysts as explants in tissue culture, using a medium that had been honed for optimum cloning efficiency of both mouse and human EC cells, I immediately noted an outgrowth of EC-like cells. These cells were clearly recognizeable as the sought-after pluripotential cells, and they passed every test: They formed teratomas in vivo, and they differentiated in vitro. They bore the cell surface antigens that we expected. They stained strongly positive for alkaline phosphatase, were karyotypically normal and, most importantly, made splendid chimeras."

These cells were the embryonic stem cells (ES cells) that became critical for the success of gene targeting. Evans and Kaufman published their report on ES cells in a seminal paper in Nature in July, 1981 [15]. Gail Martin, a former co-worker of Evans, reported similar findings half a year later [16]. In their Nature paper, Evans and Kaufman pointed out the possibility of using ES cells for gene modification. They wrote [15]:

"Their [i.e. ES cells] use as a vehicle for the transfer into the mouse genome of mutant alleles, either selected in cell culture or inserted into the cells via transformation with specific DNA fragments, has been presented as an attractive proposition. In many of these studies the use of pluripotential cells directly isolated from the embryos under study should have great advantages."

Evans' team set up blastocyst injection techniques to test whether indeed ES cells could contribute to functional germ cells and thus be used to create a chimeric mouse. They reported successful germline transmission in 1984, in another landmark paper in Nature [17].

The next step was to determine whether ES cells could be used to introduce genetic material into the germline. Evans and his co-workers infected ES cells with a recombinant retrovirus before injecting them into blastocysts [18]. Retroviral DNA was identified in the founders and transmitted to the F1 offspring, demonstrating introduction of the foreign DNA into the mouse germline [19]. In October, 1986, Evans et al. reported their findings in Nature and concluded that "cultured embryonic cells provide an efficient means for the production of transgenic animals" [19]. In December of that year, another laboratory reported germline transmission of a neomycin resistance gene that they had introduced into ES cells by retroviral infection [20].

Evans now took the important step of introducing a mutant form of a specific, endogenous gene into the mouse genome. He and his co-workers transferred a mutant gene for hypoxanthine phosphoribosyltransferase (HPRT), which is defective in Lesch-Nyhan syndrome, an X-linked monogenic defect of purine metabolism [21]. Several copies of the mutated HPRT gene were introduced into the genome of the ES cells by retroviral infection in culture. Mutated ES cells were injected into blastocysts and contributed to chimeras. The mutations were transmitted germline and identified in the male offspring as loss of HPRT activity. In a paper published in Nature back-to-back with the one from Evans' lab, Hooper et al in Edinburgh reported germline transmission of another mutated HPRT gene, a spontaneous deletion mutation in ES cells [22]. For the first time, models of human disease had been created by genetic manipulation of ES cells.

In their paper [21], Evans and his co-workers point out that their success "opens up the possibility of deriving strains carrying specifically induced alterations in other genes" and suggest that "it may also eventually be possible to produce specific alterations in endogenous genes through homologous recombination with cloned copies modified in vitro", citing the work of Capecchi and Smithies [23, 24]. Indeed, the combination of the two technologies revolutionized experimental medicine, as we now know nearly 20 years later.

The mouse has been a favourite animal for genetic studies for many decades and was an obvious choice for the first attempts to introduce new genes into the mammalian genome. Work in several laboratories had defined conditions for manipulating fertilized mouse eggs and blastocysts in culture. Using these culture techniques, SV40 virus DNA was introduced into blastocysts, which were subsequently implanted into pseudopregnant foster mothers. SV40 DNA could be detected in the offspring but it was impossible to demonstrate with certainty whether the DNA was integrated into the host genome, or remained as episomes [25]. A few years later, the first transgenic mouse was created by infecting embryos with Moloney leukemia virus [26]. A DNA copy of the viral RNA was present in the genome of the transgenic mice and was transferred to the offspring in a Mendelian fashion, therefore virus DNA had been introduced into the mouse germline. Subsequent development has made it possible to introduce and overexpress a large number of transgenes in mice and also other mammals [27]. However, integration of the foreign DNA in the genome occurs at random and the number of copies varies. Although an important tool in life science, transgene technology of this kind lacks precision with regard to the inserted gene and cannot be used to manipulate endogenous genes in a predetermined manner. These inherent problems with the transgenic overexpression technique limit its usefulness.

The principle of recombination between homologous genes has been known for half a century and was recognized by a Nobel Prize to Joshua Lederberg in 1958 for his studies in bacteria. In the 70s, it became evident that eukaryotes employ a similar machinery to mediate exchange of genetic information between homologous chromosomes during meiosis. Early studies in yeast were followed by experiments demonstrating recombination between retroviral DNA sequences in the mammalian genome and introduced oligomeric retroviral DNA. Pioneering work by Richard Axel (2004 Nobel Prize for the discovery of odorant receptors) showed that cultured mammalian cells defective in thymidine kinase could be rescued by introduction of the herpes virus thymidine kinase (tk) gene [28]. Mario Capecchi decided to improve the method and used a fine glass pipette to inject DNA directly into the nucleus [29]. This improved the efficiency of gene transfer dramatically and Capecchi's method was rapidly adopted by other investigators to introduce new genes into fertilized mouse embryos and produce transgenic mice [30]. However, the transferred gene was still introduced at random in the host genome.

Capecchi now made a crucial observation: when the tk gene was injected, copies were integrated in only one or two loci of the host genome, with multiple copies forming head-to-tail concatemers. He reasoned that such concatemers could only be generated by two mechanisms: either by replication or by homologous recombination. A series of careful experiments were performed, which unequivocally demonstrated that head-to-tail concatemers were generated by homologous recombination [31]. This, in turn, provided evidence that mammalian somatic cells possess an efficient enzymatic machinery for mediating homologous recombination. If this machinery could be harnessed to accomplish homologous recombination between a newly introduced DNA molecule and the same DNA sequence in the recipient cell's genome, any cellular gene could be mutated.

Capecchi now submitted a grant proposal to the U.S. National Institutes of Health to test the feasilibity of gene targeting in mammalian cells. It was rejected since the reviewers considered it extremely unlikely that the introduced DNA would find its matching sequence within the host genome (cited by Capecchi in a later review [32])! At about the same time, Martin Evans et al in England proposed a similar strategy in a grant application to the UK Medical Research Council, which was also turned down for being over-ambitious!

Capecchi decided to continue working on homologous recombination in spite of being turned down by NIH. He generated recipient cell lines that carried a defective neomycin resistance gene (neor) and was able to repair it by introducing a functional neor gene [23]. Correction occurred at a relatively high frequency (in one cell per 1,000 injected cells), making it likely that homologous recombination could be used to manipulate genes of the mammalian genome.

In parallel with Capecchi's work, Oliver Smithies had developed the concept that homologous recombination might be used to repair mutated genes. As early as the 1960s he had already established that an allelic variant of haptoglobin had occurred through recombinatorial events [33]. Later on, he cloned human fetal globin genes and concluded that Gγ and Aγ had arisen through a process involving homologous recombination [34]. He devised a stepwise selection procedure for recovering targeted cells carrying modified genes. The strategy was successful and he reported in a landmark paper in the September 19, 1985 issue of Nature the successful integration by homologous recombination of a plasmid into the chromosomal β-globin gene of human erythroleukaemia cells [24].

By 1985, Capecchi had shown that homologous recombination occcurs with high frequency in mammalian cells and Smithies had used homologous recombination to insert a plasmid DNA sequence into a chromosomal gene of a human cell. However, all this work was carried out in cell culture. Could homologous recombination be used to target genes in the germline and obtain strains of genetically modified animals? Both Capecchi and Smithies had heard of Martin Evans' ES cells and decided to give them a try. With the help of Evans, they both set up ES cell culture for use in homologous recombination experiments.

Smithies first used homologous recombination to correct a mutant HPRT gene in cultured ES cells [35]. For this purpose, an ES cell line was used that carried a deletion mutation; this cell line had previously been used for production of mutant mice. The HPRT gene was repaired with a plasmid carrying the missing promoter and first 2 exons and Smithies showed that treated cells survived and grew in HAT selection medium, which requires HPRT enzyme activity. Smithies and his co-authors concluded that "This modification of a chosen gene in pluripotent ES cells demonstrates the feasibility of this route to manipulate mammalian genomes in predetermined ways" [35].

Capecchi's team also chose the HPRT gene for their early studies. Standard methods were available for selectively growing cells with functional HPRT enzymes and had already been used for several years for selection of mutants, hybridoma cells in monoclonal antibody production etc. Thomas and Capecchi [36] introduced a neomycin resistance gene into an exon of the HPRT gene in ES cells and showed that clones of transfected cells had lost HPRT but gained neoR activity. They concluded in their Cell paper that "It is hoped that this combination of using ES cells as the recipient cell line and site-specific mutagenesis achieved by gene targeting will provide the means for generating mice of any desired genotype." [36] They continued by outlining an experimental strategy:

"An advantage of this scenario is that the first generation chimera will usually be heterozygous for the targeted mutation and that subsequent breeding can be used to generate the homozygous animal. Thus, only one of the two loci need be inactivated, and recessive lethals can be maintained as heterozygotes. If successful, this technology will be used in the future to dissect the developmental pathway of the mouse as well as to generate mouse models for human disease." [36]

This vision has become reality and is now a cornerstone of experimental medicine.

It was important to proceed from the "model gene" HPRT to a general strategy that would allow targeting of genes whose function cannot be selected for in cell culture. Thomas and Capecchi [36] had pointed out that the frequency of homologous recombination vs random integration was 1/1,000, which should be high enough to permit targeting of non-selectable genes as well. This observation prompted work to develop the methods needed for such approaches. The following year, Capecchi's positive-negative selection strategy for enriching ES cells containing a targeted disruption of any transfected gene was presented in Nature [37] (Fig 2). A neomycin resistance element (neoR) is introduced into an exon of the replacement vector, which also has a thymidine kinase (HSV-tk) element at its end. Homologous recombination of the targeted gene will result in neoR expression but the tk element will be lost since it was outside of the recombining DNA sequences. In contrast, random integration of the replacement vector will introduce tk as well as neoR into the gene. This strategy was successfully used to disrupt the int-2 gene, which is a member of the fibroblast growth factor (FGF) family [37].

All the components were now in place for producing gene-targeted mouse strains: the development of ES cell culture, the demonstration that gene modification in such cells can be transmitted to the germline and registered in the offspring, the observation that homologous recombination occurs with high frequency in the mammalian genome, the application of gene transfer methods to ES cells, and the invention of strategies for enriching transfected cells. Several laboratories joined the race and 1989 saw the birth of several different knockout mice [38-41]. Fig 3 shows the molecular evidence for correction of the mutant HPRT gene in the germline by Smithies and co-workers [39].

After the establishment of gene targeting technology, several important modifications and developments, in several laboratories, have extended its use in significant ways. An ingenious development of gene targeting has been made by introducing recognition sites for the enzyme Cre recombinase, so-called loxP sites, into existing genes. When mice carrying such "floxed" genes are mated with transgenic mice expressing Cre recombinase, the target gene of the offspring is modified through Cre action [42-44]. Another site-specific recombinase, Flp, is also frequently used to construct conditional targeting of genes in mice [45]. The activity of the Cre, or Flp, gene can be controlled by placing it under a suitable promoter to achieve tissue-specific gene targeting [46]. Expression of Cre and hence targeting of the floxed gene can be restricted to e.g. T cells (lck promoter), cardiac muscle (cardiac myosin promoter), neurons (enolase promoter) or epithelia (cytokeratin promoter).

Cre expression can also be controlled temporally, by introducing an element into the promoter which requires a ligand such as a drug for induction [47]. Tetracyclin, type I-interferon and tamoxifen (which binds to an estrogen receptor-binding element) have all been used to obtain drug-inducible promoters. In this way, a desired gene can be targeted by administrating the drug. By introducing a tamoxifen site into a tissue-specific promoter, gene targeting can be obtained selectively in a certain tissue when the mouse is treated with the drug.

Cre-lox technology can also be used to replace an existing gene with another one [48]. Such "knock-in" has been used e.g. to replace murine immunoglobulin or MHC genes with human ones in order to "humanize" the mouse with regard to immune function. It has also been used to replace an allele with another one, the latter for instance being an allele suspected to cause disease.

Gene targeting has transformed scientific medicine by permitting experimental testing of hypotheses regarding the function of specific genes. Prior to gene targeting, our understanding of the role of genes in higher organisms was deduced from observations of spontaneous mutations in patients and experimental animals, linkage and association studies, administration of gene products to animals and, to some extent, from cell culture experiments. However, cell culture is not helpful for understanding functions and diseases involving multicellular, integrative responses. Insights into organ systems such as the nervous system,the cardiovascular system, and the immune system, were fragmentary at best, as was knowledge of mammalian development. As the cardiovascular physiologist Heimo Ehmke put it, "cells don't have blood pressure" [49]. The possibility of observing the effects on the intact organism of destroying a candidate gene transformed these areas of research. For instance, cardiovascular physiologists switched from rats to mice as models, downscaling their instruments and techniques in order to study the genetic regulation of hemodynamics. A new era of genetic physiology was born.

The genomes of man and mouse contain about 22,400 genes. Several thousand of them have already been investigated by gene targeting. Collectively, these studies have provided a wealth of information about gene function in development and disease. They have helped fuse mechanistic molecular biology with integrative life sciences such as embryology, physiology and immunology and have prompted new technical developments in physiological sciences. For medicine, the modeling of human diseases by gene targeting in mice has been particularly informative.

At this stage, it may be helpful to recapitulate the criteria first proposed by Claude Bernard for the scientific method in medicine [50]: Medical scientists use observations, hypotheses and deductions to propose explanations, theories, for natural phenomena. Predictions from these theories are tested by experiment. Any theory which is cogent enough to make predictions can be tested reproducibly in this way. Therefore, the scientific method is essentially a cautious means of building a supportable, evidence-based understandingof our natural world. Experiments are crucial in this process.

Prior to gene targeting, genetic medicine lacked the means for experimental testing. If we make an analogy with Robert Koch's approach to infectious diseases [51], genetic medicine could apply the first of Koch's postulates (i.e. observe an association between microbe, or in this case, gene or allele, and disease) and with the advent of gene cloning, the second one (isolate the microbe/gene from the diseased individual and establish it in culture), but applying Koch's third postulate (induce the disease by transferring the microbe/gene to a host organism) required gene targeting. By mutating a gene to destroy its function (knock-out) or switching it to a disease-associated allele (knock-in), disease is induced if the hypothesis is correct. Alternative approaches based on genetic epidemiology are currently being developed but currently available methods do not have the precision of hypothesis-based experiments. This digression into scientific theory may suffice to make the point that only by targeting candidate genes did it become possible to formally establish causality between gene and disease. Let us now look at some specific examples of the impact of gene targeting in medicine.

The first area to which experimental geneticists turned their attention after the birth of gene targeting in mammals was monogenic diseases. The Lesch-Nyhan syndrome, a defective nucleotide metabolism caused by a mutation in the HPRT gene, actually served as the model condition during development of the technology, both in Evans' and Smithies' laboratories (see above). One of the reasons for chosing this particular medical condition was because selection conditions for isolating transduced cells were available for HPRT. The first examination of HPRT-/- mice was disappointing since neither neuropathological nor behavioural features of human disease could be observed [21, 39]. This prompted analysis of purine salvage pathways in mice and led to the findings that mice depend largely on adenine phosphoribosyltransferase (APRT) for purine salvage and are therefore not as sensitive to HPRT deficiency as humans. Administration of an APRT inhibitor to HPRT-/- mice induced persistent self-injurious behaviour resembling the clinical features in human disease [52]. This is an illustration of the need for sophisticated analysis of integrative functions when characterising the phenotype of gene-targeted mice.

Cystic fibrosis is one of the most common monogenic disease and was chosen for gene-targeting studies by Smithies and his co-workers [53, 54]. The defective gene had been identified by linkage studies in patient families followed by molecular cloning. It turned out to be a cAMP-activated choride channel and was termed cystic fibrosis transmembrane conductance regulator (CFTR). By knocking out CFTR in mice, a condition was generated that reproduced many features of the human disease. Thus, CFTR-/- homozygotes displayed defective chloride transport in epithelia of airways and intestines, failure to thrive, meconium ileus, and pathological alterations of gastrointestinal glands. These studies were among the first to create a model of a human disease by gene targeting in mice. They have been followed by an avalanche of such knock-out models.

The pathogenesis of inherited heart diseaseshave been explored successfully by gene targeting approaches [55, 56]. For instance, targeting of genes encoding components of the contractile apparatus in cardiomyocytes leads to cardiomyopathy; targeted mutations in connexin proteins of gap junctions cause conduction defects; disrupted genes for transcription factors involved in heart development lead to congenital heart malformations; and targeting of genes controlling energy metabolism causes cardiomyopathy.

Complex diseases involving the action of more than one gene, and in addition, gene-environment interactions, represent a particular challenge for medical research. Inheritance, penetration and interactions are usually poorly understood, it has been difficult to dissect the contribution of an individual genetic factor, and the distinction between causation and correlation has been problematic. In order to prove causation in such a complex system, experiments must permit detection of the effects of changing only a single variable at one time. Gene targeting made such experiments possible and has permitted proof of causation in complex diseases.

Oliver Smithies has been the leader in this development. Together with Nobuyo Maeda, he focused on two important, complex diseases, hypertension and atherosclerosis (reviewed in [57]). Twin studies suggest that genetic factors may account for approx 70% of familial aggregation of essential hypertension. However, at least 10 genes have been shown to alter blood pressure and their gene products appear to interact in complex ways. In spite of the discovery that angiotensinogen (AGT) gene polymorphism is associated with essential hypertension, the genetics of this disease has remained poorly understood [58]. Little is known about the number of genes actually involved in human essential hypertension, their quantitative effect on blood pressure, their mode of transmission, or their interaction with other genes and environmental components.

Smithies suspected that gene dose effects would impact on blood pressure levels and designed a new method for titrating gene dosage by producing mice with one, two or three functional copies of the AGT gene [59]. "Conventional" gene targeting was used to produce the one- and two-copy mice and gap-repair gene targeting to produce mice with three copies of the AGT gene. This resulted in proportionally higher levels of gene products (i.e. plasma angiotensinogen protein) and, importantly, proportionally higher blood pressure with increasing gene copy number. When Smithies et al targeted another important gene for blood pressure regulation, the one coding for the angiotensin-converting enzyme (ACE), no such linear relationship was observed, in spite of the effectiveness of ACE inhibitors in reducing blood pressure. The investigators submitted their data to a computer simulation for complex interacting systems and could propose a model for blood pressure control through the renin-angiotensin system, which has proven to be useful for understanding essential hypertension [60]. It shows that gene dosage, gene expression, and gene product clearance/catabolism must all be considered when evaluating the genetic regulation of blood pressure.

In 1992, Nobuyo Maeda, working in Smithies' department at the University of North Carolina, developed a mouse model of atherosclerosis by targeting the gene for apolipoprotein E (Apoe) [61]. The same gene was targeted independently by investigators at Rockefeller University [62]. The Apoe-/- mouse develops spontaneous atherosclerosis which is remarkably similar to human disease. The following year, Michael Brown and Joseph Goldstein (1985 Nobel Prize for discoveries concerning cholesterol metabolism) and their co-workers targeted the gene for the low density lipoprotein (LDL) receptor (Ldlr) and obtained a mouse that develops atherosclerosis when fed a cholesterol-rich diet [63]. The introduction of the two mouse models with defective Apoe and Ldlr genes have completely changed atherosclerosis research. By crossbreeding them with other gene-targeted mice, it has been possible to deduce the importance of genes regulating inflammation, lipid metabolism, blood pressure and other factors proposed to be involved in atherosclerotic cardiovascular disease [64]. They are also used abundantly in the pharmaceutical industry for development and testing of new drugs against coronary artery disease.

Gene targeting has been exceptionally useful in cancer research. A large number of protooncogenes, tumor suppressor genes, angiogenetic factors etc have been targeted in different tissues in mice to shed light on the induction and spreading of tumours [65]. Gene targeting of tumour suppressor genes have helped clarify their role in the formation of tumours. For instance, mice carrying a targeted p53 gene were predisposed to tumour development [66]. Conditional targeting (using Cre-lox technology) of the adenomatous polyposis coli (APC) gene induces colorectal tumors in mice and APC-targeted mice have become useful models for research on solid tumours [67]. Targeting of genes for endothelial growth factors and proteolytic enzymes have been essential for understanding mechanisms of neoangiogenesis and metastasis of solid tumours and are also used for developing therapeutic strategies to prevent spreading [68].

Contemporary research into most if not all major human diseases involves gene targeting in mice and there are "knockout models" for endocrine, metabolic, neurological, inflammatory and other disorders. Gene-targeted mouse models have also become increasingly important in studies of host defense against pathogens. Indeed, gene targeted mice have become indispensable in virtually all aspects of medical research.

Gene targeting has transformed physiology and medicine. Among the basic biomedical sciences, it is difficult to imagine contemporary medical research without the use of gene targeted models. The ability to generate predictable designer mutations in mouse genes has led to penetrating new insights into development, immunology, neurobiology, physiology, and metabolism. It has also allowed disease models of human pathologies to be generated in a tractable mammalian system and consequently enabled experimental dissection of disease states, identification of new therapy targets and the development of test systems for pharmacology. Finally, it is obvious that the development, in the future, of novel therapies to correct genetic defects in man will build on the experience of gene modification in mice that is based on the discoveries made by Mario Capecchi, Martin Evans and Oliver Smithies.

Gran K Hansson Professor of cardiovascular research at Karolinska Institutet Member of the Nobel Committee for Physiology or Medicine

To cite this page MLA style: "The 2007 Nobel Prize in Physiology or Medicine - Advanced Information". Nobelprize.org. Nobel Media AB 2014. Web. 3 Jun 2017. <http://www.nobelprize.org/nobel_prizes/medicine/laureates/2007/advanced.html>

Read this article:
The 2007 Nobel Prize in Physiology or Medicine - Advanced ...

Carleton College to hold its 143rd Commencement Ceremony June 10 – Carleton College News

Carleton College will award the Bachelor of Arts degree to the 505 graduating members of the Class of 2017 onSaturday,June 10, in a ceremony beginning at 9:30 a.m. on the lawn west of Hulings Hall on the Carleton campus. A celebratory picnic on the Bald Spot will follow. In the event of severe weather, commencement will be held indoors at the Recreation Center. Seating is available to accommodate all guests, whether outdoors or indoors, and no tickets are required. The ceremony will also be broadcasted live online (https://apps.carleton.edu/events/commencement/livestream/).

Following President Steve Poskanzers opening remarks,Reina Desrouleaux '17, chemistry major from Silver Spring,Maryland (whose speech is titled [insert meaningful life experience here]) and Eli Ruffer '17, chemistry major from Highland Park, Illinois (whose speech is titled Tyler, the Prospective Student)will address the Class of 2017, families and friends, and faculty. In additionally, Carleton College will confer an honorary doctorate upon Kathy L. Hudson 82, former Deputy Director for Science, Outreach, and Policy at the National Institutes of Health, who will briefly address the class.

The highest honor given by the College, conferred honoris causafor the sake of honorthis years honorary degree recipient is Dr. Kathy L. Hudson, former Deputy Director for Science, Outreach, and Policy at the National Institutes of Health (NIH).

Throughout her distinguished career, Hudson has served the public by ensuring that advances in genomics and other rapidly moving areas of medical research are paired with wise and effective public policies.

After earning a B.A. in biology from Carleton College and a M.S. in microbiology from the University of Chicago, Hudson obtained her Ph.D. in molecular biology from the University of California, Berkeley. Although she trained for a career in research, Hudson discovered that her real passion was science policy. As an American Association for the Advancement of Science (AAAS) Fellow in Washington DC, she worked for the U.S. House of Representatives and then the Congressional Office of Technology Assessment.

After a stint in the office of the Assistant Secretary for Health at the Department of Health and Human Services, Hudson joined the National Human Genome Research Institute (NHGRI) as assistant director. While there she made a compelling case to scientists, public policy experts, and lawmakers about the need for federal legislation to guard against genetic discrimination. She also helped to broker an historic agreement between the public and private human genome projects, which was announced by President Bill Clinton in the White House in 2000.

In 2002, Hudson left NHGRI to found and direct the Genetics and Public Policy Center at Johns Hopkins University. She became a leader in educating and advising about science and policy issues in genetics. Also at Hopkins, Hudson was an Associate Professor in the Institute of Bioethics and the Institute of Genetic Medicine. It was Hudson who did much of the work to assemble the talented and dedicated team that, in 2008 after years of effort, achieved passage of the landmark Genetic Information Nondiscrimination Act.

In 2009, Hudson returned to the National Institutes of Health, becoming the Deputy Director for Science, Outreach, and Policy. In that capacity helped found and launch the National Center for Advancing Translational Sciences. She also had a major hand in the design and launch of three national scientific projects the BRAIN Initiative, the Precision Medicine Initiative, and the Cancer Moonshot. In addition, she led efforts to revise the rules that govern participation of human subjects in research, modernize clinical trial reporting, expand scientific data sharing, and develop appropriate oversight for rapidly moving areas of medical research, including stem cells and gene editing.

On top of her many duties and responsibilities, Hudson made time to serve as a strong and tireless advocate for the role of women in science. She personally mentored a group of young women who are now moving into key leadership roles with a wide range of innovative biomedical research and policy initiatives.

Earlier this year Hudson left government service, and is working as an advisor to companies and research institutes as they forge new directions at the forefront of biomedical research.

For further information, including disability accommodations, contact the Carleton College Office of College Communications at(507) 222-4309or emailkraadt@carleton.edu. The commencement site is located on the Carleton campus between College and Winona Streets in Northfield.

More:
Carleton College to hold its 143rd Commencement Ceremony June 10 - Carleton College News

New research, plus basic care, could help cats – News-Press Now

At the University of Missouri's College of Veterinary Medicine in Columbia, genome research may help improve the health of cats.

"Continued collaboration with geneticists and veterinarians could lead to the rapid discovery of undiagnosed genetic conditions in cats," said Dr. Leslie Lyons, a professor of Comparative Medicine in the College of Veterinary Medicine.

The university is studying rare feline genetic disorders. The college is involved in developing complete feline DNA sequencing to identify anomalies causing disease. This should allow clinicians to develop an effective course of treatment to improve a cat's health.

"The goal of genetic testing is to identify disease early," Lyons said, "so that effective and proactive treatment can be administered."

This research at the university has identified a gene associated with progressive retinal blindness in the African black-footed cat and a gene in the silver tabby producing a fatal disorder. These gene identifications may help to diagnose other cats and allow them to receive appropriate treatment, Lyons said.

But in the meantime, local veterinarians say feline health starts in the home.

If you want your cat to be safe, make it a house cat, according to veterinarians. Then do simple things to ensure your feline isn't becoming sick, including vaccinations for rabies, leukemia and feline respiratory diseases.

Simple things such as brushing your cat daily will decrease the number of hair balls. Experts also suggest regular litter box changes, regular feeding times and portion control to keep a cat healthy.

Avoid feeding table scraps even when a feline begs for bones, fresh eggs, dough and dairy products like milk or yogurt, said Dr. Mark Gutzmer, veterinarian and an owner of All Creatures Animal Hospital. These things may harm a cat's digestive health, he said. And cats have a tendency to become overweight, so they shouldn't have any scraps, the doctor said.

Owners should observe their feline pets and specifically check out their fur coat, skin, eyes, mouth and teeth, if they will let you, Gutzmer said.

A cat's pelt should have shinny hair with the skin being free of flakes. Eyes should be bright and clear without mucus or other substances around the edges of the eye. Cats should have bright pink gums and when looking at their back teeth.

Two other factors for observing cat health would be activity and digestion.

"A healthy young cat should be playful and active throughout a good portion of the day," said Kyle Ripperger, a veterinarian with East Hills Veterinary Clinic.

House cats, once they're comfortable with their surroundings, tend to develop a routine and owners should be familiar with it, Ripperger said.

"They kinda do the same things all the time," Ripperger said.

Any change in that routine could be significant, both doctors said.

Always let your veterinarian know of changes in a cat's behavior, such as a lack of appetite, vomiting, diarrhea, unexplained weight loss, sitting for long times in a litter box and the appearance of what seems to be depression.

Weight loss is a clue something is going on, both doctors said.

Even drinking excess water can be a sign something isn't right, Ripperger said.

And both doctors say to always bring your pet in for a yearly checkup. This may allow the doctors to get ahead of any symptoms you haven't noticed.

See the rest here:
New research, plus basic care, could help cats - News-Press Now

Blood test offers hope for better lung cancer treatment – Sioux City Journal

BOSTON (AP) Researchers have taken an important step toward better lung cancer treatment by using blood tests to track genetic changes in tumors as they progress from their very earliest stages.

With experimental tests that detect bits of DNA that tumors shed into the blood, they were able to detect some recurrences of cancer up to a year before imaging scans could, giving a chance to try new therapy sooner.

It's the latest development for tests called liquid biopsies, which analyze cancer using blood rather than tissue samples. Some doctors use these tests now to guide care for patients with advanced cancers, mostly in research settings. The new work is the first time tests like this have been used to monitor the evolution of lung tumors at an early stage, when there's a much better chance of cure.

Only about one third of lung cancer cases in the United States are found at an early stage, and even fewer in other parts of the world. But more may be in the future as a result of screening of longtime smokers at high risk of the disease that started a few years ago in the U.S.

Early-stage cases are usually treated with surgery. Many patients get chemotherapy after that, but it helps relatively few of them.

"We have to treat 20 patients to cure one. That's a lot of side effects to cure one patient," said Dr. Charles Swanton of the Francis Crick Institute in London.

The new studies he led suggest that liquid biopsies might help show who would or would not benefit from chemotherapy, and give an early warning if it's not working so something else can be tried.

Cancer Research UK, a charity based in England, paid for the work, and results were published online Wednesday by Nature and the New England Journal of Medicine .

To be clear: This kind of care is not available yet the tests used in these studies are experimental and were customized in a lab to analyze the genes in each patient's cancer. But the technology is advancing rapidly.

The company that generated the tests for the study in Nature California-based Natera Inc. plans to offer the tests for research by universities and drug companies later this year and hopes to have a version for routine use in cancer care next year.

"This is coming, and it's coming fast," said Dr. David Gandara, a lung specialist at the University of California, Davis, who had no role in the studies but consults for two companies developing liquid biopsies. A test that could spare many people unnecessary treatment "would be huge," he said.

In the studies, researchers analyzed tumors from about 100 people with non-small cell lung cancer, the most common form of the disease. Even in these early-stage cases, they found big variations in the number of gene flaws, and were able to trace how the tumors' genes changed over time.

People with many gene or chromosome problems were four to five times more likely to have their cancer return, or to die from their disease within roughly two years.

They also looked at 14 patients whose cancers recurred after surgery, and compared them to 10 others whose did not. Blood tests after surgery accurately identified more than 90 percent of them that were destined to relapse, up to a year before imaging tests showed that had occurred.

The results suggest that using liquid biopsy tests to help select and adjust treatments is "now feasible," at least from a scientific standpoint, the authors write.

A big issue is cost, though. Liquid biopsies sold now in the U.S. cost nearly $6,000. Tests that more narrowly track a patient's particular tumor gene changes, like the one in these studies, may cost less. They may save money in the long run, by preventing futile treatment, but this has yet to be shown.

Go here to read the rest:
Blood test offers hope for better lung cancer treatment - Sioux City Journal

No Excuses: Exercise Can Overcome the ‘Obesity Gene’ – Glens Falls Post-Star

THURSDAY, April 27, 2017 (HealthDay News) -- Even if obesity is "in your genes," regular exercise can help keep extra pounds at bay, a new study suggests.

Researchers found that when people carried a particular gene variant that raises obesity risk, regular exercise seemed to reduce the effects of their DNA -- by about one-third.

The gene in question is known as FTO. Studies show that people with a particular variant of the gene have a heightened risk of obesity.

But the gene's effects are not huge, or written in stone. Research has found that people who carry two copies of the FTO variant (one inherited from each parent) weigh about 6.5 pounds more than non-carriers, on average.

The new findings underscore one way to counter the gene's impact: Exercise.

"There are genes that appear to directly impact weight, but the effects are small," said lead researcher Mariaelisa Graff, of the University of North Carolina at Chapel Hill. "You still have a lot of choice over your behavior."

The study results are not exactly surprising, according to Dr. Timothy Church, an obesity researcher who was not involved in the work.

"This shows, once again, that genes are not your destiny," said Church. He is a professor of preventative medicine at Louisiana State University's Pennington Biomedical Research Center.

Church said regular exercise is particularly key in preventing excess weight gain in the first place -- and in keeping the pounds off after someone loses weight.

Exercise is less effective in helping obese people shed weight, Church said. Diet changes are the critical step there.

But the bottom line is that exercise matters, regardless of your genes, according to Dr. Chip Lavie, of the John Ochsner Heart and Vascular Institute, in New Orleans.

Lavie, who was not involved in the study, pointed to findings from his own research.

"[We] have published data that suggests the main cause of increasing obesity over the past five decades is the dramatic decline in physical activity," he said.

Gym memberships aside, Americans these days are less active at work, at home (through housework) and during leisure time, according to Lavie.

And the benefits of exercise go beyond weight control, he stressed. Physical activity boosts people's fitness levels -- which, Lavie said, is critical in preventing heart disease and living a longer, healthier life.

The new findings are based on over 200,000 adults, mostly of European descent, who'd taken part in previous health studies.

Graff and her colleagues analyzed information on their weight and exercise habits, and looked at how those factors "interacted" with 2.5 million gene variants.

FTO is the gene that is most strongly linked to obesity, Graff said.

And overall, her team found, active people who carried the obesity-linked FTO variant appeared more resistant to its effects than sedentary people.

On average, exercise weakened the variant's effects by about 30 percent, the researchers reported in the April 27 issue of PLOS Genetics.

There were some hints that exercise also affected some other weight-related genes. But the only clear relationship was with the FTO variant, according to Graff.

That, she noted, could be related to the broad way the study looked at exercise. The 23 percent of people who were least active were considered "inactive," while everyone else was deemed "active."

Church said he thinks research into the genetics of body weight will increasingly become useful.

If certain gene variants affect people's response to a low-carb diet or aerobic exercise, for example, that could help in "tailoring" weight-loss plans, he suggested.

"The science is rapidly evolving," Church said, "and there's still a lot to learn. But I think that's the direction this is going."

Link:
No Excuses: Exercise Can Overcome the 'Obesity Gene' - Glens Falls Post-Star

‘Sleep gene’ offers clues about why we need our zzzs – WSU News

By Eric Sorensen, WSU science writer

SPOKANE, Wash. Washington State University researchers have seen how a particular gene is involved in the quality of sleep experienced by three different animals, including humans. The gene and its function open a new avenue for scientists exploring how sleep works and why animals need it so badly.

Sleep must be serving some important function, but as scientists we still dont understand what that is, said Jason Gerstner, assistant research professor in WSUs Elson S. Floyd College of Medicine and lead author of a paper in the open-access journal Science Advances. One way to get closer to that is by understanding how it is regulated or what processes exist that are shared across species.

As a doctoral student at the University of Wisconsin, Gerstner looked at genes that change expression over the sleep-wake cycle and found expression of the gene FABP7 changed over the day throughout the brain of mice.

He and colleagues saw that mice with a knocked out FABP7 gene slept more fitfully compared to mice with the gene intact. This suggested the gene is required for normal sleep in mammals.

To see if FABP7 is indeed required for normal sleep in humans, Gerstner and colleagues in Japan looked at data from nearly 300 Japanese men who underwent a seven-day sleep study that included an analysis of their DNA. It turned out that 29 of them had a variant of the gene responsible for the production of FABP7.

Like the mice, they tended to sleep more fitfully. While they would get the same amount of sleep as other people, their sleep was not as good, with more waking events when they should be sleeping.

Finally, the researchers made transgenic fruit flies. They inserted mutated and normal human FABP7 genes into star-shaped glial cells called astrocytes. Glial cells were long thought to be mere supporting characters to neurons, the processors of information in the brain. But researchers more recently have found that, like neurons, glial cells release chemical neurotransmitters and control behavior.

To monitor the flies sleep, the researchers used a commercial Drosophila activity monitor that automatically records activity changes using an infrared beam to determine if a fly is awake or asleep. If the beam is unbroken for five or more minutes, the machine concludes the fly is asleep.

It turned out that flies with the mutated FABP7 gene broke the beam more frequently during the normal sleep time. Like mice and humans without a properly functioning FABP7 gene, mutant FABP7 flies slept more fitfully.

This suggests that theres some underlying mechanism in astrocytes throughout all these species that regulates consolidated sleep, said Gerstner.

Moreover, he said, Its the first time weve really gained insight into a particular cells and molecular pathways roles in complex behavior across such diverse species.

Even more remarkable is that fruit flies have been on the planet for some 60 million years.

That suggests we have found an ancient mechanism that persisted over evolutionary time, he said. Evolution does not keep something around that long if it is not important.

While the researchers are excited about finding a gene with an apparently strong influence on sleep, they stress that other genes are almost certainly involved in the process.

FABP7 proteins are involved in what is called lipid signaling, shuttling fats to a cell nucleus to activate genes controlling growth and metabolism. Gerstner and his colleagues will now look to see how these functions might intersect with theories about why sleep matters. Among those theories are that sleep is important for neuronal activity, energy use and storage, and memory and learning.

Gerstners collaborators include scientists in Japan, Wisconsin and Pennsylvania, as well as WSU research intern Samantha Riedy, WSU professor Marcos Frank, and Hans Van Dongen, director of the WSU Sleep and Performance Research Center.

Funders include the National Institutes of Health and the U.S. Office of Naval Research.

The research is in keeping with WSUs Grand Challenges, major initiatives aimed at large societal problems. It is particularly relevant to the Sustaining Health challenge.

News media contact: Jason Gerstner, WSU Elson S. Floyd College of Medicine, 509-368-6660, j.gerstner@wsu.edu

Excerpt from:
'Sleep gene' offers clues about why we need our zzzs - WSU News

Study reverses thinking on genetic links to stress, depression – Washington University School of Medicine in St. Louis

Visit the News Hub

Scientists re-examine data exploring connection between serotonin gene, depression, stress

For years, scientists have been trying to determine what effect a gene linked to the brain chemical serotonin may have on depression in people exposed to stress. But now, analyzing information from more than 40,000 people who have been studied over more than a decade, researchers led by a team at Washington University School of Medicine in St. Louis have found no evidence that the gene alters the impact stress has on depression.

New research findings often garner great attention. But when other scientists follow up and fail to replicate the findings? Not so much.

In fact, a recent study published in PLOS One indicates that only about half of scientific discoveries will be replicated and stand the test of time. So perhaps it shouldnt come as a surprise that new research led by Washington University School of Medicine in St. Louis shows that an influential 2003 study about the interaction of genes, environment and depression may have missed the mark.

Since its publication in Science, that original paper has been cited by other researchers more than 4,000 times, and some 100 other studies have been published about links between a serotonin-related gene, stressful life events and depression risk. It indicated that people with a particular variant of the serotonin transporter gene were not as well-equipped to deal with stressful life events and, when encountering significant stress, were more likely to develop depression.

Such conclusions were widely accepted, mainly because antidepressant drugs called selective serotonin reuptake inhibitors (SSRIs) help relieve depression for a significant percentage of clinically depressed individuals, so many researchers thought it logical that differences in a gene affecting serotonin might be linked to depression risk.

But in this new study, the Washington University researchers looked again at data from the many studies that delved into the issue since the original publication in 2003, analyzing information from more than 40,000 people, and found that the previously reported connection between the serotonin gene, depression and stress wasnt evident. The new results are published April 4 in the journal Molecular Psychiatry.

Our goal was to get everyone who had gathered data about this relationship to come together and take another look, with each research team using the same tools to analyze data the same way, said the studys first author, Robert C. Culverhouse, PhD, an assistant professor of medicine and of biostatistics. We all ran exactly the same statistical analyses, and after combining all the results, we found no evidence that this gene alters the impact stress has on depression.

Over the years, dozens of research groups had studied DNA and life experiences involving stress and depression in the more than 40,000 people revisited in this study. Some previous research indicated that those with the gene variant were more likely to develop depression when stressed, while others didnt see a connection. So for almost two decades, scientists have debated the issue, and thousands of hours of research have been conducted. By getting all these groups to work together to reanalyze the data, this study should put the questions to rest, according to the researchers.

The idea that differences in the serotonin gene could make people more prone to depression when stressed was a very reasonable hypothesis, said senior investigator Laura Jean Bierut, MD, the Alumni Endowed Professor of Psychiatry at Washington University. But when all of the groups came together and looked at the data the same way, we came to a consensus. We still know that stress is related to depression, and we know that genetics is related to depression, but we now know that this particular gene is not.

Culverhouse noted that finally, when it comes to this gene and its connection to stress and depression, the scientific method has done its job.

Experts have been arguing about this for years, he said. But ultimately the question has to be not what the experts think but what the evidence tells us. Were convinced the evidence finally has given us an answer: This serotonin gene does not have a substantial impact on depression, either directly or by modifying the relationship between stress and depression.

With this serotonin gene variant removed from the field of potential risk factors for depression, Culverhouse and Bierut said researchers now can focus on other gene-environment interactions that could influence the onset of depression.

Culverhouse, RC, et al. Collaborative meta-analysis finds no evidence of a strong interaction between stress and 5-HTTLPR genotype contributing to the development of depression. Molecular Psychiatry. April 4, 2017.

This work was supported by the National Institute on Drug Abuse and the National Institute of Mental Health of the National Institutes of Health (NIH), grant numbers R21 DA033827, MH089995 and R01 DA026911. Other funding provided by the Wellcome Trust and other funding agencies from countries around the world. For a complete list of funding agencies and grants, please refer to the paper.

Potential conflicts of interest involving researchers who are authors of the study also are listed at the end of the paper. Some have received consultancy/speaking fees from various pharmaceutical companies and other business interests. LJ Bierut is one of the listed inventors on US Patent 8 080 371, Markers for Addiction, covering the use of certain DNA SNPs in determining the diagnosis, prognosis and treatment of addiction.

Washington University School of Medicines 2,100 employed and volunteer faculty physicians also are the medical staff of Barnes-Jewish and St. Louis Childrens hospitals. The School of Medicine is one of the leading medical research, teaching and patient-care institutions in the nation, currently ranked seventh in the nation by U.S. News & World Report. Through its affiliations with Barnes-Jewish and St. Louis Childrens hospitals, the School of Medicine is linked to BJC HealthCare.

See the article here:
Study reverses thinking on genetic links to stress, depression - Washington University School of Medicine in St. Louis

Too Much Information? FDA Clears 23AndMe to Sell Home Genetic Tests for Alzheimer’s and Parkinson’s – Scientific American

Genetic testing company 23AndMe is back with a controversial new offering, after the U.S. Food and Drug Administration on Thursday green-lighted the companys request to market a fresh batch of direct-to-consumer tests. Soon, with a simple saliva swab dropped in the mail, customers will be able to get answers about their genetic risk for developing 10 maladiesincluding Parkinsons disease and late-onset Alzheimers.

The FDA approval will likely reignite a long-simmering debate about when and how such tests should be used. Even when there are strong links between certain gene variants and medical conditions, genetic information often remains difficult to interpret. It must be balanced against other factors including health status, lifestyle and environmental influences, which could sharpen or weaken risk. If disease risk news is delivered at homewithout a genetic counselor or doctor on hand to offer contextmany geneticists fear it can lead to unnecessary stress, confusion and misunderstandings.

Against that backdrop, the FDAs decision came with caveats: Results obtained from the tests should not be used for diagnosis or to inform treatment decisions, the agency said in a statement. It added that false positive and false negative findings are possible.

But geneticist Michael Watson, executive director of the American College of Medical Genetics and Genomics, thinks consumers will have trouble making such distinctions and says he doubts people will view them as a mere novelty. Watson also worries 23AndMes wares may create other problems: Follow-up testing for some of these conditions may be quite pricey, he says, and insurance companies might not cover that cost if a person has no symptoms. He also notes that some of the conditions involved may have no proved treatments, leaving consumers with major concernsand few options to address them, aside from steps like making some lifestyle changes.

The makeup of 23AndMes reports to consumers is still being finalized, but the company says it does not expect to grade or rank a persons risk of developing any of the 10 conditions approved for analysis. Instead it will simply report a person has a gene variant associated with any of the maladies and is at an increased risk, the company told Scientific American.

The FDA decision may significantly widen the companys market and top off a years-long debate about what sort of genetic information should be available to consumers without professional medical oversight. After the FDAs 2013 decision to stop 23AndMe from sharing data about disease risk with its customers, the company was still able to offer them information about their genetic ancestry. It has also been selling consumer tests for genes that would indicate whether people are carriers for more than 30 heritable conditions, including cystic fibrosis and Tay-Sachs disease.

This month 23AndMe plans to release its first set of genetic health-risk reports for late-onset Alzheimers disease, Parkinsons disease, hereditary thrombophilia (a blood-clotting disorder), alpha 1-antitrypsin deficiency (a condition that raises the risk of lung and liver disease), and a new carrier status report for Gauchers disease (an organ and tissue disorder). Reports for other tests will follow, the company says.

In considering whether to approve the tests, the FDA says it reviewed studies that demonstrated the 23AndMe procedures correctly and consistently identified variants associated with the 10 conditions. Further data from peer-reviewed scientific literature demonstrated the links between these gene variants and conditions, and supported the underlying science.

The FDA also announced on Thursday that it plans to offer the company exemptions for similar genetic tests in the future, without requiring them to be submitted for premarket review. That decision could leave the door open to offering tests for other conditions that have questionable reproducibility, says Jim Evans, a genetics and medicine professor at the University of North Carolina School of Medicine.

Read the original here:
Too Much Information? FDA Clears 23AndMe to Sell Home Genetic Tests for Alzheimer's and Parkinson's - Scientific American

Screening the dark genome for disease – Science Daily

Researchers have developed a method to swiftly screen the non-coding DNA of the human genome for links to diseases that are driven by changes in gene regulation. The technique could revolutionize modern medicine's understanding of the genetically inherited risks of developing heart disease, diabetes, cancer, neurological disorders and others, and lead to new treatments.

The study appeared online in Nature Biotechnology on April 3, 2017.

"Identifying single mutations that cause rare, devastating diseases like muscular dystrophy has become relatively straightforward," said Charles Gersbach, the Rooney Family Associate Professor of Biomedical Engineering at Duke University. "But more common diseases that run in families often involve lots of genes as well as genetic reactions to environmental factors. It's a much more complicated story, and we've been wanting a way to better understand it. Now we've found a way."

The new technique relies on the gene-hacking system called CRISPR/Cas9. Originally discovered as a natural antiviral defense mechanism in bacteria, the system recognizes and homes in on the genetic code of previous intruders and then chops up their DNA. In the past several years, researchers have harnessed this biologic system to precisely cut and paste DNA sequences in living organisms.

In the current study, researchers added molecular machinery that can control gene activity by manipulating the web of biomolecules that determines which genes each cell activates and to what degree.

With the new tool, Gersbach and his colleagues are exploring the 98 percent of our genetic code often referred to as the "dark matter of the genome."

"Only a small fraction of our genome encodes instructions to make proteins that guide cellular activity," said Tyler Klann, the biomedical engineering graduate student who led the work in Gersbach's lab. "But more than 90 percent of the genetic variation in the human population that is associated with common disease falls outside of those genes. We set out to develop a technology to map this part of the genome and understand what it is doing."

The answer, says Klann, lies with promoters and enhancers. Promoters sit directly next to the genes they control. Enhancers, however, which modulate promoters, can be just about anywhere due to the genome's complex 3D geometry, making it difficult to discern what they're actually doing.

"If an enhancer is dialing a promoter up or down by 10 or 20 percent, that could logically explain a small genetic contribution to cardiovascular disease, for example," said Gersbach. "With this CRISPR-based system, we can more strongly turn these enhancers on and off to see exactly what effect they're having on the cell. By developing therapies that more dramatically affect these targets in the right direction, we could have a significant effect on the corresponding disease."

That's all well and good for exploring the regions of the genome that researchers have already identified as being linked to diseases, but there are potentially millions of sites in the genome with unknown functions. To dive down the dark genome rabbit hole, Gersbach turned to colleagues Greg Crawford, associate professor of pediatrics and medical genetics, and Tim Reddy, assistant professor of bioinformatics and biostatistics. All three professors work together in the Duke Center for Genomic and Computational Biology.

Crawford developed a way of determining which sections of DNA are open for business. That is, which sections are not tightly packed away, providing access for interactions with biomachinery such as RNA and proteins. These sites, the researchers reason, are the most likely to be contributing to a cell's activity in some way. Reddy has been developing computational tools for interpreting these large genomic data sets.

Over the past decade, Crawford has scanned hundreds of types of cells and tissues affected by various diseases and drugs and come up with a list of more than 2 million potentially important sites in the dark genome -- clearly far too many to investigate one at a time. In the new study, Crawford, Reddy and Gersbach demonstrate a high-throughput screening method to investigate many of these potentially important genetic sequences in short order. While these initial studies screened hundreds of these sites across millions of base pairs of the genome, the researchers are now working to scale this up 100- to 1000-fold.

"Small molecules can target proteins and RNA interference targets RNA, but we needed something to go in and modulate the non-coding part of the genome," said Crawford. "Up until now, we didn't have that."

The method starts by delivering millions of CRISPR systems loaded into viruses, each targeting a different genetic point of interest, to millions of cells in a single dish. After ensuring each cell receives only one virus, the team screens them for changes in their gene expression or cellular functions.

For example, someone researching diabetes could do this with pancreatic cells and watch for changes in insulin production. Those cells that show interesting alterations are then isolated and sequenced to determine which stretch of DNA the CRISPR affected, revealing a new genetic piece of the diabetes puzzle.

The technique is already producing results, identifying previously known genetic regulatory elements while also spotting a few new ones. The results also showed it can be used to turn genes either on or off, which is superior to other tools for studying biology which only turn genes off. Different cell types also produced different -- but partially overlapping -- results, highlighting the biological complexity in gene regulation and disease that can be interrogated with this technology.

"Now that we have this tool, we can go in and annotate the functions of these previously unknown but important stretches of our genome," said Gersbach. "With so many places to look, and the ability to do it quickly and robustly, we'll undoubtedly find new segments that are important for disease, which will provide new avenues for developing therapeutics."

See the article here:
Screening the dark genome for disease - Science Daily

New gene discovered driving drug resistance – Medical Xpress

April 4, 2017 Scanning electron micrograph of human T lymphocyte or T cell. Credit: NIAID/NIH

Scientists in Salford, U.K., have identified a gene which is 'revving the engine of cancer' against the world's most common breast cancer drug.

For reasons unknown, 50% of patients with breast cancer treated with the estrogen receptor-blocking drug tamoxifen eventually become resistant to the treatment.

In a paper published this week in the journal Oncotarget, biochemists tested a hypothesis that the mechanism of tamoxifen resistance is related to energy-generating mitochondria in cancer cells.

In doing so, they identified the protein NQ01 as the 'trigger' which determines whether cells would survive tamoxifen or not.

Michael P Lisanti, Professor of Translational Medicine in the Biomedical Research Centre at the University of Salford said: "In simple terms, the process of poisoning the cell (with tamoxifen) actually has the opposite effect, stimulating the cancer cells to respond by revving their engines in order to survive."

Lisanti and collaborators Dr Federica Sotgia and Dr Marco Fiorillo tested their idea that cancer cells were fighting against tamoxifen by using their mitochondria the 'powerhouse of the cell' - that produces all their energy.

In the laboratory they directly compared sensitive cells with tamoxifen-resistant cancer cells, and demonstrated that higher mitochondrial power is what distinguishes a drug-sensitive cell from a resistant cell.

Then they used a combination of protein profiling, genetics and metabolism to identify which genes were necessary to confer tamoxifen-resistance. They observed that by adding just a single gene, NQ01, the cells would survive.

Finally, they used a chemical inhibitor of NQ01 (dicoumarol), which is a relative of warfarin, to successfully sensitise tamoxifen-resistant cells.

Professor Lisanti concludes: "This is the first evidence that tamoxifen resistance is related to a specific metabolic behaviour, ie. increased mitochondrial power, which is important because this is not related to tamoxifen's effect on the estrogen receptor.

"It also confirms that tamoxifen resistance is not a mechanism related to estrogen."

Dr Marco Fiorillo suggests: "Now that we have identified the target, this will allow us and others to design new drugs to overcome tamoxifen resistance. There are already existing experimental drugs for targeting NQO1 and GCLC, for other reasons, so making inhibitors to target these enzymes is a practical reality."

Explore further: Researchers discover key to drug resistance in common breast cancer treatment

More information: Marco Fiorillo et al. Mitochondrial "power" drives tamoxifen resistance: NQO1 and GCLC are new therapeutic targets in breast cancer, Oncotarget (2017). DOI: 10.18632/oncotarget.15852

Journal reference: Oncotarget

Provided by: University of Salford

A new cancer-drug delivery system shows the ability to exploit the oxygen-poor areas of solid tumors that make the growths resistant to standard chemotherapy and radiation treatment.

Multiple myeloma is a cancer of the plasma cells, which are white blood cells produced in bone marrow that churn out antibodies to help fight infection. When plasma cells become cancerous, they produce abnormal proteins, ...

Smoking by either parent helps promote genetic deletions in children that are associated with the development and progression of the most common type of childhood cancer, according to research headed by UC San Francisco. ...

A team led by researchers from the Stanford University School of Medicine has demonstrated a way to diagnose cancer without resorting to surgery, raising the possibility of far fewer biopsies.

The one-size-fits-all approach to early stage breast cancer creates a paradox: Millions of dollars are spent on unnecessary surgeries and radiation to treat women with low-risk 'in situ' lesions, an estimated 85% of which ...

On the list of dreaded medical tests, a prostate biopsy probably ranks fairly high. The common procedure requires sticking a needle into the prostate gland to remove tissue for assessment. Thousands of men who undergo the ...

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

The rest is here:
New gene discovered driving drug resistance - Medical Xpress

NKPD1 gene variant increases depression risk – Medical Xpress

April 4, 2017 Credit: George Hodan/Public Domain

A study of people from an isolated village in the Netherlands reveals a link between rare variants in the gene NKPD1 and depressive symptoms. The findings are published in the current issue of Biological Psychiatry. The study, led by co-first authors Najaf Amin, PhD, of Erasmus University Medical Center in the Netherlands and Nadezhda Belonogova of the Russian Academy of Sciences in Novosibirsk, Russia, helps researchers understand the molecular pathology of the disease, which could eventually improve how depression is diagnosed and treated.

Genetics play a strong role in risk for depression, but the identification of specific genes contributing to the disorder has eluded researchers. "By sequencing all of the DNA that codes for mRNA and ultimately, proteins, Dr. Amin and colleagues found a single gene that may account for as much as 4% of the heritable risk for depression," said Doctor John Krystal, Editor of Biological Psychiatry.

To identify the gene, the researchers assessed data from the Erasmus Rucphen Family study, which was composed of a collection of families and their descendents living in social isolation until the past few decades. In a population like this, genetic isolation leads to an amplification of rarely occurring variants with little other genetic variation, providing a more powerful cohort for the discovery of rare variants. Nearly 2,000 people who had been assessed for depressive symptoms were included in the analysis.

Using whole-exome sequencing to examine portions of DNA containing genetic code to produce proteins, Amin and colleagues found that several variants of NKPD1 were associated with higher depressive symptom scores. The association between depressive symptoms and NKPD1 were also replicated in an independent sample of people from the general population, although the replication sample highlighted different variants within NKPD1.

"The involvement of NKPD1 in the synthesis of sphingolipids and eventually of ceramides is interesting," said Dr. Amin, referring to the predicted role of NKPD1 in the body. Altered sphingolipid levels in blood have been associated with depression and have been proposed as a therapeutic target for major depressive disorder.

"We are the first to show a possible genetic connection in this respect," said Dr. Amin, adding that this implies that such a therapy might be beneficial for patients carrying risk variants in the NKPD1 gene.

As with other psychiatric disorders, depression lacks genetic or biochemical markers to aid diagnosis and treatment of the disorder. According to Dr. Amin, moving depression treatment into the era of precision and personalized medicine will require a transition to objective and unbiased measurements where patients are stratified based on the molecular pathology of the disease. "NKPD1 may be one such molecular mechanism," she said.

Explore further: Earlier and more severe depression symptoms associated with high genetic risk for major psychiatric disorders

More information: Najaf Amin et al. Nonsynonymous Variation inNKPD1Increases Depressive Symptoms in European Populations, Biological Psychiatry (2017). DOI: 10.1016/j.biopsych.2016.08.008

Journal reference: Biological Psychiatry

Provided by: Elsevier

Clinical features of major depressive disorder (MDD) may help identify specific subgroups of depressed patients based on associations with genetic risk for major psychiatric disorders, reports a study in Biological Psychiatry. ...

An international group of more than 190 scientists who analyzed the genomes of 298,420 individuals have found genetic variants that may influence our sense of wellbeing, depression and neuroticism.

Antidepressants are commonly prescribed for the treatment of depression, but many individuals do not experience symptom relief from treatment. The National Institute of Mental Health's STAR*D study, the largest and longest ...

Depression symptoms that steadily increase in older adults are more strongly linked to dementia than any other types of depression, and may indicate the early stages of the disease, according to the first ever long-term study ...

(Medical Xpress)A very large team of researchers made up mostly of members in China and calling itself the CONVERGE consortium, has identified two genetic variants that appear to be associated with major depressive disorder ...

New research findings often garner great attention. But when other scientists follow up and fail to replicate the findings? Not so much.

How often do you act impulsively without considering the consequences? What if you could learn how to be more patient?

Scientists at the University of Basel have shown that LSD reduces activity in the region of the brain related to the handling of negative emotions like fear. The results, published in the scientific journal Translational ...

Scientists at the Center for Infection and Immunity (CII) at Columbia University's Mailman School of Public Health are the first to report immune signatures differentiating two subgroups of myalgic encephalomyelitis/chronic ...

The common antibiotic doxycycline can disrupt the formation of negative associations in the brain, according to new research from UCL and the University of Zurich.

A study of people from an isolated village in the Netherlands reveals a link between rare variants in the gene NKPD1 and depressive symptoms. The findings are published in the current issue of Biological Psychiatry. The study, ...

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

Continued here:
NKPD1 gene variant increases depression risk - Medical Xpress

SOD1 gene – Genetics Home Reference – NIH

At least 200 mutations in the SOD1 gene have been found to cause amyotrophic lateral sclerosis (ALS), a condition characterized by progressive muscle weakness, a loss of muscle mass, and an inability to control movement. Most of these mutations change one of the protein building blocks (amino acids) in the superoxide dismutase enzyme. About half of all Americans with ALS caused by SOD1 gene mutations have a particular mutation that replaces the amino acid alanine with the amino acid valine at position 5 in the enzyme, written as Ala5Val or A5V. (Because of variations in the ways amino acids are counted in proteins, this mutation is sometimes called Ala4Val or A4V.) ALS caused by the A5V mutation is generally associated with a shorter life expectancy compared with ALS caused by other genetic mutations.

ALS is caused by the death of nerve cells that control muscle movement (motor neurons). It is unclear why these cells are particularly sensitive to SOD1 gene mutations. Researchers have suggested several ways in which the altered enzyme may cause the death of motor neurons. These possibilities include an increase in harmful superoxide radicals, increased production of other types of toxic radicals, increased cell death, or accumulation of clumps (aggregates) of misfolded superoxide dismutase that may be toxic to cells.

Read more from the original source:
SOD1 gene - Genetics Home Reference - NIH

Study reveals gene therapy may help in treating cardiac disease – The Siasat Daily

California: In a breakthrough study, researchers have found a potential treatment for life-threatening cardiac diseases by using gene therapy.

Danon disease is a very rare, life-threatening condition where the fundamental biological process of removing and recycling proteins does not work.

This impairment results in dysfunction of the heart, skeletal muscle, neurologic system, eyes, and liver. Most patients die or require heart transplants by the third decade of life.

In the study, which was published in Science Translational Medicine, researchers have identified a novel way to treat Danon disease using gene therapy.

Heart transplant is not always available for patients and does not treat the other organs affected in Danon disease. We knew we needed to find therapies specifically designed to address the underlying cause, said the lead researcher Eric Adler.

Danon disease is a result of mutations in the gene LAMP2. For nearly a decade, Adler and a team of researchers at UC San Diego Health have been working to determine whether gene therapy could provide a new treatment approach.

Gene therapy involves either replacing or repairing a gene that causes a medical problem or adding genes to help the body treat disease. In this case, Adler and the team focused on adding a specially designed gene that restores the LAMP2 function, resulting in improved cardiac and liver function.

We utilised mice that were a model for Danon disease and missing this specific LAMP gene. We applied gene therapy to a group of these mice and compared to mice that did not receive treatment, said Adler.

The mice that received gene therapy expressed positive results in heart, liver and muscle function. The hearts overall function of ejecting blood and relaxing improved, as did the bodys ability to degrade proteins and metabolism.

Danon disease is more common in males, and symptoms begin in early childhood or adolescence.

In many cases, the condition is inherited by a parent, typically the mother. We believe Danon disease is actually more common than we think, but it is often misdiagnosed, said Adler.

By utilising gene therapy, we were able to identify a possible new treatment approach other than a heart transplant. This study is a significant step for patients with Danon disease, Adler added.

Prior studies in Adlers lab have focused on using a patients skin cells to create stem cells. These stem cells were used to create a heart model, allowing researchers to study Danon disease at the cellular level.

The approach has provided new insight into the diseases pathology and led to the idea of using gene therapy. Our work is also proof that using stem cells to model diseases has great potential for helping develop new medicines, said Adler.

The next step, said Adler, is testing in patients with Danon disease. A Phase I clinical trial for safety and efficacy has begun.

This is the first trial using gene therapy to treat a genetic cardiac disorder and three patients are currently being treated, which means were that much closer to finding a cure for this terrible disease, and may be able to use similar methods to treat other diseases, said Adler.

Get the latest updates in Technology, Entertainment, Sports, Politics and Top Stories on WhatsApp & Telegram by subscribing to our channels. You can also download our app for Android and iOS.

Read the rest here:
Study reveals gene therapy may help in treating cardiac disease - The Siasat Daily

Ionis and partner announce enrollment completion of global Phase 3 GENERATION HD1 study for Huntington’s disease – BioSpace

CARLSBAD, Calif., April 20, 2020 /PRNewswire/ -- Ionis Pharmaceuticals, Inc. (NASDAQ: IONS), the leader in RNA-targeted therapeutics, today announced that its partner Roche, also known as Genentech in the United States, has completed enrollment for GENERATION HD1, a global Phase 3 study evaluating the efficacy and safety of tominersen (previously IONIS-HTTRx or RG6042), an investigational antisense therapy for people living with Huntington's disease (HD).

"Completion of the enrollment of this Phase 3 study is an important landmark for the clinical development of tominersen and for families affected by Huntington's disease. While there is much work ahead of us, we are now closer to potentially providing a treatment for people living with this devastating disease. We are grateful to Huntington's disease patients, their families and healthcare providers for their courage and resilience, particularly in the current challenging environment," said Brett P. Monia, Ph.D., Ionis' chief executive officer. "At Ionis, knowing that sick people depend on us fuels our passion for discovering and delivering novel antisense medicines like tominersen, the first and only therapy in pivotal trials targeting the underlying cause of HD."

GENERATION HD1 is evaluating the efficacy and safety of tominersen treatment administered once every two months (eight weeks) or every four months (16 weeks) over a period of 25 months, compared to placebo. The study has completed enrollment with 791 patients across approximately 100 sites around the world.

HD is a devastating, and ultimately fatal, hereditary disease resulting in deterioration in mental abilities and physical control. Currently, there is no approved disease-modifying treatment for HD. There are approximately 3 to 10 per 100,000 people worldwide affected by HD. In the U.S. alone, there are approximately 40,000 people with symptomatic HD and more than 200,000 people at risk of having inherited the gene that causes HD.

About tominersenTominersen, previously IONIS-HTTRx or RG6042, is an investigational antisense therapy designed to reduce the production of all forms of the huntingtin protein (HTT), including its mutated variant, mHTT. Tominersen is the first therapy in pivotal trials targeting the underlying cause of HD. In December 2017, Roche licensed the investigational molecule from Ionis.

In the Phase 1/2 study, 46 people with early stage HD were treated with tominersen or placebo for 13 weeks. The data demonstrated significant, dose-dependent reductions in mHTT in the cerebrospinal fluid (CSF) of treated participants with a favorable safety and tolerability profile.

Tominersen is being investigated in a Phase 3 study (GENERATION HD1), an open label extension study in HD patients and a Phase I pharmacokinetics and pharmacodynamics study (GEN-PEAK). These studies, in addition to the non-interventional HD Natural History Study, are important elements of the clinical program to thoroughly evaluate the potential of tominersen to be the first disease-modifying medicine for the treatment of HD. The Phase 3 GENERATION HD1 study is expected to complete in 2022. The timing for this study's completion remains unchanged.

Additional information about tominersen clinical trials may be found at https://clinicaltrials.gov/ct2/show/NCT03761849.

About Ionis Pharmaceuticals, Inc.As the leader in RNA-targeted drug discovery and development, Ionis has created an efficient, broadly applicable, drug discovery platform called antisense technology that can treat diseases where no other therapeutic approaches have proven effective. Our drug discovery platform has served as a springboard for actionable promise and realized hope for patients with unmet needs. We created the first and only approved treatment for children and adults with spinal muscular atrophy as well as the world's first RNA-targeted therapeutic approved for the treatment of polyneuropathy in adults with hereditary transthyretin amyloidosis. Our sights are set on all the patients we have yet to reach with a pipeline of more than 40 novel medicines designed to potentially treat a broad range of disease, including neurological, cardiovascular, infectious, and pulmonary diseases.

To learn more about Ionis visit http://www.ionispharma.com or follow us on twitter @ionispharma.

Ionis' Forward-looking StatementThis press release includes forward-looking statements regarding Ionis' alliance with Roche and the development, activity, therapeutic potential, commercial potential and safety of tominersen (IONIS-HTTRx or RG6042). Any statement describing Ionis' goals, expectations, financial or other projections, intentions or beliefs is a forward-looking statement and should be considered an at-risk statement. Such statements are subject to certain risks and uncertainties, particularly those inherent in the process of discovering, developing and commercializing medicines that are safe and effective for use as human therapeutics, and in the endeavor of building a business around such medicines. Ionis' forward-looking statements also involve assumptions that, if they never materialize or prove correct, could cause its results to differ materially from those expressed or implied by such forward-looking statements. Although Ionis' forward-looking statements reflect the good faith judgment of its management, these statements are based only on facts and factors currently known by Ionis. As a result, you are cautioned not to rely on these forward-looking statements. These and other risks concerning Ionis' programs are described in additional detail in Ionis' annual report on Form 10-K for the year ended December 31, 2019, which is on file with the SEC. Copies of this and other documents are available from the Company.

In this press release, unless the context requires otherwise, "Ionis," "Company," "we," "our," and "us" refers to Ionis Pharmaceuticals and its subsidiaries.

Ionis Pharmaceuticals is a trademark of Ionis Pharmaceuticals, Inc.

View original content to download multimedia:http://www.prnewswire.com/news-releases/ionis-and-partner-announce-enrollment-completion-of-global-phase-3-generation-hd1-study-for-huntingtons-disease-301043200.html

SOURCE Ionis Pharmaceuticals, Inc.

Company Codes: NASDAQ-NMS:IONS

The rest is here:
Ionis and partner announce enrollment completion of global Phase 3 GENERATION HD1 study for Huntington's disease - BioSpace

A Rare Genetic Mutation Reveals Secrets of the Common Cold – Discover Magazine (blog)

(Credit: nenetus/Shutterstock)

A rare mutation that nearly killed a young girl has revealed insights into the common cold.

Researchers from theNational Institute of Allergy and Infectious Diseases conducted a genetic analysis of a child who had been laid low by repeated boutsof rhinovirus (the virus that causes colds) and influenza infections severe enough toplace her on life support. By combing through her genome, they found a single mutation that they say obstructed her bodys natural disease-fighting pathway. The finding not only helped to solve a medical mystery, it could also give us new assets in the fight against common viral infections.

When a virus enters our cells and begins churning out copies of itself, the cells normally react by releasing signaling proteins called interferons. This cellular distress signal alerts other cells in the vicinity to be on guard against the virus, and is a crucial step in our bodies ability to fight disease. This particular patient, however, had a mutation that prevented her cells from recognizing and responding to rhinovirus, allowing it to proliferate throughout her body unchecked.

Her health eventually improved somewhat as her immune system grew stronger, although at five years of age, she is still at risk.

Researchers were already aware that the misbehaving protein called MDA5 was involved with signaling the immune system when a virus began to spread, but their findings help to pin down an explicit link to rhinovirus infections. It gives us more insight into how the common cold is contracted and spreads through the body, which is helpful, given that we still dont have an effective means of preventing the virus. Although colds arent normally seen as a serious disease, they can be deadly for individuals with asthma, COPD and other lung diseases, as well as the elderly.

While the research here, published Monday in the Journal of Experimental Medicine,was confined to a single person with a rare mutation, the benefits could extendmuch further. Up until they encountered an individual with this mutation, the researchers were unaware of the role MDA5 proteins play in helping to protect us against colds, a discovery that could guide treatments in the future. The concept is similar to how researchers working with so-called knockout mice with specific genes disabled uncover the secrets of our genomes. Sometimes, its only by seeing what goes wrong when a gene is missing that we can figure out what it does. Cases such as this highlight the value of rare occurrences in medicine, according to study lead author Helen Su.

Its not just because were trying to treat the rare people, but because they give us an insight that would not be easily obtained by studying lots of normal people, she says. They can actually be very instructive.

Though this is the only confirmed MDA5 mutation of this nature, more likely exist, Su says. She and her colleagues also combed through a database of 60,000 genomes looking for similar mutations in the IFIH1 gene that codes for the protein. They found several variations that they say could lead to impaired immune functions, although none seemed to materialize in these individuals, indicating that multiple factors are likely at play.

As for the rest of us, dont worry, the mutation is rare, and if youre reading this youre almost certainly fine. Su emphasizes that colds arent aproblem for healthy individuals, and only become a problem when other issues arise.

In some rare cases, however, all it takes is a single genetic variation for a common illness to take a deadly turn.

View post:
A Rare Genetic Mutation Reveals Secrets of the Common Cold - Discover Magazine (blog)

Blindness Gene Therapy Becomes Most Expensive Medicine In U.S.

With a sticker price of $850,000, a gene therapy for blindness will be the most expensive medicine sold in the United States.

The treatment called Luxturna is manufactured by Spark Therapeutics and helps treat Leber congenital aumaurosis, an inherited condition that leads to blindness. The rare condition only affects two to three people per 100,000. The treatment was expected to cost $1 million, but the company said it brought prices down over concerns of accessibility to the drug.

We wanted to balance the value and the affordability concerns with a responsible price that would ensure access to patients, said Jeffrey Marrazzo in an interview Wednesday.

The drug received approval from the U.S. Food and Drug Administration in December, and given its success, most insurers are likely to cover the treatment.

If they decided not to cover it they would immediately have to face negative publicity, said Meredith Rosenthal, a professor of health economics at Harvard University to the Toronto Star.

To further allay concerns over the cost of the drug, Philadelphia-based Spark will use unconventional pricing models and schemes with insurers. Spark reached an agreement with insurer Harvard Pilgrim Wednesday on a rebates program to reimburse the insurer a portion of the procedure if patients dont see the expected improvement in vision.

As far as the price, and the structures to pay the price, I think its all pretty much in line with what were seeing in other innovative therapies, said Dr. Stuart Orkin, a pediatric oncologist at the Dana-Farber Cancer Institute and Boston Childrens Hospital to health website STAT. I do applaud them for thinking through the payment schemes. Its better than if they had just put out a price and said, you know, Youre paying it.'

Luxturna, Spark argues, is much cheaper in the long run than a lifetime of blindness. Luxturna is injected into both eyes to provide patients with a functioning copy of a gene that is defective in their eyes. The non-profit Institute for Clinical and Economic Review, however, said that the drug would need to be far cheaper to be a cost-effective treatment.

At least one medicine in Europe was more expensive, surpassing the $1 million mark, but the gene therapy for a rare protein disorder has been discontinued.

See the original post:
Blindness Gene Therapy Becomes Most Expensive Medicine In U.S.

A New Gene Editing Technique Could Finally Allow Us to Treat ALS – Futurism

In BriefResearchers from UC San Diego's School of Medicine have tested a modified CRISPR-Cas9 technique designed to target RNA instead of DNA. Rcas9 could potentially improve the lives of patients with ALS, Huntington's disease, or myotonic dystrophy by delaying the progression of their disorders.

The most efficient and effective gene-editing tool in use today is CRISPR-Cas9. Just this year, researchers have successfully used it fora wide variety of experiments, from modifying garden vegetables to encoding a GIF in bacterial DNA. Most recently, the tool was used to remove a genetic disease from a human embryo.

Although undeniably powerful, CRISPR-Cas9 does have its limitations; it can only target DNA. To extend its capabilities to includeRNA editing, researchers from the University of California San Diego (UCSD) School of Medicinedeveloped amodification of CRISPR, and theyre calling their toolRNA-targeting Cas9 (RCas9).

In a study published in Cell, the UCSD team tested their technique by correcting the kinds of molecular mistakes that cause people to develop microsatellite repeat expansion diseases, such ashereditary amyotrophic lateral sclerosis (ALS)and Huntingtons disease.

During standard CRISPR-CAs9 gene editing, a guide RNA is instructed to deliver a Cas9 enzyme to a specific DNA molecule. The researchers from UCSD instead instructed it to target an RNA molecule.

Tests conducted in the laboratory showed that RCas9 removed 95 percent ofproblem-causing RNA for myotonic dystrophy types 1 and 2, Huntingtons disease, and one type of ALS. The technique also reversed 93 percent of the dysfunctional RNA targets in the muscle cells of patients with myotonic dystrophy type 1, resulting in healthier cells.

This is exciting because were not only targeting the root cause of diseases for which there are no current therapies to delay progression, but weve re-engineered the CRISPR-Cas9 system in a way thats feasible to deliver it to specific tissues via a viral vector, senior author Gene Yeo, a cellular and molecular medicine professor at UCSD School of Medicine, explained in a press release.

Across the globe, an estimated 450,000 patients are said to be living with ALS. Roughly 30,000 of those are from the U.S. where 5,600 people are diagnosed with the diseases every year. The exact number of Huntingtons disease cases, however, isnt quite as easy to pin down. One estimate says that around 30,000 Americans display symptoms of it, while more than 200,000 are at risk.

Regardless of the exact numbers, these two neurological diseases clearly affect a significant number of people. This prevalence and the absence of a known curemakes the UCSD teams research all the more relevant. Even more exciting is the fact that the same kinds of RNA mutations targeted by this study are known to cause more than 20 other genetic diseases.

Our ability to program the RCas9 system to target different repeats, combined with low risk of off-target effects, is its major strength, co-first author of the study Ranjan Batra said in the UCSD press release.

However, the researchers do know that what theyve accomplished is just a first step. While RCas9 works in a lab, they still have to figure out how it will fare when tested in actual patients.

The main thing we dont know yet is whether or not the viral vectors that deliver RCas9 to cells would elicit an immune response, explained Yeo. Before this could be tested in humans, we would need to test it in animal models, determine potential toxicities, and evaluate long-term exposure.

Ultimately, while RCas9 couldnt exactly deliver a cure, it could potentially extend patients healthy years. For disease like ALS and Huntingtons, thats a good place to start.

Read more from the original source:
A New Gene Editing Technique Could Finally Allow Us to Treat ALS - Futurism

Combination therapy targets genetic mutation found in many cancers – Medical Xpress

June 2, 2017 Credit: CC0 Public Domain

A study at The University of Texas MD Anderson Cancer Center has shown promise for effective treatment of therapy-resistant cancers caused by a mutation of the RAS gene found in many cancers. The pre-clinical study combined therapies targeting the inhibitors polyADP ribose polymerase (PARP) and mitogen-activated protein kinase (MEK). The findings were published this week in Science Translational Medicine.

Mutations in the RAS gene account for more than 90 percent of pancreatic cancers, 50 percent of colorectal cancers, and 30 percent of lung cancers, and a significant portion of many other types of tumors. Unfortunately, these cancers are usually resistant to traditional treatments contributing to poor patient outcomes.

"Nowhere is the need for targeted therapies greater than for cancers driven by oncogenic RAS, which represents the most common type of potentially targetable mutation in cancer," said Gordon Mills, M.D., Ph.D., chair of Systems Biology. "Our study demonstrated that the rational combination of PARP and MEK inhibitors warrants clinical investigation in patients with RAS-mutant tumors where there are few effective therapeutic options."

PARP inhibitors block a key pathway for cellular DNA repair, effectively stopping many cancers with defects in DNA repair from growing, but the disease soon gains resistance due to the tumor's cell ability to adapt to stresses caused by the therapy. MEK inhibitors also are used to affect pathways often overactive in some cancers.

Mills' team found that combinations of PARP and MEK inhibitors evoked "unexpected cytotoxic effects" in vitro and in vivo in multiple RAS-mutant tumor models across tumor lineages where RAS mutations are prevalent. The combination therapy worked independent of mutations in tumor suppressor genes including BRCA1, BRCA2 and p53, suggesting the dual therapy's potential as a treatment for multiple RAS-mutant cancers. It also was effective for tumors that had become resistant to PARP, as well as in cells that did not have aberrations in BRCA1 and BRCA2, suggesting the combination could expand to a wide spectrum of patients likely to benefit.

"The sensitivity of RAS-mutant cells to the combination appears to be independent of intrinsic gene expression patterns, as observed across multiple different lineages," said Mills. "Because the synergistic responses to MEK1 and PARP1 combinations also were independent of p53 mutation status, the approach should be effective in both normal and mutant p53 tumors. Together, the in vitro and in vivo data argue that a MEK1 and PARP1combination offer the potential to induce cell death and increase the magnitude, duration and spectrum of PARP activity."

Currently, clinical trials in this area of investigation are under consideration at MD Anderson.

Explore further: New findings may enhance PARP inhibitors therapy in breast cancer

More information: C. Sun el al., "Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers," Science Translational Medicine (2017). stm.sciencemag.org/lookup/doi/10.1126/scitranslmed.aal5148

Findings from a new study reveal that PARP inhibitors, an emerging class of drugs being studied in cancer clinical trials, may be enhanced by combining them with inhibitors targeting an oncogene known as c-MET which is overexpressed ...

Researchers from the Chinese Academy of Sciences have discovered that the metabolic enzyme phosphoglycerate mutase 1 (PGAM1) helps cancer cells repair their DNA and found that inhibiting PGAM1 sensitizes tumors to the cancer ...

Researchers from the Wellcome Trust Sanger Institute and their collaborators have discovered that a greater number of breast cancers are genetically similar to rarer cases with faulty BRCA1 or BRCA2 genes. The results published ...

Poly (ADP-Ribose) polymerase (PARP) inhibitors, shown to have clinical activity when used alone in women with familial breast and ovarian cancers linked to BRCA mutations, may be a novel treatment strategy in women with HER2-positive ...

About one-third of patients with ovarian cancer who wouldn't be expected to respond to a PARP inhibitor had partial shrinkage of their tumor when a kinase inhibitor was added to treatment, report scientists from Dana-Farber ...

A new imaging test may provide the ability to identify ovarian cancer patients who are candidates for an emerging treatment that targets a key enzyme cancer cells need to survive. Currently, epithelial ovarian cancer patients ...

Adding a new anti-hormonal drug to the standard treatment for advanced prostate cancer has been shown to reduce the risk of dying by almost 40 percent, according to two studies published Saturday.

Adding abiraterone to hormone therapy at the start of treatment for prostate cancer improves survival by 37 per cent, according to the results of one of the largest ever clinical trials for prostate cancer presented at the ...

The immunotherapy drug pembrolizumabalready FDA-approved for other forms of cancer-has been found to be effective in patients with metastatic triple negative breast cancer, according to an international clinical trial ...

A study at The University of Texas MD Anderson Cancer Center has shown promise for effective treatment of therapy-resistant cancers caused by a mutation of the RAS gene found in many cancers. The pre-clinical study combined ...

The use of immunotherapy to treat cancer is celebrating its first successes but there are still many knowledge gaps in the underlying mechanisms of action. In a study of mice with soft tissue tumors, ETH researchers have ...

A compound found in olive oil may help to prevent cancer developing in the brain, a study shows.

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

More here:
Combination therapy targets genetic mutation found in many cancers - Medical Xpress