[Full text] Association of CYP3A5 Gene Polymorphisms and Amlodipine-Induced Periph | PGPM – Dove Medical Press

Introduction

Calcium channel blockers (CCBs) are widely used in the treatment of hypertension. In addition to their antihypertensive effects, CCBs may also lead to many adverse reactions, including peripheral edema, dizziness, flushing, fatigue, headache, palpitations, and gingival enlargement.14 Peripheral edema, particularly of lower limbs, is the most common side effect of CCBs. CCBs-induced peripheral edema is more common in women and is related to age, upright posture and duration of CCBs therapy.2,5 Although it has been known for decades that CCBs could cause peripheral edema, there are still very few genetic markers that could be used in clinical treatment.

Amlodipine is a first-line antihypertensive drug and a long-acting 1,4-dihydropyridine calcium channel blocker.6 It became the fifth most prescribed medication in the United States in 2018.7 However, therapy trials report that the incidence of amlodipine-induced peripheral edema is about 19%, which also probably serves as the main reason behind the limited use of this drug.8,9 To our knowledge, factors that increase the serum concentration of drugs may also increase the risk of side effect. Accordingly, genetic factors affecting the metabolism of amlodipine will consequently influence its clearance, thus possibly resulting in peripheral edema. In recent years, numerous studies have focused on identifying genetic factors determining the responses to amlodipine therapy, including aspects of efficacy and safety.1012 Nevertheless, the genetic predisposition to amlodipine-induced peripheral edema remains unclear. However, some studies shine light on CYP genes, which are involved in the metabolism of amlodipine.13,14

CYP3A is a subfamily of cytochrome P450 and is involved in the metabolism of many compounds.10,1517 CYP3A4 and CYP3A5 are the two major enzymes of CYP3A family and play important roles in the dehydrogenation of amlodipine.18 However, it has been reported that CYP3A4, rather than CYP3A5, plays a more important role in amlodipine clearance in vivo.16 However, according to our observations, almost all SNPs of CYP3A4 are very rare in Chinese Han or East Asian.1921 A recent study evaluated the influence of CYP3A polymorphisms on the pharmacokinetic (PK) parameters of 10 CYP3A substrates including amlodipine, and it turned out CYP3A4 polymorphisms did not show a pronounced influence on PK of amlodipine.22 So, as far as we know, CYP3A4 does not suffice as an explanation towards the incidence of amlodipine-induced adverse responses. Additionally, the contribution of CYP3A5 in amlodipine metabolism is still controversial. CYP3A5 expression varies among individuals due to gene polymorphisms that result in non-productive mRNA splicing and lower or undetectable expression of the protein.14,23,24 Pharmacogenomics studies have found that CYP3A5 gene polymorphisms have significant impact on drug metabolism, efficacy, and toxicity.25,26 Recent studies have reported a possible relationship between CYP3A5 and amlodipine. CYP3A5*3/*3 genotypes might be associated with blood pressure response to amlodipine,27 and CYP3A5*3 may affect the disposition of amlodipine.13 In Chinese hypertensive patients who underwent renal transplantation, researchers have demonstrated that CYP3A5*3 polymorphism affects the antihypertensive efficacy of amlodipine. In such patients with CYP3A5*3/*3, the reduction in diastolic blood pressure (DBP) was significantly higher than those with other genotypes.28 Additionally, compared with CYP3A5*1/*1, CYP3A5*1/*3 patients exhibit a lower metabolic ratio of amlodipine in vivo and hypertensive patients with CYP3A5*1/*3 genotypes show a higher decrease in blood pressure.14 These findings suggest that CYP3A5 genetic variants are really involved in amlodipine response, having an impact on treatment effectivity of amlodipine.

Although studies have identified that CYP3A5 polymorphisms do play a role in blood pressure control and treatment responses of amlodipine, the effect of a particular genotype in amlodipine-induced peripheral edema remains unclear. Here, we aim to test whether any genetic variants associate with amlodipine-induced peripheral edema. For this, we used the targeted region sequencing method to genotype the SNPs in all known haplotypes of CYP3A5, and evaluated their effect on amlodipine-induced peripheral edema in Chinese Han hypertensive patients.

We recruited a total of 240 patients with essential hypertension who have ever been treated with amlodipine or L-amlodipine for more than 4 weeks. The participants were outpatients from Beijing Chaoyang Hospital of Capital Medical University and the Second Hospital of Jilin University. The demographic characteristics and baseline data were collected, including gender, age, body mass index (BMI), smoking and drinking status, blood pressure, heart rate and use of medications. Participants were also questioned about peripheral edema symptoms (leg or ankle edema). Two trained physicians evaluated the adverse drug reactions caused by the amlodipine or L-amlodipine based on the WHO-UMC scale.29 We classified the participant as a peripheral edema case if WHO-UMC causality categories were certain, probable, or possible. The others were grouped as controls.

The DNA Sampling Swabs (Taitong Gene Testing Equipments Co., Ltd., Suzhou, China) were used for collecting oral buccal mucosa cells. Genomic DNA was isolated from the swabs, using Hi-Swab DNA kit (TIANGEN, Beijing, China) according to the manufacturers instructions. Quantification of the DNA concentration was performed using the manufacturers protocol with the Qubit dsDNA HS Assay Kit (Yeasen, Shanghai, China). Methods for targeted region capture and NGS have been reported previously.30 Illumina HiSeq X was used to sequence the captured DNA with paired-end reads of 150 bp length. Table S1 lists the targeted regions of sequencing and the 24 SNPs of CYP3A5.

High-quality sequencing reads were obtained from the raw data by removing reads that contained adapters, were with unknown bases, or had a low-quality using the Trimmomatic (v0.36)31 program. The high-quality reads were aligned to human reference genome hg19 using the Burrows-Wheeler Aligner (BWA, v0.7.15)32 using the default parameters. The Genome Analysis ToolKit (GATK, v3.8)33 was used for indels realignment, quality score recalibration, variant calling, and genotyping (using Haplotype Caller).

Demographic and clinical characteristics of different groups were compared by t-test or Chi-square (2) test according to the data category. The associations between gene polymorphisms and the risk of peripheral edema were assessed by codominant model, dominant model, recessive model and allele model by calculating the odds ratios (ORs) and 95% confidence intervals (CIs) using logistic regression with or without adjustment by gender and alcohol status. Stratification was done by gender. Analyses above were carried out on R-4.03. PLINK 1.934 was used to calculate the minor allele frequency and assess HardyWeinberg equilibrium (HWE) for each SNP. Additionally, linkage disequilibrium (LD) block and haplotype were assessed by Haploview35 software. The D and r2 values for all pairs of SNPs were calculated. P value<0.05 was considered as the significant level.

Two hundred and forty enrolled patients were separated into 64 cases and 176 controls. The general characteristics of the study population are summarized in Table 1. In agreement with previous reports, a higher incidence of CCB-induced peripheral edema was observed in women. A significant difference was found in the sex ratio between cases and controls (P=0.00048). In addition, the frequency of drinkers was 29.69% in the case group and 46.02% in the control group (P=0.034). There was no significant difference between the two groups regarding other characteristics.

Table 1 Characteristics of the Study Population

All the observed SNPs and the minor allele frequencies (MAF) in two groups are listed in Table 2. Except rs15524, rs4646453 and rs776746, the other SNPs were rare or not detected in the studied population. Therefore, we focused on these three SNPs (MAF>0.05) for further studies. Distributions of genotype frequencies of the SNPs did not show any deviation in HardyWeinberg equilibrium (P>0.05).

Table 2 Observed CYP3A5 Variations and Frequencies

The genotype and allele allocations of the test polymorphisms differed considerably between cases and controls (Table 3). In more detail, the frequencies of alleles rs15524 G, rs4646453 A, and rs776746 T were significantly lower in cases than those in the control group (G vs A: OR=0.53, P=0.011; A vs C: OR=0.54, P=0.019; T vs C: OR=0.58, P=0.03; respectively). Furthermore, there was a statistically significant difference in genotype of the rs15524 and rs4646453 between the two groups in dominant model with or without adjustment by gender and alcohol status (GG+AA vs AA: OR=0.5, P=0.021; AA+AC vs CC: OR=0.54, P=0.04). As for rs776746, the presence of TT+CT genotype demonstrated a significantly lower risk after gender and alcohol status adjustment (TT+CT vs CC: OR=0.57, adjusted P=0.044).

Table 3 Genotype Frequencies of Study SNPs in Case and Control Groups

Stratified analyses were performed to access the differential effect of gender on the association between amlodipine-induced edema and the polymorphisms. No significant differences were identified, but the relevant genotypes still showed lower risk in all subgroups. The details are presented in Table 4.

Table 4 Stratified Analyses Between SNPs and Risk of Amlodipine-Induced Peripheral Edema

We employed Haploview program to assess the Linkage disequilibrium (LD) block and haplotype of the three SNPs of CYP3A5. The LD analysis indicated that these SNPs were in strong LD with each other (Figure 1, rs15524 and rs4646453 D=0.965, r2=0.821; rs15524 and rs776746 D=0.979, r2=0.919; rs776746 and rs4646453 D=0.989, r2=0.898). Frequencies of four haplotypes were found to be more than 1% in the haplotype analysis (Table 5). The most represented haplotype in the whole cohort of controls and cases was ACC, followed by GAT, GCC and GCT. Two haplotypes (ACC and GAT) were significantly associated with the risk of amlodipine-induced peripheral edema. The frequency of the ACC haplotype was higher in the cases than controls (79.7% vs 67.9%, P=0.012), whereas the frequency of the GAT haplotype was lower in the cases (17.2% vs 28.1%, P=0.015).

Figure 1 Linkage disequilibrium coefficients (|D|) and LD block among the three polymorphisms of CYP3A5.

Table 5 Haplotype Frequencies of CYP3A5 Gene and the Association with the Amlodipine-Induced Peripheral Edema Risk

Studies have demonstrated that genetic polymorphisms may influence the gene function, thus causing alterations in the pharmacokinetics (PK) and pharmacodynamics (PD) of the gene-associated drugs. Moreover, genetic variants have been confirmed for their association with appearance of adverse reactions to drugs, such as ACE inhibitors-induced cough36 and rosuvastatin-induced myotoxicity.37 To our knowledge, this is the first study to investigate the relationships of CYP3A5 polymorphisms and amlodipine-induced peripheral edema by using a casecontrol retrospective study in the Chinese population. Our study reveals that amlodipine-induced peripheral edema may associate with genetic polymorphisms in CYP3A5 gene. We find that the distribution of allele and genotype frequencies of the three SNPs are significantly different between cases and controls. Specifically, the alleles rs15524 G, rs4646453 A, and rs776746 T reduce the risk of amlodipine-induced peripheral edema. On the other hand, A, C, and C increase the risk and haplotype analysis also confirms this. These findings support the hypothesis that genetic variation in CYP3A5 is involved in the development of amlodipine-induced peripheral edema.

CYP3A5 is highly polymorphic with significant inter-individual variation in the enzyme activity contributing to the absorption, metabolism and tissue distribution of drugs.24,38 Genetic polymorphisms of CYP3A5 may potentially alter its proteins expression and function, and subsequently influence the clearance of any drugs metabolized by CYP3A5.38 The most studied SNP related to CYP3A5 functional variation is rs776746 (also known as CYP3A5*3 or CYP3A5*3C). Homozygous carriers of this SNP (*3/*3 or CC) lack functional CYP3A5 protein because of the frameshift mutation and truncation of the translated protein.39 Previous studies have confirmed that CYP3A5*3 is associated with drug metabolism, and CYP3A5*3/*3 carriers have decreased metabolism of nifedipine40 and tacrolimus,41 compared to CYP3A5*1/*1 and CYP3A5*1/*3 carriers. Patients with CYP3A5*3/*3 who are treated with tacrolimus may have an increased risk of nephrotoxicity as compared to patients without it.25,26 These findings suggest that the CYP3A5 *3/*3 carriers have low activity of CYP3A5 enzyme and decreased metabolism for certain drugs dependent on it, resulting in the appearance of an adverse reaction to the drug. Conforming to these findings, our study also indicates that there is an association between CYP3A5*3 and the occurrence of amlodipine-induced peripheral edema and that *3/*3 (CC) carriers have a higher risk of peripheral edema.

CYP3A5*1D (rs15524) is another frequently studied SNP in CYP3A5, which is in the 3-untranslated region (UTR). CYP3A5*1D is differentially distributed among populations ranging from 77.5% in Americans to 71.4% in East Asians, 40.6% in Africans, and even rarer in Europeans (less than 8%) (GnomAD).21 SNPs in UTRs may influence the stability of mRNA, thus affecting the expression and activity of the enzyme.42,43 According to a study identifying the potential SNPs related to miRNA, rs15524 may influence the hsa-miR-500a-5p that targets CYP3A5, affecting its expression.44 In addition, studies have found that CYP3A5*1D influences the pharmacokinetics of many drugs, including tacrolimus45 and carbamazepine.46 Therefore, rs15524 may act as a genetic marker and should be considered while studying or prescribing drugs metabolized by CYP3A5. Here, we find that the frequencies of rs15524 G allele and GA+GG genotypes in cases are lower than controls, which indicates that this SNP is associated with reduced risk of incidence of amlodipine-induced peripheral edema. In other words, CYP3A5*1D/*1D carriers may have an increased risk than others.

As for CYP3A5*1E (rs4646453), it is associated with a decreasing risk of amlodipine-induced peripheral edema. However, there is litter information about the function of the rs4646453. To some extent, this is possible because the SNP locates in the intron region. Studies have revealed that CYP3A5 rs4646453 is in LD with rs776746,47 and there is a strong LD between rs15524 and rs776746.39,46 Our findings are consistent with these studies and further promote that the presence of LDs with rs776746 may partly explain the role of rs15524 and rs4646453 in amlodipine-induced peripheral edema.

Given all these findings, there are some limitations in our present study that we would like to acknowledge. First, the population in our study is Chinese Han, and it is known that the frequency of mutations differs among ethnic groups. Hence, our results may apply well to Chinese population but are probably not suitable for populations of other ethnic groups. Second, we have performed a retrospective study and not a prospective one. The study is also limited by the smaller number of samples. The third limitation is not involving other genes contributing to amlodipine metabolism, such as CYP3A4. As we mentioned before, polymorphisms of CYP3A4 are rare in Chinese, and the evidence so far that CYP3A4 polymorphisms influence amlodipine metabolism is scarce. So, we took no account of them currently. But it would be better for future studies to include these related genes to better explain the connection between genetic factors and amlodipine-induced peripheral edema. Hence, more casecontrol studies with large number of polyethnic samples and involvement of diversified factors are necessary.

None the less, our study does investigate the novel relationship between the genetic variants and amlodipine-induced peripheral edema. In conclusion, we provide evidence that CYP3A5 polymorphisms are involved in the occurrence of amlodipine-induced peripheral edema, and the three genetic variants of CYP3A5 have the potential to serve as novel biomarkers for amlodipine-induced adverse reactions. Our findings thus provide new insights into amlodipine-induced peripheral edema and are of importance in developing and prescribing personalized and precise medicine for hypertension.

The raw data are available on reasonable request to the correspondence author Songnian Hu.

All the participants have signed the written informed consent, and approval was obtained from the Research Ethics Board of Beijing Chaoyang Hospital of Capital Medical University and the Research Ethics Board of the Second Hospital of Jilin University. This study has been conducted in accordance with the World Medical Association Declaration of Helsinki.

We thank all the participants included in this study.

All authors declare that they have no conflicts of interest for this work.

1. Flynn JT, Pasko DA. Calcium channel blockers: pharmacology and place in therapy of pediatric hypertension. Pediatr Nephrol. 2000;15(3):302316. doi:10.1007/s004670000480

2. Sica DA. Pharmacotherapy review: calcium channel blockers. J Clin Hypertens (Greenwich). 2006;8(1):5356. doi:10.1111/j.1524-6175.2005.04140.x

3. Fares H, DiNicolantonio JJ, OKeefe JH, Lavie CJ. Amlodipine in hypertension: a first-line agent with efficacy for improving blood pressure and patient outcomes. Open Heart. 2016;3(2):e000473. doi:10.1136/openhrt-2016-000473

4. Kala N, Babu S, Manjeu J, Aadivalavan A, Khan R. Allele-specific polymerase chain reaction for the detection of single nucleotide polymorphism in amlodipine-induced gingival enlargement. J Clin Pharm Ther. 2018;43(1):110113. doi:10.1111/jcpt.12587

5. Makani H, Bangalore S, Romero J, et al. Peripheral edema associated with calcium channel blockers: incidence and withdrawal ratea meta-analysis of randomized trials. J Hypertens. 2011;29(7):12701280. doi:10.1097/HJH.0b013e3283472643

6. Kes S, Caglar N, Canberk A, et al. Treatment of mild-to-moderate hypertension with calcium channel blockers: a multicentre comparison of once-daily nifedipine GITS with once-daily amlodipine. Curr Med Res Opin. 2003;19(3):226237. doi:10.1185/030079903125001677

7. The ClinCalc DrugStats Database. Amlodipine drug usage statistics, United States; 20082018. Available from: https://clincalc.com/DrugStats/Drugs/Amlodipine. Accessed December 9, 2020.

8. Epstein BJ, Roberts ME. Managing peripheral edema in patients with arterial hypertension. Am J Ther. 2009;16(6):543553. doi:10.1097/MJT.0b013e3181afbf9f

9. Messerli FH, Grossman E. Pedal edemanot all dihydropyridine calcium antagonists are created equal. Am J Hypertens. 2002;15(11):10191020. doi:10.1016/S0895-7061(02)03087-X

10. Bhatnagar V, Garcia EP, OConnor DT, et al. CYP3A4 and CYP3A5 polymorphisms and blood pressure response to amlodipine among African-American men and women with early hypertensive renal disease. Am J Nephrol. 2010;31(2):95103. doi:10.1159/000258688

11. Irvin MR, Lynch AI, Kabagambe EK, et al. Pharmacogenetic association of hypertension candidate genes with fasting glucose in the GenHAT study. J Hypertens. 2010;28(10):20762083. doi:10.1097/HJH.0b013e32833c7a4d

12. Sorensen IF, Vazquez AI, Irvin MR, et al. Pharmacogenetic effects of candidate gene complexes on stroke in the GenHAT study. Pharmacogenet Genomics. 2014;24(11):556563. doi:10.1097/FPC.0000000000000088

13. Kim KA, Park PW, Lee OJ, et al. Effect of CYP3A5*3 genotype on the pharmacokinetics and pharmacodynamics of amlodipine in healthy Korean subjects. Clin Pharmacol Ther. 2006;80(6):646656. doi:10.1016/j.clpt.2006.09.009

14. Lu Y, Zhong H, Tang Q, et al. Construction and verification of CYP3A5 gene polymorphisms using a Saccharomyces cerevisiae expression system to predict drug metabolism. Mol Med Rep. 2017;15(4):15931600. doi:10.3892/mmr.2017.6214

15. Eichelbaum M, Burk O. CYP3A genetics in drug metabolism. Nat Med. 2001;7(3):285287. doi:10.1038/85417

16. Zhu Y, Wang F, Li Q, et al. Amlodipine metabolism in human liver microsomes and roles of CYP3A4/5 in the dihydropyridine dehydrogenation. Drug Metab Dispos. 2014;42(2):245249. doi:10.1124/dmd.113.055400

17. Katoh M, Nakajima M, Shimada N, Yamazaki H, Yokoi T. Inhibition of human cytochrome P450 enzymes by 1,4-dihydropyridine calcium antagonists: prediction of in vivo drug-drug interactions. Eur J Clin Pharmacol. 2000;55(1112):843852. doi:10.1007/s002280050706

18. Dorofeeva MN, Shikh EV, Sizova ZM, et al. Antihypertensive effect of amlodipine in co-administration with omeprazole in patients with hypertension and acid-related disorders: cytochrome P450-associated aspects. Pharmgenomics Pers Med. 2019;12:329339. doi:10.2147/PGPM.S217725

19. Whirl-Carrillo M, McDonagh EM, Hebert JM, et al. Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2012;92(4):414417. doi:10.1038/clpt.2012.96

20. Gaedigk A, Ingelman-Sundberg M, Miller NA, et al. The pharmacogene variation (PharmVar) consortium: incorporation of the human cytochrome P450 (CYP) allele nomenclature database. Clin Pharmacol Ther. 2018;103(3):399401. doi:10.1002/cpt.910

21. Lek M, Karczewski KJ, Minikel EV, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285291. doi:10.1038/nature19057

22. Saiz-Rodriguez M, Almenara S, Navares-Gomez M, et al. Effect of the most relevant CYP3A4 and CYP3A5 polymorphisms on the pharmacokinetic parameters of 10 CYP3A substrates. Biomedicines. 2020;8(4):94. doi:10.3390/biomedicines8040094

23. Wojnowski L. Genetics of the variable expression of CYP3A in humans. Ther Drug Monit. 2004;26(2):192199. doi:10.1097/00007691-200404000-00019

24. Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet. 2001;27(4):383391. doi:10.1038/86882

25. Min SI, Kim SY, Ahn SH, et al. CYP3A5 *1 allele: impacts on early acute rejection and graft function in tacrolimus-based renal transplant recipients. Transplantation. 2010;90(12):13941400. doi:10.1097/TP.0b013e3181fa93a4

26. Satoh S, Saito M, Inoue T, et al. CYP3A5 *1 allele associated with tacrolimus trough concentrations but not subclinical acute rejection or chronic allograft nephropathy in Japanese renal transplant recipients. Eur J Clin Pharmacol. 2009;65(5):473481. doi:10.1007/s00228-008-0606-3

27. Zhang YP, Zuo XC, Huang ZJ, et al. CYP3A5 polymorphism, amlodipine and hypertension. J Hum Hypertens. 2014;28(3):145149. doi:10.1038/jhh.2013.67

28. Huang Y, Wen G, Lu Y, et al. CYP3A4*1G and CYP3A5*3 genetic polymorphisms alter the antihypertensive efficacy of amlodipine in patients with hypertension following renal transplantation. Int J Clin Pharmacol Ther. 2017;55(2):109118. doi:10.5414/CP202559

29. Uppsala Monitoring Centre. The use of the WHO-UMC system for standardised case causality assessment; 2018. Available from: https://www.who-umc.org/media/164200/who-umc-causality-assessment_new-logo.pdf. Accessed December 9, 2020.

30. Guo Z, Wang H, Tao J, et al. Development of multiple SNP markers panels affordable to breeders through genotyping by target sequencing (GBTS) in maize. Mol Breeding. 2019;39. doi:10.1007/s11032-019-0940-4

31. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):21142120. doi:10.1093/bioinformatics/btu170

32. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics. 2010;26(5):589595. doi:10.1093/bioinformatics/btp698

33. DePristo MA, Banks E, Poplin R, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 2011;43(5):491498. doi:10.1038/ng.806

34. Purcell S, Neale B, Todd-Brown K, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81(3):559575. doi:10.1086/519795

35. Barrett JC, Fry B, Maller J, Daly MJ. Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics. 2005;21(2):263265. doi:10.1093/bioinformatics/bth457

36. Mas S, Gasso P, Alvarez S, et al. Pharmacogenetic predictors of angiotensin-converting enzyme inhibitor-induced cough: the role of ACE, ABO, and BDKRB2 genes. Pharmacogenet Genomics. 2011;21(9):531538. doi:10.1097/FPC.0b013e328348c6db

37. Liu JE, Liu XY, Chen S, et al. SLCO1B1 521T > C polymorphism associated with rosuvastatin-induced myotoxicity in Chinese coronary artery disease patients: a nested case-control study. Eur J Clin Pharmacol. 2017;73(11):14091416. doi:10.1007/s00228-017-2318-z

38. Lamba JK, Lin YS, Schuetz EG, Thummel KE. Genetic contribution to variable human CYP3A-mediated metabolism. Adv Drug Deliv Rev. 2002;54(10):12711294. doi:10.1016/S0169-409X(02)00066-2

39. Xie HG, Wood AJ, Kim RB, Stein CM, Wilkinson GR. Genetic variability in CYP3A5 and its possible consequences. Pharmacogenomics. 2004;5(3):243272. doi:10.1517/phgs.5.3.243.29833

40. Haas DM, Quinney SK, Clay JM, et al. Nifedipine pharmacokinetics are influenced by CYP3A5 genotype when used as a preterm labor tocolytic. Am J Perinatol. 2013;30(4):275281. doi:10.1055/s-0032-1323590

41. Niioka T, Satoh S, Kagaya H, et al. Comparison of pharmacokinetics and pharmacogenetics of once- and twice-daily tacrolimus in the early stage after renal transplantation. Transplantation. 2012;94(10):10131019. doi:10.1097/TP.0b013e31826bc400

42. Mei J, Yan T, Huang Y, et al. A DAAM1 3-UTR SNP mutation regulates breast cancer metastasis through affecting miR-208a-5p-DAAM1-RhoA axis. Cancer Cell Int. 2019;19:55. doi:10.1186/s12935-019-0747-8

43. Saunders MA, Liang H, Li WH. Human polymorphism at microRNAs and microRNA target sites. Proc Natl Acad Sci U S A. 2007;104(9):33003305. doi:10.1073/pnas.0611347104

44. Wei R, Yang F, Urban TJ, et al. Impact of the interaction between 3-UTR SNPs and microRNA on the expression of human xenobiotic metabolism enzyme and transporter genes. Front Genet. 2012;3:248. doi:10.3389/fgene.2012.00248

45. Liu J, Ouyang Y, Chen D, et al. Donor and recipient P450 gene polymorphisms influence individual pharmacological effects of tacrolimus in Chinese liver transplantation patients. Int Immunopharmacol. 2018;57:1824. doi:10.1016/j.intimp.2018.02.005

46. Wang P, Yin T, Ma HY, et al. Effects of CYP3A4/5 and ABCB1 genetic polymorphisms on carbamazepine metabolism and transport in Chinese patients with epilepsy treated with carbamazepine in monotherapy and bitherapy. Epilepsy Res. 2015;117:5257. doi:10.1016/j.eplepsyres.2015.09.001

47. Hyland PL, Freedman ND, Hu N, et al. Genetic variants in sex hormone metabolic pathway genes and risk of esophageal squamous cell carcinoma. Carcinogenesis. 2013;34(5):10621068. doi:10.1093/carcin/bgt030

See original here:

[Full text] Association of CYP3A5 Gene Polymorphisms and Amlodipine-Induced Periph | PGPM - Dove Medical Press

With fertile land and water growing scarce, high-yield wheat and barley could help save millions from hunger – Genetic Literacy Project

By the time I am 60 years old in 2050, our global population will have increased to 9.7 billionpeople. Thats an additional ~2 billion human beings that will need to be fed. With over275 million hectares(680 million acres) of irrigated land globally, researchers note that to grow enough food for our projected population increase, we will need crops to produce more output on existing land.

Experts estimate that to provide for the 2050 population forecast, annual cereal production will need to rise by 50% to about3 billion tonnes. To do this, we must implement plant breeding technologies as one part of a comprehensive solution for global hunger.

The big news, as reported in the journalNature, is that researchers have sequenced new variations of genomes in barley and wheat. The international team includes scientists from the University of Adelaides Waite Research Institute, along with the10+ Genome Project, spearheaded by Curtis Pozniak, a professor at the University of Saskatchewan, Canada. Pozniak is in collaboration with the International Barley Pan Genome Sequencing Consortium, led by Nils Stein, professor at theLeibniz Institute of Plant Genetics and Crop Plant Research.

What does that mean for society today? Because barley and wheat are staple crops on a global level, scientists may have found a way to produce the high yield necessary to feed more mouths within our lifetime. And its not just a boon to cereal production; these discoveries bring us one step closer to unlocking the entire gene set, otherwise known aspan-genome,in wheat and barley, which has ramifications for all future research in plant genomics and cereal farming.

Heres how the research unfolded: Scientists conducting field tests in Chile found a way to increase the amount of protein (expansin) in the plants, which controls growth rate.The result: grains that were up to 12% larger than usual with higher yields as well. In the past, there had always been a tradeoff between grain size and number.

This is especially good news because wheat provides about 20% of the calories consumed by humans, and the current yield is increasing at only about 1% annually a far cry from the 50% needed to supply the population by 2050. Field results were a critical component, as they helped prove the effectiveness of these variations, by showing that the plants could perform under typical agricultural conditions. The teams of researchers are now working to make this research available to farmers and the greater food industry to help inform their decisions on crop production.

Currently, more than 800 million peopleworldwideare chronically hungry, and about 2 billion are nutritionally deficient. This is ahugepublic health concern. Whats more, fertile land and water supply are becoming scarcer, and production increases are falling off amplifying the need for more productive land.

Gene-editing technologies can only address global hunger and land and water availability if theyve gained consumer trust.GMOsand gene-editing are some of the most studied plant technologies. They also have the capacity to increase yield and lower chemical fertilizer and pesticide use, provide crops with better resilience to poor climate conditions, ward off pests, reduce post-harvest loss, and produce more nutrient-dense foods.

And yet, even with 30 years of research and countless commercial applications proving that gene-edited or GMO crops are as safe as conventionally grown crops, there is still hesitation from mainstream culture.

What if you were to wake up at 60, 70 or 80 years old, and instead of looking at flourishing families youre looking at 900 million people going hungry,landonce used for playgrounds now dedicated to growing food, and the population still multiplying? While this may seem like a stretch, if we dont accept plant breeding technology and realize its fundamental impact on food security, we may not meet increasing global food demands. More and more people will go hungry.

It seems like a luxury to even discuss consumer food production preference when people in developing countries are dying of starvation. With COVID running rampant, Africa is unable to make use of new plant technology, including GMOs, due to bottlenecks caused by the pandemic. This, asRuramiso Mashumba, an African smallholder farmer shared with us, is not a matter of preference but truly, a matter of life and death.

So, while our issue here in the United States remains a social challenge of widespread consumer adoption,developing countries are struggling with political barriers, preventing them from using lifesaving technology.

We hope to see more plant technologies such as this emerge and we hope that consumers do their research and come to understand the safety and vital nature of these developments.

What will 2050 look like? Your decisions today have an impact. Understanding the safety of new plant technologies, and the good they can do, is of chief importance to feeding generations to come.

Hayley N. Philip is a writer and researcher for Dirt to Dinner with a focus in health and nutrition.

A version of this article was originally posted at Dirt To Dinner and has been reposted here with permission. Dirt To Dinner can be found on Twitter @Dirt_To_Dinner

Read this article:

With fertile land and water growing scarce, high-yield wheat and barley could help save millions from hunger - Genetic Literacy Project

Study Identifies Breast Cancer Risk and Disease-Causing Mutations in Women Over 65 – Cancer Network

A study presented at the American Society of Human Genetics 2020 Virtual Meeting provided estimates of prevalence and breast cancer risks associated with pathogenic variants (PVs) in known breast cancer predisposition genes for the US population in women over the age of 65.1

Women with onset of breast cancer over age 65 typically do not qualify for genetic testing, however this study demonstrated that frequency of PVs and risk of breast cancer is not negligible in this patient population.

The median age of diagnosis for BC is 62 years, yet little is known about the frequency of pathogenic variants (PVs) in BC cancer predisposition genes in women over the age of 65, who represent a large percentage of women with BC, but often do not qualify for genetic testing, the investigators wrote in an abstract. The purpose of this study was to investigate the frequency of PVs in predisposition genes and to estimate residual risk of [breast cancer] in women over the age of 65.

In this study, research sequenced germline DNA from women over the age of 65 from population-based studies in the CARRIERS consortium to identify PVs in cancer predisposition genes using a custom multigene amplicon-based panel. In total, 26,707 women over the age of 65 were included in this study, with 13,762 (51.5%) cases and 12,945 (48.5%) controls. Notably, family history of breast cancer was present for 26% of cases and 18% of controls.

The frequency of PVs in 12 established breast cancer predisposition genes was found to be 3.18% for cases and 1.48% for controls. Genes with the highest frequencies observed includedATM(0.48%),BRCA1(0.18%),BRCA2 (0.49%),CHEK2(0.67%), andPALB2(0.23%).

This shows that a large number of women in this age category are predisposed to breast and other cancers, Nicholas Boddicker, PhD, a research associate at the Mayo Clinic, explained in a press release.2

Moreover, genes revealed to be associated with moderate risk of breast cancer included BRCA1 (OR, 3.37; 95% I, 1.68-7.51), BRCA2 (OR, 2.64; 95% CI, 1.78-4.02), PALB2 (OR, 3.09; 95% CI, 1.71-5.98), and CHEK2 (OR, 2.13; 95% CI, 1.53-3.02). However, ATM(OR, 1.38; 95% CI, 0.96-2.00) was not significantly associated with risk of breast cancer (P = .086).

Further, investigators found that the residual risk of breast cancer between the ages of 66 and 85 was 9.8% (95% CI, 6.8%-14.4%) forATM, 18.3% (95% CI, 9.5%-35.7%) forBRCA1, 18.6% (95% CI, 12.5%-28.0%) forBRCA2, 14.9% (95% CI, 10.8%-20.6%) forCHEK2, and 15.8% (95% CI, 9.0%-28.3%) forPALB2. For the general population, residual risk of breast cancer was 6.8%.

According to Boddicker, the frequency of disease-causing variants and the risks presented in this study can be used to inform cancer screening, risk management, and possibly clinical testing guidelines for women over 65.

In this study, women over 65 with no prior breast cancer found to have pathogenic variants in one of several genes would have remaining risk of breast cancer nearing 20% and could qualify for MRI surveillance in addition to mammography, he said. Without genetic testing, many of these women would not normally be screened this way.

Moving forward, the investigators indicated there are further areas which need to be explored, including combining other factors and measurements of risk with genetic testing to help better personalize risk estimates for women. In addition, more efforts to characterize these effects in other racial and ethnic groups are also still needed.

References:

1. Boddicker NJ, Hart S, Yadav S, et al. Residual breast cancer risk in genetically predisposed women diagnosed over age 65. Presented at the American Society of Human Genetics 2020 Virtual Meeting. Abstract #: 2412.

2. Breast Cancer Risk and Disease-Causing Mutations in Women Over Age 65 [news release]. Rockville, Maryland. Published October 26, 2020. Accessed November 17, 2020. https://www.ashg.org/publications-news/press-releases/breast-cancer-risk-disease-causing-mutations-women-over-age-65/

Visit link:

Study Identifies Breast Cancer Risk and Disease-Causing Mutations in Women Over 65 - Cancer Network

Scientists say West Africans originally migrated to East Africa – Quartz Africa

Africa is the cradle of humankind. All humans are descendants from this common pool of ancestors. Africa and its multitude of ethnolinguistic groups are therefore fundamental to learning more about humankind and our origins.

A human genome refers to the complete set of genetic information found in a human cell. We inherit our genomes from our parents. Studying the variations in different peoples genomes gives important clues to how genetic information influences peoples appearance and health. It can also tell us about our ancestry. To date, very few African individuals have been included in studies looking at genetic variation. Studying African genomes not only fills a gap in the current understanding of human genetic variation, but also reveals new insights into the history of African populations.

My colleagues and I, who are all members of the Human Heredity and Health (H3Africa) consortium, contributed to a landmark genetics study. This study focused on 426 individuals from 13 African countries. More than 50 different ethnolinguistic groups were represented in the studyone of the most diverse groups of Africans ever to be included in such an investigation. We sequenced the whole genome of each of these individualsthis means we could read every part of the genome to look for variation.

We were able to show that Zambia was most probably an intermediate site on the likely route of migration from West Africa to east and southern Africa.

This study contributes a major, new source of African genomic data, which showcases the complex and vast diversity of African genetic variation. And it will support research for decades to come.

Our findings have broad relevance, from learning more about African history and migration, to clinical research into the impact of specific variants on health outcomes.

One of the key outcomes was the discovery of more than 3 million new genetic variants. This is significant because we are learning more about human genetic diversity in general, and discovering more differences that could be linked to disease or traits in the future.

This study also adds details to what is known about the migration and expansion of groups across the continent. We were able to show that Zambia was most probably an intermediate site on the likely route of migration from West Africa to east and south Africa. Evidence supporting movement from east Africa to central Nigeria between 1,500 and 2,000 years ago was also revealed, through the identification of a substantial amount of east African ancestry in a central Nigerian ethnolinguistic group, the Berom.

The study also enabled us to reclassify certain variants that were previously suspected to cause disease. Variants that cause serious genetic diseases are often rare in the general population, mostly because their effect is so severe that a person with such a variant often does not reach adulthood. But we observed many of these variants at quite common levels in the studied populations. One wouldnt expect that these types of disease-causing variants would be this common in healthy adults. This finding helps to reclassify these variants for clinical interpretation.

Finally, we found a surprising number of regions with signatures of natural selection that have not been previously reported. Selection means that when individuals are exposed to environmental factors like a viral infection, or a drastic new dietary component, some gene variants may confer an added adaptive advantage to the humans that bear them in their genome.

Our best interpretation of these findings is that as humans across Africa were exposed to different environmentssometimes as a result of migrationthese variants were likely important to surviving in those new conditions. This has left an imprint on the genome and contributes to genomic diversity across the continent.

Our data has shown that we have not yet found all the variation in the human genome. There is more to learn by adding new, unstudied population groups. We know that less than a quarter of participants in genomics research are of non-European ancestry. Most available genetic data come from just three countriesthe UK (40%), the US (19%) and Iceland (12%).

It is essential to keep adding more genomic data from all global populationsincluding Africa. This will ensure that everyone can benefit from the advances in health that precision medicine offers. Precision medicine refers to the customization of healthcare to fit the individual. Including personal genetic information could radically change the nature and scope of healthcare options that would work best for that individual.

The Human Heredity and Health consortium is now in its eighth year of existence, and supports more than 51 diverse projects. These include studies focusing on diseases like diabetes, HIV, and tuberculosis. The reference data generated through our study are already being put to use by many of the consortiums studies.

Next, we are planning to take an even deeper look at the data to better understand what other types of genetic variation exist. We are also hoping to add further unstudied populations to grow and enrich this data set.

Building capacity for genomics research on the African continent is a key goal of Human Heredity and Health. An important aspect of this study is that it was driven and conducted by researchers and scientists from the African continent. Researchers from 24 institutions across Africa participated and led this investigation. This study showcases the availability of both infrastructure and skills for large-scale genomics research on the continent. It also highlights the prospect of future world-class research on this topic from Africa.

Zan Lombard, Principal Medical Scientist, Associate Professor, University of the Witwatersrand

This article is republished from The Conversation under a Creative Commons license. Read the original article.

The rest is here:

Scientists say West Africans originally migrated to East Africa - Quartz Africa

Major new study unveils complexity and vast diversity of Africa’s genetic variation – The Conversation CA

Africa is the cradle of humankind. All humans are descendants from this common pool of ancestors. Africa and its multitude of ethnolinguistic groups are therefore fundamental to learning more about humankind and our origins.

A human genome refers to the complete set of genetic information found in a human cell. We inherit our genomes from our parents. Studying the variations in different peoples genomes gives important clues to how genetic information influences peoples appearance and health. It can also tell us about our ancestry. To date, very few African individuals have been included in studies looking at genetic variation. Studying African genomes not only fills a gap in the current understanding of human genetic variation, but also reveals new insights into the history of African populations.

My colleagues and I, who are all members of the Human Heredity and Health (H3Africa) consortium, contributed to a landmark genetics study. This study focused on 426 individuals from 13 African countries. More than 50 different ethnolinguistic groups were represented in the study one of the most diverse groups of Africans ever to be included in such an investigation. We sequenced the whole genome of each of these individuals this means we could read every part of the genome to look for variation.

This study contributes a major, new source of African genomic data, which showcases the complex and vast diversity of African genetic variation. And it will support research for decades to come.

Our findings have broad relevance, from learning more about African history and migration, to clinical research into the impact of specific variants on health outcomes.

One of the key outcomes was the discovery of more than three million new genetic variants. This is significant because we are learning more about human genetic diversity in general, and discovering more differences that could be linked to disease or traits in the future.

This study also adds details to what is known about the migration and expansion of groups across the continent. We were able to show that Zambia was most probably an intermediate site on the likely route of migration from west Africa to east and south Africa. Evidence supporting movement from east Africa to central Nigeria between 1,500 and 2,000 years ago was also revealed, through the identification of a substantial amount of east African ancestry in a central Nigerian ethnolinguistic group, the Berom.

The study also enabled us to reclassify certain variants that were previously suspected to cause disease. Variants that cause serious genetic diseases are often rare in the general population, mostly because their effect is so severe that a person with such a variant often does not reach adulthood. But we observed many of these variants at quite common levels in the studied populations. One wouldnt expect that these types of disease-causing variants would be this common in healthy adults. This finding helps to reclassify these variants for clinical interpretation.

Finally, we found a surprising number of regions with signatures of natural selection that have not been previously reported. Selection means that when individuals are exposed to environmental factors like a viral infection, or a drastic new dietary component, some gene variants may confer an added adaptive advantage to the humans that bear them in their genome.

Our best interpretation of these findings is that as humans across Africa were exposed to different environments sometimes as a result of migration these variants were likely important to surviving in those new conditions. This has left an imprint on the genome and contributes to genomic diversity across the continent.

Our data has shown that we have not yet found all the variation in the human genome. There is more to learn by adding new, unstudied population groups. We know that less than a quarter of participants in genomics research are of non-European ancestry. Most available genetic data come from just three countries the UK (40%), the US (19%) and Iceland (12%).

It is essential to keep adding more genomic data from all global populations including Africa. This will ensure that everyone can benefit from the advances in health that precision medicine offers. Precision medicine refers to the customisation of healthcare to fit the individual. Including personal genetic information could radically change the nature and scope of healthcare options that would work best for that individual.

The Human Heredity and Health consortium is now in its eighth year of existence, and supports more than 51 diverse projects. These include studies focusing on diseases like diabetes, HIV and tuberculosis. The reference data generated through our study are already being put to use by many of the consortiums studies.

Read more: What we've learnt from building Africa's biggest genome library

Next, we are planning to take an even deeper look at the data to better understand what other types of genetic variation exist. We are also hoping to add further unstudied populations to grow and enrich this data set.

Building capacity for genomics research on the African continent is a key goal of Human Heredity and Health. An important aspect of this study is that it was driven and conducted by researchers and scientists from the African continent. Researchers from 24 institutions across Africa participated and led this investigation. This study showcases the availability of both infrastructure and skills for large-scale genomics research on the continent. It also highlights the prospect of future world-class research on this topic from Africa.

Link:

Major new study unveils complexity and vast diversity of Africa's genetic variation - The Conversation CA

Future Visioning the Role of CRISPR Gene Editing: Navigating Law and Ethics to Regenerate Health and Cure Disease – IPWatchdog.com

Despite the projected growth in market applications and abundant investment capital, there is a danger that legal and ethical concerns related to genetic research could put the brakes on gene editing technologies and product programs emanating therefrom.

As society adjusts to a new world of social distance and remote everything, rapid advancements in the digital, physical, and biological spheres are accelerating fundamental changes to the way we live, work, and relate to one another. What Klaus Schwab prophesized in his 2015 book, The Fourth Industrial Revolution, is playing out before our very eyes. Quantum computing power, a network architecture that is moving function closer to the edge of our interconnected devices, bandwidth speeds of 5G and beyond, natural language processing, artificial intelligence, and machine learning are all working together to accelerate innovation in fundamental ways. Given the global pandemic, in the biological sphere, government industrial policy drives the public sector to work hand-in-glove with private industry and academia to develop new therapies and vaccines to treat and prevent COVID-19 and other lethal diseases. This post will envision the future of gene editing technologies and the legal and ethical challenges that could imperil their mission of saving lives.

There are thousands of diseases occurring in humans, animals, and plants caused by aberrant DNA sequences. Traditional small molecule and biologic therapies have only had minimal success in treating many of these diseases because they mitigate symptoms while failing to address the underlying genetic causes. While human understanding of genetic diseases has increased tremendously since the mapping of the human genome in the late 1990s, our ability to treat them effectively has been limited by our historical inability to alter genetic sequences.

The science of gene editing was born in the 1990s, as scientists developed tools such as zinc-finger nucleases (ZFNs) and TALE nucleases (TALENs) to study the genome and attempt to alter sequences that caused disease. While these systems were an essential first step to demonstrate the potential of gene editing, their development was challenging in practice due to the complexity of engineering protein-DNA interactions.

Then, in 2011, Dr. Emmanuelle Charpentier, a French professor of microbiology, genetics, and biochemistry, and Jennifer Doudna, an American professor of biochemistry, pioneered a revolutionary new gene-editing technology called CRISPR/Cas9. Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and Cas9 stands for CRISPR-associated protein 9. In 2020, the revolutionary work of Drs. Charpentier and Doudna developing CRISPR/Cas9 were recognized with the Nobel Prize for Chemistry. The technology was also the source of a long-running and high-profile patent battle between two groups of scientsists.

CRISPR/Cas9 for gene editing came about from a naturally occurring viral defense mechanism in bacteria. The system is cheaper and easier to use than previous technologies. It delivers the Cas9 nuclease complexed with a synthetic guide RNA (gRNA) into a cell, cutting the cells genome at the desired location, allowing existing genes to be removed and new ones added to a living organisms genome. The technique is essential in biotechnology and medicine as it provides for the genomes to be edited in vivo with extremely high precision, efficiently, and with comparative ease. It can create new drugs, agricultural products, and genetically modified organisms or control pathogens and pests. More possibilities include the treatment of inherited genetic diseases and diseases arising from somatic mutations such as cancer. However, its use in human germline genetic modification is highly controversial.

The following diagram from CRISPR Therapeutics AG, a Swiss company, illustrates how it functions:

In the 1990s, nanotechnology and gene editing were necessary plot points for science fiction films. In 2020, developments like nano-sensors and CRISPR gene editing technology have moved these technologies directly into the mainstream, opening a new frontier of novel market applications. According to The Business Research Company, the global CRISPR technology market reached a value of nearly $700 million in 2019, is expected to more than double in 2020, and reach $6.7 billion by 2030. Market applications target all forms of life, from animals to plants to humans.

Gene editings primary market applications are for the treatment of genetically-defined diseases. CRISPR/Cas9 gene editing promises to enable the engineering of genomes of cell-based therapies and make them safer and available to a broader group of patients. Cell therapies have already begun to make a meaningful impact on specific diseases, and gene editing helps to accelerate that progress across diverse disease areas, including oncology and diabetes.

In the area of human therapy, millions of people worldwide suffer from genetic conditions. Gene-editing technologies like CRISPR-Cas9 have introduced a way to address the cause of debilitating illnesses like cystic fibrosis and create better interventions and therapies. They also have promising market applications for agriculture, food safety, supply, and distribution. For example, grocery retailers are even looking at how gene editing could impact the products they sell. Scientists have created gene-edited crops like non-browning mushrooms and mildew-resistant grapes experiments that are part of an effort to prevent spoilage, which could ultimately change the way food is sold.

Despite the inability to travel and conduct face-to-face meetings, attend industry conferences or conduct business other than remotely or with social distance, the investment markets for venture, growth, and private equity capital, as well as corporate R&D budgets, have remained buoyant through 2020 to date. Indeed, the third quarter of 2020 was the second strongest quarter ever for VC-backed companies, with 88 companies raising rounds worth $100 million or more according to the latest PwC/Moneytree report. Healthcare startups raised over $8 billion in the quarter in the United States alone. Gene-editing company Mammouth Biosciences raised a $45 million round of Series B capital in the second quarter of 2020. CRISPR Therapeutics AG raised more in the public markets in primary and secondary capital.

Bayer, Humboldt Fund and Leaps are co-leading a $65 million Series A round for Metagenomi, a biotech startup launched by UC Berkeley scientists. Metagenomi, which will be run by Berkeleys Brian Thomas, is developing a toolbox of CRISPR- and non-CRISPR-based gene-editing systems beyond the Cas9 protein. The goal is to apply machine learning to search through the genomes of these microorganisms, finding new nucleases that can be used in gene therapies. Other investors in the Series A include Sozo Ventures, Agent Capital, InCube Ventures and HOF Capital. Given the focus on new therapies and vaccines to treat the novel coronavirus, we expect continued wind in the sails for gene-editing companies, particularly those with strong product portfolios that leverage the technology.

Despite the projected growth in market applications and abundant investment capital, there is a danger that legal and ethical concerns related to genetic research could put the brakes on gene-editing technologies and product programs emanating therefrom. The possibility of off-target effects, lack of informed consent for germline therapy, and other ethical concerns could cause government regulators to put a stop on important research and development required to cure disease and regenerate human health.

Gene-editing companies can only make money by developing products that involve editing the human genome. The clinical and commercial success of these product candidates depends on public acceptance of gene-editing therapies for the treatment of human diseases. Public attitudes could be influenced by claims that gene editing is unsafe, unethical, or immoral. Consequently, products created through gene editing may not gain the acceptance of the government, the public, or the medical community. Adverse public reaction to gene therapy, in general, could result in greater government regulation and stricter labeling requirements of gene-editing products. Stakeholders in government, third-party payors, the medical community, and private industry must work to create standards that are both safe and comply with prevailing ethical norms.

The most significant danger to growth in gene-editing technologies lies in ethical concerns about their application to human embryos or the human germline. In 2016, a group of scientists edited the genome of human embryos to modify the gene for hemoglobin beta, the gene in which a mutation occurs in patients with the inherited blood disorder beta thalassemia. Although conducted in non-viable embryos, it shocked the public that scientists could be experimenting with human eggs, sperm, and embryos to alter human life at creation. Then, in 2018, a biophysics researcher in China created the first human genetically edited babies, twin girls, causing public outcry (and triggering government sanctioning of the researcher). In response, the World Health Organization established a committee to advise on the creation of standards for gene editing oversight and governance standards on a global basis.

Some influential non-governmental agencies have called for a moratorium on gene editing, particularly as applied to altering the creation or editing of human life. Other have set forth guidelines on how to use gene-editing technologies in therapeutic applications. In the United States, the National Institute of Health has stated that it will not fund gene-editing studies in human embryos. A U.S. statute called The Dickey-Wicker Amendment prohibits the use of federal funds for research projects that would create or destroy human life. Laws in the United Kingdom prohibit genetically modified embryos from being implanted into women. Still, embryos can be altered in research labs under license from the Human Fertilisation and Embryology Authority.

Regulations must keep pace with the change that CRISPR-Cas9 has brought to research labs worldwide. Developing international guidelines could be a step towards establishing cohesive national frameworks. The U.S. National Academy of Sciences recommended seven principles for the governance of human genome editing, including promoting well-being, transparency, due care, responsible science, respect for persons, fairness, and transnational co-operation. In the United Kingdom, a non-governmental organization formed in 1991 called The Nuffield Council has proposed two principles for the ethical acceptability of genome editing in the context of reproduction. First, the intervention intends to secure the welfare of the individual born due to such technology. Second, social justice and solidarity principles are upheld, and the intervention should not result in an intensifying of social divides or marginalizing of disadvantaged groups in society. In 2016, in application of the same, the Crick Institute in London was approved to use CRISPR-Cas9 in human embryos to study early development. In response to a cacophony of conflicting national frameworks, the International Summit on Human Gene Editing was formed in 2015 by NGOs in the United States, the United Kingdom and China, and is working to harmonize regulations global from both the ethical and safety perspectives. As CRISPR co-inventor Jennifer Doudna has written in a now infamous editorial in SCIENCE, stakeholders must engage in thoughtfully crafting regulations of the technology without stifling it.

The COVID-19 pandemic has forced us to rely more on new technologies to keep us healthy, adapt to working from home, and more. The pandemic makes us more reliant on innovative digital, biological, and physical solutions. It has created a united sense of urgency among the public and private industry (together with government and academia) to be more creative about using technology to regenerate health. With continued advances in computing power, network architecture, communications bandwidths, artificial intelligence, machine learning, and gene editing, society will undoubtedly find more cures for debilitating disease and succeed in regenerating human health. As science advances, it inevitably intersects with legal and ethical norms, both for individuals and civil society, and there are new externalities to consider. Legal and ethical norms will adapt, rebalancing the interests of each. The fourth industrial revolution is accelerating, and hopefully towards curing disease.

Continue reading here:

Future Visioning the Role of CRISPR Gene Editing: Navigating Law and Ethics to Regenerate Health and Cure Disease - IPWatchdog.com

Future Visioning The Role Of CRISPR Gene Editing: Navigating Law And Ethics To Regenerate Health And Cure Disease – Technology – United States -…

"Despite the projected growth in market applications andabundant investment capital, there is a danger that legal andethical concerns related to genetic research could put the brakeson gene editing technologies and product programs emanatingtherefrom."

There are thousands of diseases occurring in humans, animals,and plants caused by aberrant DNA sequences. Traditional smallmolecule and biologic therapies have only had minimal success intreating many of these diseases because they mitigate symptomswhile failing to address the underlying genetic causes. While humanunderstanding of genetic diseases has increased tremendously sincethe mapping of the human genome in the late 1990s, our ability totreat them effectively has been limited by our historical inabilityto alter genetic sequences.

The science of gene editing was born in the 1990s, as scientistsdeveloped tools such as zinc-finger nucleases (ZFNs) and TALEnucleases (TALENs) to study the genome and attempt to altersequences that caused disease. While these systems were anessential first step to demonstrate the potential of gene editing,their development was challenging in practice due to the complexityof engineering protein-DNA interactions.

Then, in 2011, Dr. Emmanuelle Charpentier, a French professor ofmicrobiology, genetics, and biochemistry, and Jennifer Doudna, anAmerican professor of biochemistry, pioneered a revolutionary newgene-editing technology called CRISPR/Cas9. Clustered Regularly InterspacedShort Palindromic Repeats (CRISPR) and Cas9 stands forCRISPR-associated protein 9. In 2020, the revolutionary work ofDrs. Charpentier and Doudna developing CRISPR/Cas9 were recognizedwith the Nobel Prize for Chemistry. The technology was also thesource of a long-running and high-profile patent battle between two groups ofscientsists.

CRISPR/Cas9 for gene editing came about from a naturallyoccurring viral defense mechanism in bacteria. The system ischeaper and easier to use than previous technologies. It deliversthe Cas9 nuclease complexed with a synthetic guide RNA (gRNA) intoa cell, cutting the 'cell's genome at the desired location,allowing existing genes to be removed and new ones added to aliving organism's genome. The technique is essential inbiotechnology and medicine as it provides for the genomes to beedited in vivo with extremely high precision, efficiently, and withcomparative ease. It can create new drugs, agricultural products,and genetically modified organisms or control pathogens and pests.More possibilities include the treatment of inherited geneticdiseases and diseases arising from somatic mutations such ascancer. However, its use in human germline genetic modification ishighly controversial.

The following diagram from CRISPR Therapeutics AG, a Swisscompany, illustrates how it functions:

In the 1990s, nanotechnology and gene editing were necessaryplot points for science fiction films. In 2020, developments likenano-sensors and CRISPR gene editing technology have moved thesetechnologies directly into the mainstream, opening a new frontierof novel market applications. According to The Business ResearchCompany, the global CRISPR technology market reached a value ofnearly $700 million in 2019, is expected to more than double in2020, and reach $6.7 billion by 2030. Market applications targetall forms of life, from animals to plants to humans.

Gene editing's primary market applications are for thetreatment of genetically-defined diseases. CRISPR/Cas9 gene editingpromises to enable the engineering of genomes of cell-basedtherapies and make them safer and available to a broader group ofpatients. Cell therapies have already begun to make a meaningfulimpact on specific diseases, and gene editing helps to acceleratethat progress across diverse disease areas, including oncology anddiabetes.

In the area of human therapy, millions of people worldwidesuffer from genetic conditions. Gene-editing technologies likeCRISPR-Cas9 have introduced a way to address the cause ofdebilitating illnesses like cystic fibrosis and create betterinterventions and therapies. They also have promising marketapplications for agriculture, food safety, supply, anddistribution. For example, grocery retailers are even looking athow gene editing could impact the products they sell. Scientistshave created gene-edited crops like non-browning mushrooms andmildew-resistant grapes - experiments that are part of an effort toprevent spoilage, which could ultimately change the way food issold.

Despite the inability to travel and conduct face-to-facemeetings, attend industry conferences or conduct business otherthan remotely or with social distance, the investment markets forventure, growth, and private equity capital, as well as corporateR&D budgets, have remained buoyant through 2020 to date.Indeed, the third quarter of 2020 was the second strongest quarterever for VC-backed companies, with 88 companies raising roundsworth $100 million or more according to the latest PwC/Moneytreereport. Healthcare startups raised over $8 billion in the quarterin the United States alone. Gene-editing company MammouthBiosciences raised a $45 million round of Series B capital in thesecond quarter of 2020. CRISPR Therapeutics AG raised more in thepublic markets in primary and secondary capital.

Bayer, Humboldt Fund and Leaps are co-leading a $65 million Series A round for Metagenomi, abiotech startup launched by UC Berkeley scientists. Metagenomi,which will be run by Berkeley's Brian Thomas, is developing atoolbox of CRISPR- and non-CRISPR-based gene-editing systems beyondthe Cas9 protein. The goal is to apply machine learning to searchthrough the genomes of these microorganisms, finding new nucleasesthat can be used in gene therapies. Other investors in the Series Ainclude Sozo Ventures, Agent Capital, InCube Ventures and HOFCapital. Given the focus on new therapies and vaccines to treat thenovel coronavirus, we expect continued wind in the sails forgene-editing companies, particularly those with strong productportfolios that leverage the technology.

Despite the projected growth in market applications and abundantinvestment capital, there is a danger that legal and ethicalconcerns related to genetic research could put the brakes ongene-editing technologies and product programs emanating therefrom.The possibility of off-target effects, lack of informed consent forgermline therapy, and other ethical concerns could cause governmentregulators to put a stop on important research and developmentrequired to cure disease and regenerate human health.

Gene-editing companies can only make money by developingproducts that involve editing the human genome. The clinical andcommercial success of these product candidates depends on publicacceptance of gene-editing therapies for the treatment of humandiseases. Public attitudes could be influenced by claims that geneediting is unsafe, unethical, or immoral. Consequently, productscreated through gene editing may not gain the acceptance of thegovernment, the public, or the medical community. Adverse publicreaction to gene therapy, in general, could result in greatergovernment regulation and stricter labeling requirements ofgene-editing products. Stakeholders in government, third-partypayors, the medical community, and private industry must work tocreate standards that are both safe and comply with prevailingethical norms.

The most significant danger to growth in gene-editingtechnologies lies in ethical concerns about their application tohuman embryos or the human germline. In 2016, a group of scientistsedited the genome of human embryos to modify the gene forhemoglobin beta, the gene in which a mutation occurs in patientswith the inherited blood disorder beta thalassemia. Althoughconducted in non-viable embryos, it shocked the public thatscientists could be experimenting with human eggs, sperm, andembryos to alter human life at creation. Then, in 2018, abiophysics researcher in China created the first human geneticallyedited babies, twin girls, causing public outcry (and triggeringgovernment sanctioning of the researcher). In response, the WorldHealth Organization established a committee to advise on thecreation of standards for gene editing oversight and governancestandards on a global basis.

Some influential non-governmental agencies have called for amoratorium on gene editing, particularly as applied to altering thecreation or editing of human life. Other have set forth guidelineson how to use gene-editing technologies in therapeuticapplications. In the United States, the National Institute ofHealth has stated that it will not fund gene-editing studies inhuman embryos. A U.S. statute called "The Dickey-WickerAmendment" prohibits the use of federal funds for researchprojects that would create or destroy human life. Laws in theUnited Kingdom prohibit genetically modified embryos from beingimplanted into women. Still, embryos can be altered in researchlabs under license from the Human Fertilisation and EmbryologyAuthority.

Regulations must keep pace with the change that CRISPR-Cas9 hasbrought to research labs worldwide. Developing international guidelines could be a steptowards establishing cohesive national frameworks. The U.S.National Academy of Sciences recommended seven principles for thegovernance of human genome editing, including promoting well-being,transparency, due care, responsible science, respect for persons,fairness, and transnational co-operation. In the United Kingdom, anon-governmental organization formed in 1991 called The NuffieldCouncil has proposed two principles for the ethical acceptabilityof genome editing in the context of reproduction. First, theintervention intends to secure the welfare of the individual borndue to such technology. Second, social justice and solidarityprinciples are upheld, and the intervention should not result in anintensifying of social divides or marginalizing of disadvantagedgroups in society. In 2016, in application of the same, the CrickInstitute in London was approved to use CRISPR-Cas9 in humanembryos to study early development. In response to a cacophony ofconflicting national frameworks, the International Summit on HumanGene Editing was formed in 2015 by NGOs in the United States, theUnited Kingdom and China, and is working to harmonize regulationsglobal from both the ethical and safety perspectives. As CRISPRco-inventor Jennifer Doudna has written in a now infamous editorialin SCIENCE, "stakeholders must engage in thoughtfullycrafting regulations of the technology without stiflingit."

The COVID-19 pandemic has forced us to rely more on newtechnologies to keep us healthy, adapt to working from home, andmore. The pandemic makes us more reliant on innovative digital,biological, and physical solutions. It has created a united senseof urgency among the public and private industry (together withgovernment and academia) to be more creative about using technologyto regenerate health. With continued advances in computing power, networkarchitecture, communications bandwidths, artificial intelligence,machine learning, and gene editing, society will undoubtedly findmore cures for debilitating disease and succeed in regeneratinghuman health. As science advances, it inevitably intersects withlegal and ethical norms, both for individuals and civil society,and there are new externalities to consider. Legal and ethicalnorms will adapt, rebalancing the interests of each. The fourthindustrial revolution is accelerating, and hopefully towards curingdisease.

Originally published by IPWatchdog.com, November 24,2020.

The content of this article is intended to provide a generalguide to the subject matter. Specialist advice should be soughtabout your specific circumstances.

See the rest here:

Future Visioning The Role Of CRISPR Gene Editing: Navigating Law And Ethics To Regenerate Health And Cure Disease - Technology - United States -...

Understanding the immunology of COVID-19 – SelectScience

Watch this on-demand webinar with Dr. Petter Brodin to learn about new insights into the immune response to SARS-CoV-2

A popular SelectScience webinar that provides important new insights into the immune system responses to SARS-CoV-2 infection is now available on demand. The studies, conducted by Dr. Petter Brodin's group at Karolinska Institute in Stockholm, took a systems-level approach to analyze both the cellular and protein components involved, using methodologies including mass cytometry, flow cytometry and high-multiplex proteomics.

A longitudinal study of severe COVID-19 patients identified distinct patterns of immune cell coregulation in four different stages of the disease and demonstrated a shared trajectory of immunological recovery that may provide future biomarkers of disease progression. In an investigation of multisystem inflammatory syndrome in children (MIS-C), a relatively rare complication of SARS-CoV-2 infection in children, important differences in inflammatory response were seen between MIS-C and severe COVID-19 in adults. Moreover, while some similarities were observed between inflammatory responses in MIS-C and Kawasaki disease, important differences were also apparent, particularly in the T cell subsets involved.

Read on for highlights from the live Q&A discussion with Dr. Brodin or register to watch the full webinar on demand >>

PB: If we start with MIS-C and Kawasaki disease, then Kawasaki disease occurs in young children 2-4 years of age in the wintertime. It's a viral infection of a different kind and the thing about Kawasaki disease is that children present with a rash and sometimes heart involvement. Initially, when this MIS-C presentation started to occur, people mistook them for Kawasaki Disease. However, we've now learned that Kawasaki disease and MIS-C often involve different populations of children. MIS-C typically involves older kids, children of teenage years and often much more severe in presentation than the typical Kawasaki disease. They often have abdominal involvement with vomiting, stomach ache, and so on, which is not typical in Kawasaki disease. There are clearly clinical differences between MIS-C and Kawasaki disease.

When it comes to acute COVID and these other post-infectious conditions, they are quite distinct. Acute COVID typically begins with a respiratory infection, coughing, fever, and then, later on, might develop into a hyperinflammatory disease. At that time, during the hyperinflammatory later phases of the infection, then there can be similarities between MIS-C and acute COVID, but that is sort of in the later stages.

PB: This has been probably the most important issue to sort out since we started to learn about this new virus because what's pretty evident is that for the majority of patients and people infected with SARS-CoV-2, the infection is rather mild. A lot of people have fevers and a cough, and so on. Young children more frequently are asymptomatic, but then in all age groups, some individuals develop very severe disease. Most commonly, of course, men more than women, and older people more than young people. There is a very big variation in presentation with patients with COVID-19.

We've learned quite a bit over these past 10 months, with 30,000 papers published. There has been an extraordinary development in understanding both the virus, but also the immune response to the virus. We know now that men suffer often more severe disease than women when it comes to acute COVID, are more likely to end up in intensive care units and more likely to die. We think that this is related to differences in the immune system between men and women because the infection rate, the likelihood of being infected, is not different in men and women, as far as we know.

What are those immune system differences? There have been a couple of reports, and we know from other people's work that, for example, vaccine responses differ between men and women. We also know that many autoimmune diseases, particularly diseases such as lupus, which involves interferon responses, are much more prominent in women than in men, more common in women than in men. A lot of evidence points towards differences in men and women with respect to innate, initial antiviral immune responses, both before COVID-19 but also now.

I think that is probably the best determinant we have to date, to explain the differences in COVID-19 severity. It has to do with the ability to mount a robust early immune response to the virus, involving type 1 interferons but also other factors probably.

PB: I think that relates to the MIS-C work, which was done in children. The question implies that there are genetic differences when it comes to the likelihood of getting the infection. That particular question we have not studied. It's very difficult to study whether people are resistant to a particular virus. Those people are very difficult to find. We are looking into genetic host factors that would explain both why some children develop MIS-C, while most children obviously don't, and also those factors, genetics and other things, that might determine why an individual develops severe COVID versus a milder COVID. There has been some progress made in that area by researchers such as Jean-Laurent Casanovas Lab at the Rockefeller Institute, Helen Su at the NIH, leading a large consortium called Human Genetic Effort. Their patients with rare immunodeficiencies involving viral sensing and interferon responses have been reported and those are individuals that are very rare, but they presented with life-threatening COVID-19. That's related in general to the infection, not specifically children.

PB: My guess is that it might involve prior coronaviruses, but that remains to be determined. I believe, and I think quite a few people believe, that the coronaviruses are so abundant that not only children would carry immunity to such viruses but probably also quite a few adults. Therefore, it does not entirely, in my opinion, explain why children are so able to manage this infection without severe disease in general. I think probably this points more to differences in the immune system. If you think about it from an evolutionary point of view, or life history point of view, children are experts at responding to new pathogens because the younger a child is, the less experience that child would have, and the more able the child must be to respond to a new infection. While adult people, and especially older people, they can get by quite well by relying on their memory responses of prior exposures. Typically, older people might be less equipped to respond to new pathogens. This can be explained by many different factors, the lower number of naive cells in the adaptive immune system, thymic involution, and then lack of production of naive T cells, and so on. I think there are many different pieces to this puzzle, and we only know a little bit of that at the moment.

Q: What do you see are the biggest advantages of combining the two platforms used in your studies?

PB: Sometimes people say that immune responses don't occur in the blood, and so there's no point in looking in the blood. Instead, all the relevant responses occur in tissues. Obviously, it's true that the blood is not the main siteof immune activity; it is definitely tissue, specific responses that we cannot see in the blood. Given the fact that we can sample the blood so easily and we can collect non-determinable samples, there is real potential in detecting important signals in the blood, even if the immune response is actually going on primarily in a distal tissue, like the lung.What do we do to study the blood in the best possible way? My group has reasoned that by looking at the various components of blood and the immune cells and proteins that make up the blood immune system, and the circulating immune system, and doing that in the most comprehensive way that we can, we believe this gives us a very strong potential, sort of an ability to actually look at the immune response in younger children, or over time in a patient. This combination of technologies, the Olink platform for plasma protein measurements which gives very reproducible signals with very low background signal, and then the mass cytometry assay, which gives us very broad coverage of the immune cell components, we think it's a very strong combination of features.

Watch this on-demand webinar to find out more in-depth insights about the immune responses of COVID-19>>

Read more here:

Understanding the immunology of COVID-19 - SelectScience

Scientists successfully implanted human genes into monkeys to increase their brain mass – Boing Boing

From Popular Mechanics:

Researchers from Germany's Max Planck Institute of Molecular Cell Biology and Genetics in Germany and Japan's Central Institute for Experimental Animals introduced a specifically human gene,ARHGAP11B, into the fetus of a common marmoset monkey, causing the enlargement of its brain's neocortex. The scientistsreported their findings in Science.

Those findings include a good indication that the ARHGAP11B gene played a key role in the development of the human neocortex, and, by extension, human evolution overall. (A 2015 study saw similar results with ARHGAP11B implanted in mice embryos, which "produced many more neural progenitor cells and could even undergo folding of their normally unfolded neocortex.")

In a press alert, the scientists behind the study were careful to specifically refer to their subjects as "transgenic non-human primates," which differentiates them from transgenic common marmosets, or humans (we're not given a cool classification for the sake of objectivity). They also included this ethical caveat:

We confined our analyses to marmoset fetuses, because we anticipated that the expression of this human-specific gene would affect the neocortex development in the marmoset. In light of potential unforeseeable consequences with regard to postnatal brain function, we considered it a prerequisite and mandatory from an ethical point of view to first determine the effects of ARHGAP11B on the development of fetal marmoset neocortex.

So, we're still a little ways away from a horrible interactive Planet of the Apes remix. That's good.

Somewhat related: I re-read Karen Joy Fowler's award-winning fiction-about-science novel We Are All Completely Beside Ourselves this summer, which I think every human being should read if they have not yet.

Uh-Oh, Scientists Used Human Genes to Make Monkey Brains Bigger [Tim Childers / Popular Mechanics]

Human-specific ARHGAP11B increases size and folding of primate neocortex in the fetal marmoset [Michael Heide, Christiane Haffner, Ayako Murayama, Yoko Kurotaki, Haruka Shinohara, Hideyuki Okano, Erika Sasaki, and Wieland B. Huttner]

Human brain size gene triggers bigger brain in monkeys [EurekaAlert!]

Image: Public Domain via Wikimedia Commons

Here is the original post:

Scientists successfully implanted human genes into monkeys to increase their brain mass - Boing Boing

Thirteen TUM researchers among the most cited worldwide – India Education Diary

Thirteen researchers from the Technical University of Munich (TUM) are among the most cited in their respective fields. This is shown by the current edition of the ranking Highly Cited Researchers.In their publications, scientists cite the most important papers previously published on the respective topic. The frequency of citations is therefore a good indicator of the quality and worldwide recognition of a research work and its authors. Using the Web of Science database, which records scientific publications from a wide range of disciplines, the US company Clarivate Analytics once a year identifies the Highly Cited Researchers.

The current edition of the ranking lists the scientists who were cited most frequently in their respective fields from 2009 to 2019. Researchers who were cited frequently in various fields are listed in the Cross-Field category. In total, the list includes about 6,400 persons in no particular order, including the following researchers from TUM:

Agricultural Sciences:

Chemistry:

Clinical Medicine:

Psychiatry and Psychology:

Environment and Ecology:

Cross-Field:

Go here to see the original:

Thirteen TUM researchers among the most cited worldwide - India Education Diary

Amgen To Present At The Evercore ISI Virtual HealthCONx Conference – GuruFocus.com

THOUSAND OAKS, Calif., Nov. 25, 2020 /PRNewswire/ -- Amgen (NASDAQ:AMGN) will present at the Evercore ISI Virtual HealthCONx Conference at 1:50 p.m. ET on Tuesday, Dec. 1, 2020. David M. Reese, M.D., executive vice president of Research and Development and Peter H. Griffith, executive vice president and chief financial officer at Amgen will present at the conference. Live audio of the presentation can be accessed from the Events Calendar on Amgen's website, http://www.amgen.com, under Investors. A replay of the webcast will also be available on Amgen's website for at least 90 days following the event.

About AmgenAmgen is committed to unlocking the potential of biology for patients suffering from serious illnesses by discovering, developing, manufacturing and delivering innovative human therapeutics. This approach begins by using tools like advanced human genetics to unravel the complexities of disease and understand the fundamentals of human biology.

Amgen focuses on areas of high unmet medical need and leverages its expertise to strive for solutions that improve health outcomes and dramatically improve people's lives. A biotechnology pioneer since 1980, Amgen has grown to be one of the world's leading independent biotechnology companies, has reached millions of patients around the world and is developing a pipeline of medicines with breakaway potential.

For more information, visit http://www.amgen.com and follow us on http://www.twitter.com/amgen.

CONTACT: Amgen, Thousand Oaks Megan Fox, 805-447-1423 (media)Trish Rowland, 805-447-5631 (media) Arvind Sood, 805-447-1060 (investors)

View original content to download multimedia:http://www.prnewswire.com/news-releases/amgen-to-present-at-the-evercore-isi-virtual-healthconx-conference-301180785.html

SOURCE Amgen

See the article here:

Amgen To Present At The Evercore ISI Virtual HealthCONx Conference - GuruFocus.com

Spaceflight does some weird things to astronauts bodies – MIT Technology Review

The Twins Study gave us a first sketch of the human bodys molecular responses to spaceflight, but these outlines needed to be filled in, says Christopher Mason, an associate professor of physiology and biophysics at Weill Cornell Medicine. The changes we saw needed more context and replication. We needed additional studies to map out the frequency of the changes we observed in other astronauts, and other organisms, that go into space, and also to see if the degree of change was similar for shorter missions.

That brings us to a new package of research that builds on the Twins Study, reanalyzing some of the original data with new techniques and providing comparisons with other astronauts. In a set of 19 studies published today in a slew of different journals (along with 10 preprints still under peer review), researchers like Mason (a senior author on 14 of the papers) studied the physiological, biochemical, and genetic changes that occurred in 56 astronauts (including Kelly) who have spent time in spacethe largest study of its kind ever conducted.

The new papers, which incorporate results from cell-profiling and gene-sequencing techniques that have become easier to run only recently, reveal that there are some features of spaceflight that consistently appear in humans, mice, and other animals when they go to space, says Mason. There appears to be a core mammalian set of adaptations and responses to the rigors of spaceflight.

The researchers highlightsix biological changesthat occur in all astronauts during spaceflight: oxidative stress (an excessive accumulation of free radicals in the bodys cells), DNA damage, dysfunction of the mitochondria, changes in gene regulation, alterations in the length of telomeres (the ends of chromosomes, which shorten with age), and changes in the gut microbiome.

Of these six changes, the biggest and most surprising one for scientists wasmitochondrial dysfunction. Mitochondria play a critical role in producing the chemical energy necessary to keep cellsand by extension, tissue and organsfunctional. Researchers found irregular mitochondrial performance in dozens of astronauts and were able to broadly characterize these changes thanks to new genomics and proteomics techniques. Afshin Beheshti, a bioinformatician at NASA and senior author of one study, says mitochondrial suppression helps explain how many of the problems astronauts experienced (like immune system deficiencies, disrupted circadian rhythm, and organ complications) are actually holistically related to each other, since they all rely on the same metabolic pathways.

When youre in space, its not just one are or organ thats affected, its the whole body thats affected, says Beheshti. "We started connecting the dots."

Other research homed in on problems observed at the genetic level. The Twins Study showed that Kellys telomeres got longer in space before shrinking back to normal or even shorter lengths soon after he returned to Earth. Telomeres are supposed to shorten with age, so lengthening makes little sense, and the Twins Study didnt provide enough data to prompt any real conclusions as to why it happened and what the effects were.

Susan Bailey, a Colorado State University expert on telomere research and a senior author for several of the papers, says the new research found that 10 other astronautsexperienced the same telomere lengthening Kelly did irrespective of mission durationas well as the same telomere shrinking once they came back to Earth.

Notably, one of the papers in the new package found that longer telomeres were also associated with climbers of Mount Everest. For Bailey and her colleagues, this suggests that telomere lengthening isaffected by oxidative stresssomething that climbers and astronauts both experience, and that disrupts proper telomere maintenance.

NASA

They are still trying to pinpoint how these pathways work and exactly what the consequences could be (its probably not a secret to longevity), but we now have a foundation to build onwe know what to look for and be aware of in future astronauts on long-duration [and deep space] exploration missions, she says.

Though some of the changes are unexpected, many are no cause for concern. What is amazing to me is how well we adapt to space, says Jeffrey Sutton, director of the Baylor College of Medicines Center for Space Medicine, who was not involved with the new research.Blood cell mutations decreased in Kelly while he was in space(a total surprise for Mason). Astronauts also exhibiteddecreased levels of biomarkers associated with agingandincreased levels of microRNAsthat regulate the vascular systems response to radiation damage and microgravity. One of the strangest findings was that astronauts gut microbiomes managed to bring space microbes found on the ISS back to Earth.

The studies individually and collectively are truly impressive, says Sutton. We have entered a new era of space biomedical research, where the approaches and tools of precision and translational medicine are being applied to advance our understanding of human adaptation to space.

Ultimately, however, the data highlights just how much havoc and stress even the healthiest bodies face during space missionswhich should have an impact on planning for longer missions. I dont think were close to sending untrained people into space for really long periods of time, says Scott Kelly.

Physiologically, he thinks its probably safe to send people to Mars and back. In the distant future, however, instead of going to Mars, were going to be going to the moons of Jupiter or Saturn, he says. Youre going to be in space for years. And at that point, well have to take a closer look at artificial gravity as a mitigation. I wouldnt want to be arriving on the surface of another planetary body and not be able to function. A year or so is workable. Several years probably isnt.

NASA

Were still far away from having to evaluate those kinds of risks. Mason and his colleagues suggest that there should be pharmacological strategies for reducing the impact of gravity on the bodies of returning astronauts.

Sutton believesprecision medicinecould play a huge role in tailoring those drugs to protect astronauts against the effects of microgravity and radiation. And the shared biological responses between astronauts and Mount Everest climbers suggest that some interventions used to protect extreme sports athletes from oxidative stress could be applied to astronauts too.

What we need is more dataand more populations to use for comparison. Mason, Bailey, and their colleagues are starting to collect cell and gene profiles of more astronauts, especially those going on future year-long missions. They also want to study people whove experienced other conditions similar in some way to spaceflight, such as radiotherapy patients, pilots, and flight attendants.

The more we know about the health effects of long-duration spaceflight, the better able we will be to help maintain the health and performance of astronauts during and after spaceflight, says Bailey. Such knowledge benefits those of us on Earth as wellwe are all concerned about getting older, and being in poor health.

This post has been updated with comments from Afshin Beheshti.

See the article here:

Spaceflight does some weird things to astronauts bodies - MIT Technology Review

Twist Bioscience Reports Fourth Quarter and Full Year Fiscal 2020 Financial Results – Business Wire

SOUTH SAN FRANCISCO, Calif.--(BUSINESS WIRE)--Twist Bioscience Corporation (NASDAQ: TWST), a company enabling customers to succeed through its offering of high-quality synthetic DNA using its silicon platform, today reported financial results and business highlights for the fourth quarter and full year fiscal 2020 ended September 30, 2020.

We ended our fiscal year with record revenue and orders against the backdrop of a global pandemic and significant uncertainty, said Emily M. Leproust, Ph.D., CEO and co-founder of Twist Bioscience. While we are proud of the new products we introduced to aid in the fight against COVID-19, which complemented our revenue, it was our core synthetic biology and next-generation sequencing (NGS) product lines that drove our overarching success.

We have aggressive plans for growth and expansion in fiscal 2021 and beyond, continuing to build our foundation for sustained success across synthetic biology, NGS, biopharma and DNA data storage.

FISCAL 2020 FINANCIAL RESULTS

FISCAL 2020 FOURTH QUARTER FINANCIAL RESULTS

"Over the last year, we delivered on revenue, orders, margin and product pipeline in a very challenging environment, commented Jim Thorburn, CFO of Twist. We have a strong balance sheet and momentum moving into fiscal 2021, and look forward to an exciting year ahead.

Fiscal Fourth Quarter 2020 and Recent Highlights

Fiscal 2021 Financial Guidance

The following statements are based on Twists current expectations for fiscal 2021. The following statements are forward-looking, and actual results could differ materially depending on market conditions and the factors set forth under Forward-Looking Statements below. Twist does not plan to update, nor does it undertake any obligation to update, this outlook in the future.

For the full fiscal year 2021, Twist provided the following financial guidance:

Fiscal 2021 First Quarter Financial Guidance

For the first quarter of fiscal 2021, Twist provided the following financial guidance:

COVID-19 Considerations

During the three months ended September 30, 2020, financial results of the Company were not significantly affected by the COVID-19 outbreak. However, the extent to which the COVID-19 outbreak affects Twists future financial results and operations is subject to a high degree of uncertainty and will depend on future developments, including the duration, spread and treatment of the outbreak domestically and abroad.

Conference Call Information

The company plans to hold a conference call and live audio webcast for analysts and investors today at 8:00 a.m. Eastern Time to discuss its financial results and provide an update on the companys business. The call can be accessed by dialing (866) 688-0947 (domestic) or (409) 217-8781 (international) and refer to the conference ID 2947139. A telephonic replay of the conference call will be available beginning approximately four hours after the call through November 30, 2020 and may be accessed by dialing (855) 859-2056 (domestic) or (404) 537-3406 (international). The replay conference ID is 2947139. The webcast replay will be available for two weeks.

Given the circumstances globally, it is recommended to dial-in at most 15 to 20 minutes prior to the call start to reduce waiting times. If a participant will be listen-only, they are encouraged to listen via the webcast on Twists investor page.

About Twist Bioscience Corporation

Twist Bioscience is a leading and rapidly growing synthetic biology and genomics company that has developed a disruptive DNA synthesis platform to industrialize the engineering of biology. The core of the platform is a proprietary technology that pioneers a new method of manufacturing synthetic DNA by writing DNA on a silicon chip. Twist is leveraging its unique technology to manufacture a broad range of synthetic DNA-based products, including synthetic genes, tools for next-generation sequencing (NGS) preparation, and antibody libraries for drug discovery and development. Twist is also pursuing longer-term opportunities in digital data storage in DNA and biologics drug discovery. Twist makes products for use across many industries including healthcare, industrial chemicals, agriculture and academic research.

Follow us on Twitter | Facebook | LinkedIn | YouTube

Investor Relations Information

Twist uses the investor relations section on its website as a means of complying with its disclosure obligations under Regulation FD. Accordingly, investors should monitor Twist's investor relations website in addition to following Twist's press releases, SEC filings, and public conference calls and webcasts.

Legal Notice Regarding Forward-Looking Statements

This press release contains forward-looking statements. All statements other than statements of historical facts contained herein are forward-looking statements reflecting the current beliefs and expectations of management made pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995, including, but not limited to, statements under the headings Fiscal 2021 Financial Guidance and Fiscal 2021 First Quarter Financial Guidance, future growth and expansion plans and Twists other expectations regarding its future financial performance, the impact of the COVID-19 pandemic on Twists future financial performance, and Twists ability to address the challenges posed by the business and economic impacts of COVID-19 pandemic, diversification and revenue growth across all product categories, introduction of new products, the use of our products by the healthcare sectors for the potential detection and treatment of diseases, and expectations regarding newly announced partnerships. Such forward-looking statements involve known and unknown risks, uncertainties, and other important factors that may cause Twists actual results, performance, or achievements to be materially different from any future results, performance, or achievements expressed or implied by the forward-looking statements. Such risks and uncertainties include, among others, the risks and uncertainties of the duration, extent and impact of the COVID-19 pandemic, including any reductions in demand for our products (or deferred or canceled orders) globally or in certain regions; the ability to attract new customers and retain and grow sales from existing customers; risks and uncertainties of rapidly changing technologies and extensive competition in synthetic biology could make the products Twist is developing obsolete or non-competitive; uncertainties of the retention of significant customers; supply chain and other disruptions caused by the COVID-19 pandemic or otherwise; risks of third party claims alleging infringement of patents and proprietary rights or seeking to invalidate Twists patents or proprietary rights; and the risk that Twists proprietary rights may be insufficient to protect its technologies. For a further description of the risks and uncertainties that could cause actual results to differ from those expressed in these forward-looking statements, as well as risks relating to Twists business in general, see Twists risk factors set forth in Twists Annual Report on Form 10-Q filed with the Securities and Exchange Commission (SEC) on August 12, 2020 and subsequent filings with the SEC. Additional risk factors may be described in the Risk Factors section of Twists Annual Report on Form 10-K to be filed with the SEC on or about November 25, 2020. In addition, many of the foregoing risks and uncertainties are, and could be, exacerbated by the COVID-19 pandemic and any worsening of global or regional business and economic environment as a result. We cannot at this time predict the extent of the impact of the COVID-19 pandemic and any resulting business or economic impact, but it could have a material adverse effect on our business, financial condition, results of operations and cash flows. Any forward-looking statements contained in this press release speak only as of the date hereof, and Twist Bioscience specifically disclaims any obligation to update any forward-looking statement, whether as a result of new information, future events or otherwise.

Twist Bioscience Corporation

Condensed Consolidated Statements of Operations

(Unaudited)

(in thousands, except per share data)

Three months ended September 30,

Twelve months ended September 30,

2020

2019

2020

2019

32,432

15,736

90,100

54,385

17,578

12,386

61,406

47,426

11,636

10,496

43,006

35,683

27,185

24,423

103,267

80,126

-

-

22,500

-

56,399

47,305

230,179

163,235

(23,967

)

(31,569

)

(140,079

)

(108,850

)

112

Read the original post:

Twist Bioscience Reports Fourth Quarter and Full Year Fiscal 2020 Financial Results - Business Wire

How to use precision medicine to personalize COVID-19 treatment according to the patients genes – TheStreet

Courtesy of Colin Allen, University of Pittsburgh and David Finegold, University of Pittsburgh

Tom Hanks and his wife, Rita Wilson, were among the earliest celebrities to catch the novel coronavirus. In an interview at the beginning of July, Hanks described how differently COVID-19 had affected each of them in March.

My wife lost her sense of taste and smell, she had severe nausea, she had a much higher fever than I did. I just had crippling body aches, he said. I was very fatigued all the time and I couldnt concentrate on anything for more than about 12 minutes.

Why does COVID-19 present such different symptoms or none at all in different people?

Preexisting conditions can only be part of the story. Hanks is over 60 and is a Type 2 diabetic, putting him in a high-risk group. Nevertheless, he survived his brush with the virus with no pneumonia and apparently without any long-lasting effects. Knowing what causes variation in different patients could help physicians tailor their treatments to individual patients an approach known as precision medicine.

In recent years, a gene-centric approach to precision medicine has been promoted as the future of medicine. It underlies the massive effort funded by the U.S. National Institutes of Health to collect over a million DNA samples under the All of Us initiative that began in 2015.

But the imagined future did not include COVID-19. In the rush to find a COVID-19 vaccine and effective therapies, precision medicine has been insignificant. Why is this? And what are its potential contributions?

We are a physician geneticist and a philosopher of science who began a discussion about the promise and potential pitfalls of precision medicine before the arrival of COVID-19. If precision medicine is the future of medicine, then its application to pandemics generally, and COVID-19 in particular, may yet prove to be highly significant. But its role so far has been limited. Precision medicine must consider more than just genetics. It requires an integrative omic approach that must collect information from multiple sources beyond just genes and at scales ranging from molecules to society.

Inherited diseases such as sickle cell anemia and Tay-Sachs disease follow a predictable pattern. But such direct genetic causes are perhaps the exception rather than the rule when it comes to health outcomes. Some heritable conditions for instance, psoriasis or the many forms of cancer depend on complex combinations of genes, environmental and social factors whose individual contributions to the disease are difficult to isolate. At best, the presence of certain genes constitutes a risk factor in a population but does not fully determine the outcome for an individual person carrying those genes.

The situation becomes yet more complicated for infectious diseases.

Viruses and bacteria have their own genomes that interact in complex ways with the cells in the people they infect. The genome of SARS-CoV-2 underlying COVID-19 has been extensively sequenced. Its mutations are identified and traced worldwide, helping epidemiologists understand the spread of the virus. However, the interactions between SARS-CoV-2 RNA and human DNA, and the effect on people of the viruss mutations, remain unknown.

Tom Hanks and his wife caught the virus and recovered in a matter of weeks. Presumably each was infected over the course of a few minutes of exposure to another infected person, involving cellular mechanisms that operate on a timescale of milliseconds.

But the drama of their illness, and that of the many victims with far worse outcomes, is taking place in the context of a global pandemic that has already lasted months and may continue for years. People will need to adopt changes in their behavior for weeks or months at a time.

What should a precision medicine approach be in a pandemic? The gene-centric vision of precision medicine encourages people to expect individualized gene-targeted fixes. But, genes, behavior and social groups interact over multiple timescales.

To capture all the data needed for such an approach is beyond possibility in the current crisis. A nuanced approach to the COVID-19 pandemic will depend heavily on imprecise population level public health interventions: mask-wearing, social distancing and working from home. Nevertheless, there is an opportunity to begin gathering the kinds of data that would allow for a more comprehensive precision medicine approach one that is fully aware of the complex interactions between genomes and social behavior.

With unlimited resources, a precision medicine approach would begin by analyzing the genomes of a large group of people already known to be exposed to SARS-CoV-2 yet asymptomatic, along with a similar-sized group with identified risk factors who are dying from the disease or are severely ill.

An early study of this kind by Precisionlife Ltd data mined genetic samples of 976 known COVID-19 cases. Of these, 68 high-risk genes were identified as risk factors for poor COVID-19 outcomes, with 17 of them deemed likely to be good targets for drug developments. But, as with all such statistical approaches, the full spectrum of causes underlying their association with the disease is not something the analysis provides. Other studies of this kind are appearing with increasing frequency, but there is no certainty in such fast-moving areas of science. Disentangling all the relevant factors is a process that will take months to years.

To date, precision medicine has proven better suited to inherited diseases and to diseases such as cancer, involving mutations acquired during a persons lifetime, than to infectious diseases. There are examples where susceptibility to infection can be caused by malfunction of unique genes such as the family of inherited immune disorders known as agammaglobulinemia, but these are few and far between.

Many physicians assume that most diseases involve multiple genes and are thus not amenable to a precision approach. In the absence of the kind of information needed for a multi-omic approach, there is a clear challenge and opportunity for precision medicine here: If it is to be the future of medicine, in order to complement and expand our existing knowledge and approaches, it needs to shift from its gene-centric origins toward a broader view that includes variables like proteins and metabolites. It must consider the relationships between genes and their physical manifestations on scales that range from days to decades, and from molecules to the global society.

Colin Allen, Distinguished Professor of History & Philosophy of Science, University of Pittsburgh and David Finegold, Professor, Department of Human Genetics, Pitt Public Health, University of Pittsburgh

This article is republished from The Conversation under a Creative Commons license. Read the original article.

Go here to see the original:

How to use precision medicine to personalize COVID-19 treatment according to the patients genes - TheStreet

Meiotic chromosome synapsis depends on multivalent SYCE1-SIX6OS1 interactions that are disrupted in cases of human infertility – Science Advances

Abstract

Meiotic reductional division depends on the synaptonemal complex (SC), a supramolecular protein assembly that mediates homologous chromosomes synapsis and promotes crossover formation. The mammalian SC has eight structural components, including SYCE1, the only central element protein with known causative mutations in human infertility. We combine mouse genetics, cellular, and biochemical studies to reveal that SYCE1 undergoes multivalent interactions with SC component SIX6OS1. The N terminus of SIX6OS1 binds and disrupts SYCE1s core dimeric structure to form a 1:1 complex, while their downstream sequences provide a distinct second interface. These interfaces are separately disrupted by SYCE1 mutations associated with nonobstructive azoospermia and premature ovarian failure (POF), respectively. Mice harboring SYCE1s POF mutation and a targeted deletion within SIX6OS1s N terminus are infertile with failure of chromosome synapsis. We conclude that both SYCE1-SIX6OS1 binding interfaces are essential for SC assembly, thus explaining how SYCE1s reported clinical mutations give rise to human infertility.

Meiotic cell division is defined by a unique and highly dynamic program of events that result in homologous chromosome synapsis, crossover (CO) formation, and subsequent homolog segregation into haploid germ cells (13). Homologous chromosome pairs are established through interhomolog recombination searches from up to 400 induced double-strand breaks (DSBs) per cell (4). Once established, local recombination-mediated alignments are converted into the single continuous synapsis of aligned homologous chromosomes through the zipper-like assembly of the synaptonemal complex (SC) (5). The SCs supramolecular protein structure mediates continuous 100-nm tethering between homologous chromosome axes and provides the necessary three-dimensional framework for crossover formation (2). Following SC disassembly, crossovers provide the sole physical links between homologs at metaphase I, so are essential for ensuring correct homolog segregation in addition to providing genetic diversity (2).

The SC has an iconic and highly conserved tripartite structure that has been observed across meiotically reproducing eukaryotes (6). This consists of lateral elements (LEs) that coat the two homologous chromosome axes and a midline central element (CE), with a series of transverse filaments that bind together these longitudinal electron-dense structures (Fig. 1A) (7). The protein components of the mammalian SC have been identified as transverse filaments protein SYCP1 (Synaptonemal complex protein 1) (8), CE proteins SYCE1, SYCE2, and SYCE3 (Synaptonemal complex central element proteins 1 to 3), SIX6OS1, and TEX12 (Testis-expressed protein 12) (912), and LE proteins SYCP2 and SYCP3 (13, 14). All transverse filament and CE components are essential for SC assembly, and their individual disruption leads to infertility owing to meiotic arrest with failure of DSB repair (10, 11, 1518). In contrast, disruption of LE components produces a sexual dimorphism of male infertility and female subfertility (19, 20), with SYCP3 deficiency in females promoting germ cell aneuploidy and embryonic death (21).

(A) Schematic of the SC demonstrating its tripartite structure of two chromosome-bound LEs and a midline CE. Synapsis is achieved through N-terminal head-to-head assembly of SYCP1 molecules, which are bound via their C termini to meiotic chromosomes. SYCP1 head-to-head assembly is structurally supported within the CE by SYCE3 (red), an SYCE1-SIX6OS1 complex (yellow), and SYCE2-TEX12 fibrous assemblies (green). (B) Human SYCE1 (top) and SIX6OS1 (bottom) sequence schematics indicating the location and consequence of infertility-associated mutations of SYCE1 and 1021 internal deletion of SIX6OS1, alongside the principal constructs used in this study. (C) SDSpolyacrylamide gel electrophoresis (SDS-PAGE) analysis of the purified recombinant proteins used in this study. The dominant degradation product of SYCE1POF is indicated by an asterisk; its identity was confirmed by the observed cleavage of degraded MBP- and His-SYCE1POF fusion proteins upon treatment with TEV protease (fig. S1, A and B), consistent with it representing C-terminal degradation down to SYCE1s structural core. Mw, weight-average molecular weight. (D) SEC-MALS analysis. SYCE1core (yellow), SYCE1POF (green), and full-length SYCE1 (violet) are dimeric species of 36, 48 (39 kDa for the degradation product), and 86 kDa, respectively (theoretical dimers: 37, 55, and 80 kDa). dRI, differential refractive index. Data for SYCE1core and full-length SYCE1 are reproduced from (28).

In recent years, a variety of cellular imaging, biochemical and structural biology approaches have begun to uncover the molecular structures, interactions, and mechanisms responsible for mammalian SC assembly. SYCP1 self-assembles into a supramolecular lattice that provides the underlying 100-nm synapsis between chromosome axes (22, 23), while SYCP3 assembles into regularly repeating filaments that support chromosomal looping (24, 25). The five CE proteins provide essential structural supports for the SYCP1 lattice that enable its continuous and cooperative extension along the entire chromosome length. In this capacity, CE proteins have been categorized as synaptic initiation factors (SYCE3, SYCE1, and SIX6OS1) and elongation factors (SYCE2 and TEX12), of which their disruption leads to complete loss of tripartite SC structure and failure of extension of short SC-like stretches, respectively (10, 11, 1618). Of synaptic initiation factors, SYCE3 forms dimers that undergo potentially limitless self-assembly (26, 27), SYCE1 forms antiparallel dimeric assemblies (28), and SIX6OS1 is an SYCE1-interacting protein of unknown structure (11). These likely act as short-range structural supports between SYCP1 molecules, possibly in transverse, longitudinal, and vertical orientations to stabilize a local three-dimensional SYCP1 lattice (22). In contrast, SYCE2 and TEX12 exist as a seemingly constitutive complex that undergoes self-assembly into fibers of many micrometers in length (29), which likely provide the long-range structural supports that stabilize continuous growth of the SYCP1 lattice along the entire chromosome axis (22).

Owing to the essential roles of meiotic recombination, synapsis, and chromosome dynamics in mammalian meiosis (15, 3034), their defects are associated with human infertility, recurrent miscarriage, and aneuploidies (35, 36). As genetic causes of infertility, they typically fall within the category of idiopathic cases, having no readily diagnosable and clinically resolvable cause. Within the 10 to 15% of couples who suffer from infertility, approximately 25% are idiopathic and of likely genetic origin, comprising 50 to 80% of cases of nonobstructive azoospermia (NOA) and premature ovarian failure (POF) (36, 37). While individual infertility mutations are inherently unlikely to become widespread in a population, they can be found within families, especially when consanguineous (38), and provide crucial insights into their common targets and the molecular mechanisms that they disrupt.

Within the SC, familial infertility mutations have been identified for SYCP3 and SYCE1 (36). All identified SYCP3 mutations are autosomal dominant and alter or delete its structural cores C terminus that mediates filamentous assembly, so likely sequester wild-type (WT) molecules into inactive complexes (24, 36). In contrast, the three identified SYCE1 mutations are autosomal recessive and were found in two familial cases of NOA and one of POF (36). The two NOA cases are splice-site mutations, c.197-2A>G and c.375-2A>G, which are predicted to result in a truncated product of amino acids 1 to 65 and an internal deletion of amino acids 126 to 155, respectively (39, 40). These remove or delete part of human SYCE1s structural core that is encoded by amino acids 25 to 179, so can be explained by disruption of its dimeric structure (Fig. 1B) (28, 36). The POF mutation c.613C>T generates a premature stop codon (p.Gln241*) to give a truncated product of amino acids 1 to 240, relative to the canonical 351amino acid isoform (Fig. 1B) (41). However, as this truncation lies outside SYCE1s structural core, the molecular mechanism that is disrupted, and thereby responsible for infertility, remains unknown.

Here, we combine mouse genetics and cellular and biochemical studies to reveal a multivalent interaction mode between SYCE1 and SIX6OS1 that is disrupted by infertility-associated mutations of SYCE1. We find that the SIX6OS1 N terminus binds and disrupts the core dimeric structure of SYCE1 (amino acids 25 to 179) to form a 1:1 complex as the first interface, and its downstream sequence binds to SYCE1 amino acids 177 to 305 as the second interface. SYCE1s infertility-associated mutations c.375-2A>G (NOA) and c.613C>T (POF) specifically disrupt the first and second interfaces, respectively. Mice harboring the SYCE1 POF mutation and a targeted deletion within SIX6OS1 (which disrupts the first interface) are infertile, with failure of SC assembly. We conclude that both SYCE1-SIX6OS1 binding interfaces are essential for SC assembly and meiotic division, thus explaining how human infertility results from the differential targeting of binding interfaces by SYCE1s reported clinical mutations.

The SYCE1 POF mutation c.613C>T encodes a premature stop codon (p.Gln241*) that is predicted to generate a truncated protein product of amino acids 1 to 240, relative to SYCE1s canonical 351amino acid isoform (Fig. 1B) (41). We previously demonstrated that an N-terminal structural core encoded by amino acids 25 to 179 (SYCE1core) forms an -helical antiparallel coiled-coil structure that mediates head-of-head dimerization of SYCE1 (28). As this core region is retained (Fig. 1B), we predicted that SYCE1s antiparallel dimeric structure would be maintained within the 1- to 240-amino acid truncated product of the POF mutation (SYCE1pof). To test this, we purified recombinant SYCE1pof, generating purified material that contained approximately equal quantities of the full protein and a degradation product of apparent size consistent with degradation to the C-terminal boundary of its structural core (Fig. 1C and fig. S1, A and B). Circular dichroism (CD) spectroscopy confirmed that SYCE1pof contains a proportion of -helical structure consistent with retention of the 25179 core structure (fig. S1C), and SYCE1pof and SYCE1core demonstrated identical melting temperatures (Tm) of 39C (fig. S1D). Furthermore, analysis by size exclusion chromatography multiangle light scattering (SEC-MALS) confirmed that the full and degraded proteins are homodimers of 48 and 39 kDa, respectively (Fig. 1D). We conclude that SYCE1pof retains the dimeric structure imposed by its core 25179 region, so its SC and meiotic defects must result from additional structural or functional roles of its deleted C terminus.

Having established its retention of core dimeric structure, we next sought to determine the structural and functional consequence of the SYCE1 POF mutation on the SC and meiotic division in vivo. We thus generated mice harboring mutations of Syce1 alleles to introduce stop codons at amino acid position 243, equivalent to the human p.Gln241* mutation (figs. S2 and S3). While heterozygotes (designated Syce1POF/WT) were fertile, both male and female homozygotes (designated Syce1POF/POF) were infertile, replicating the autosomal recessive pattern of the POF mutation in humans (41). In male mutant mice, we observed reduced testis size (63% smaller, n = 3 mice at 2 months of age; fig. S4A) and a zygotene-like arrest similar to that observed in the SYCE1 knockout (16). There was defective SC assembly, with reduced staining for SYCP1 (Fig. 2A) and SYCE3 (Fig. 2B) and no staining for SYCE1 (Fig. 2C), SIX6OS1 (Fig. 2D), and SYCE2-TEX12 (fig. S4, B and C). Analysis of SYCE1 expression in the testis of Syce1POF/POF mice confirmed the presence of Syce1 transcript and a protein product of the correct molecular weight, albeit at reduced levels in comparison with WT (fig. S4, D and E, and table S1A). The Syce1POF open reading frame achieved WT levels of protein expression in a heterologous 293T cellular system (fig. S4F). We next studied the kinetics of DSB repair. Meiotic DSBs are generated by the nuclease SPO11 and are then resected to form single-stranded DNA ends that invade into the homologous chromosome by the recombinases RAD51 (DNA repair protein RAD51 homolog 1) and DMC1 (Meiotic recombination protein DMC1/LIM15 homolog) (42). DSBs are labeled by the presence of phosphorylated H2AX (-H2AX) (43). The distribution of -H2AX in mutant spermatocytes was similar to that found in WT cells at early prophase I but show increased staining at zygotene-like arrest (Fig. 2E). The distributions of RAD51 and DMC1 were detected on aligned LEs (Fig. 2, F and G) but in absence of mismatch repair protein MLH1 (DNA mismatch repair protein Mlh1) (marker of crossing-overs) (Fig. 2H). Together, these data indicate generation of DSBs but with failure of their repair and CO formation in Syce1POF/POF. In female mutant mice, we observed no follicles in adult ovaries (fig. S5A), and embryonic oocytes demonstrated zygotene arrest with mostly unaligned chromosome axes, recapitulating the human POF syndrome. Analysis of the SC revealed similar defects, with reduction in SYCP1 and SYCE3 (Fig. 3, A and B) staining (though to a lesser extent than males), and absence of SYCE1, SIX6OS1 (Fig. 3, C and D), and SYCE2-TEX12 (fig. S5, B and C). The distribution of -H2AX, RAD51, and DMC1 labeling in zygotene-like mutant oocytes was also increased and lacked MLH1 foci (Fig. 3, E to H). Thus, the SYCE1 POF mutation leads to male and female infertility with phenotypes of failed DSB repair, synapsis, and lastly SC assembly, similar to those previously observed upon disruption of structural components of the SC CE (10, 11, 1618).

(A) Double immunolabeling of WT pachytene and Syce1POF/POF zygotene-like spermatocytes with SYCP3 (red) and SYCP1 (green). In Syce1POF/POF spermatocytes, AEs fail to synapse and show a weak staining of SYCP1 along the axial elements (AEs). a.u., arbitrary units. (B to D) Double immunolabeling of spermatocyte spreads with SYCP3 (red) and the CE proteins (green). Syce1POF/POF zygotene-like spermatocytes showed a highly reduced signal of SYCE3 (B) and the absence of (C) SYCE1 and (D) SIX6OS1 from the AEs. (E) Double immunolabeling of -H2AX (green) and SYCP3 (red) in spermatocyte spreads from WT and Syce1POF/POF mice. -H2AX staining was persistent in Syce1POF/POF zygotene-like spermatocytes, but was restricted to the sex body in WT pachytene cells. (F and G) Double immunofluorescence of (F) RAD51 or (G) DMC1 (green) and SYCP3 (red). Syce1POF/POF zygotene-like spermatocytes showed increased numbers of foci of RAD51 and DMC1 along the AEs in comparison with WT, indicating unrepaired DSBs. (H) Double immunolabeling of MLH1 (green) and SYCP3 (red) showing the absence of COs (MLH1) in arrested Syce1POF/POF spermatocytes. Fluorescence intensity levels (A, B, and E) and number of foci (F and G) from WT and zygotene-like arrested spermatocytes are quantified in the right-hand plots. Welchs t test analysis: ***P < 0.0001. Scale bars, 10 m.

(A) Double immunolabeling of oocyte spreads from WT and Syce1POF/POF mice with SYCP3 (red) and SYCP1 (green). Syce1POF/POF oocytes became arrested in a zygotene-like stage where AEs remain unsynapsed and unaligned, with reduced levels of SYCP1. (B to D) Double immunolabeling of oocyte spreads with SYCP3 (red) and the CE proteins (green). Syce1POF/POF zygotene-like oocytes showed reduced SYCE3 signal (B) and a complete absence of (C) SYCE1 and (D) SIX6OS1 from the AEs. IP, immunoprecipitation. (E) Double immunostaining of spread preparations of WT pachytene and Syce1POF/POF zygotene-like oocytes with -H2AX (green) and SYCP3 (red). In Syce1POF/POF oocytes, the levels of -H2AX increased and were more restricted to AEs in comparison with WT pachytene cells. (F to G) Double immunolabeling of (F) RAD51 or (G) DMC1 (green) and SYCP3 (red), showing higher numbers of foci in AEs from mutant oocytes. (H) Labeling of MLH1 (green) and SYCP3 (red). MLH1 foci are absent from the AEs of Syce1POF/POF oocytes. Fluorescence intensity levels (A, B, and E) and number of foci (F and G) from WT and Syce1POF/POF zygotene-like oocytes are quantified in the right-hand plots. Welchs t test analysis: ***P < 0.0001. Scale bars, 10 m.

As the Syce1POF/POF mouse strain indicated a clear structural defect in the SC, we wondered whether the POF mutation may disrupt the known interaction between SYCE1 and fellow SC CE components SIX6OS1 and SYCE3 (11). The expression of SYCE1 and SIX6OS1 in COS7 cells produced cytoplasmic signals that became colocalized in foci upon coexpression (95% cells; Fig. 4A and fig. S6), in keeping with our previous findings (11). SYCE1pof formed similar or slightly reduced numbers of foci that equally colocalized with SIX6OS1, indicating a retention of SIX6OS1 binding (89% cells; Fig. 4A). We further demonstrated a similar coimmunoprecipitation of SIX6OS1 by WT SYCE1 and SYCE1pof upon coexpression in human embryonic kidney (HEK) 293 cells (Fig. 4B). Thus, the SYCE1-SIX6OS1 interaction is retained in the SYCE1 POF mutation. Could other disrupted functions contribute to the effect of the POF mutation? The only other known SYCE1 interactor is SYCE3, which undergoes low-affinity binding, as determined by its dissociation during purification (fig. S7, A and B). In contrast with the WT protein, the expression of SYCE1pof (cytoplasmic foci) in COS7 cells failed to recruit SYCE3 (preferentially nuclear) to their cytoplasmic foci (colocalization between SYCE3 and SYCE1 was observed for 95% of cells expressing WT SYCE1 and 21% of cells expressing SYCE1pof; Fig. 4C and fig. S6). Similarly, SYCE1pof failed to coimmunoprecipitate SYCE3 upon coexpression in HEK293 cells (Fig. 4D). Thus, while the SYCE1-SIX6OS1 complex is retained, the low-affinity SYCE1-SYCE3 complex is largely abolished in the SYCE1 POF mutation.

(A) Mouse SIX6OS1 colocalized with mouse SYCE1 and SYCE1POF in a cytoplasmatic punctate pattern upon coexpression in COS7 cells; the percentage of cells exhibiting colocalization is shown in the right-hand plot (n = 100 cells). DAPI, 4,6-diamidino-2-phenylindole. (B) HEK293T cells were cotransfected with the indicated expression vectors. Protein complexes were immunoprecipitated with anti-Flag or antienhanced green fluorescent protein (EGFP) antibodies, or mouse immunoglobulin G (IgG) as a negative control, and were analyzed by immunoblotting with the indicated antibody. GFP-mSIX6OS1 coimmunoprecipitated with Flag-mSYCE1 and Flag-mSYCE1POF, suggesting that the POF mutation of SYCE1 alone is insufficient to block the interaction. (C) COS7 cells were transfected with mouse Syce3 in combination with mouse Syce1 or Syce1pof as indicated. SYCE1 colocalized with SYCE3 in its own cytoplasmatic punctate pattern, and colocalization was substantially diminished for SYCE1POF (n = 100 cells). (D) Immunoprecipitation of protein complexes from HEK293T-cotransfected cells with an anti-Myc or anti-EGFP antibody or mouse IgG. SYCE1 coimmunoprecipitated with SYCE3, and the interaction was disrupted for SYCE1 POF, suggesting that the C-terminal region of SYCE1 is required for its interaction with SYCE3. The untransfected lanes in (B) and (D) show the absence of all the proteins in total protein extracts from untransfected 293T cells. Scale bars, 20 m.

What is the molecular basis of SIX6OS1 binding by SYCE1? As this is retained in SYCE1pof, we reasoned that SIX6OS1 binding must be mediated by SYCE1s structural core. We screened SYCE1core against a library of SIX6OS1 constructs through bacterial coexpression and identified a robust interaction with amino acids 1 to 67 of SIX6OS1, herein referred to as SIX6OS1N (Figs. 1B and 5A). We were able to purify the SYCE1core-SIX6OS1N complex by reciprocal affinity chromatography, ion exchange, and size exclusion chromatography (Fig. 5B) and found it to be stable under all experimental conditions tested. We were further able to purify similar complexes for SYCE1pof (with the same degradation product as upon isolated expression) and full-length SYCE1 (Fig. 1C and fig. S1B), confirming that SIX6OS1 binding is retained by all constructs containing the 25179 core. CD analysis revealed similar -helical content for SYCE1-SIX6OS1N complexes as for their isolated SYCE1 proteins (fig. S1C). CD thermal denaturation revealed slightly increased cooperativity of unfolding and melting temperatures for SYCE1-SIX6OS1N complexes relative to their isolated SYCE1 proteins (increasing from 39 to 43C, 39 to 41C, and 38 to 40C for SYCE1core, SYCE1pof, and full length, respectively; Fig. 5C and fig. S1D). SEC-MALS analysis revealed that all three SYCE1-SIX6OS1N complexes are 1:1, with molecular weights of 27, 37, and 46 kDa, respectively (Fig. 5D and fig. S7C). Thus, the SYCE1core undergoes conformation change from an antiparallel homodimer to a 1:1 complex upon binding to SIX6OS1N (Fig. 5E).

(A) Amylose pulldown following coexpression of MBP-SIX6OS1 175, 167, 175 1021, and free MBP with His-SYCE1core. (B) SDS-PAGE of the copurification of the SYCE1core-SIX6OS1n complex. Ni-NTA, Ninitrilotriacetic acid. (C) CD thermal denaturation recording the CD helical signature at 222 nm between 5 and 95C, as % unfolded; estimated melting temperatures (Tm) are indicated. (D) SEC-MALS analysis. SYCE1core-SIX6OS1n (blue), SYCE1POF-SIX6OS1n (red) and full-length SYCE1-SIX6OS1n (black) are 1:1 complexes of 27, 37 (29 kDa for the degradation product complex), and 46 kDa, respectively (theoretical 1:1 to 27, 36, and 48 kDa), while MBP-SIX6OS1n (gray) is a 57-kDa monomer (theoretical, 53 kDa). SDS-PAGE of the SYCE1POF-SIX6OS1n sample is shown in Fig. 1C. (E) Schematic of the conformational change of the SYCE1core antiparallel dimer (yellow) into a 1:1 SYCE1core-SIX6OS1n complex (yellow-blue). (F and G) SEC-SAXS analysis. (F) SEC-SAXS P(r) interatomic distance distributions of SYCE1core-SIX6OS1n (blue), SYCE1POF-SIX6OS1n (red), and SYCE1core (yellow), revealing maximum dimensions (Dmax) of 138, 180, and 186 , respectively. Their cross-sectional radii (Rc) are indicated (fig. S7D). (G) SAXS ab initio models of SYCE1core-SIX6OS1n (blue) and SYCE1core (yellow); averaged models were generated from 20 independent DAMMIF runs. Data for SYCE1core and full-length SYCE1 are reproduced from (28).

We analyzed the conformation of the SYCE1core-SIX6OS1N complex by size exclusion chromatography small-angle x-ray scattering (SEC-SAXS; fig. S7, D and E). The SAXS real-space pair-distance P(r) distribution (the distribution of interatomic distances within a protein structure) demonstrates positive skew, indicating that SYCE1core-SIX6OS1N retains the rod-like structure of SYCE1core, but with a reduction in its molecular length from 186 to 138 (Fig. 5F). Furthermore, its cross-sectional radius is slightly increased from 9 to 11 (fig. S7F), suggesting an increase from a two- to four-helical coiled coil. These geometric changes are consistent with the SYCE1core-SIX6OS1N 1:1 complex forming a shorter but wider coiled coil than the isolated SYCE1core dimer, as indicated by their SAXS ab initio models (Fig. 5G). Furthermore, the SAXS P(r) distribution of SYCE1pof indicates a similar elongated structure but with an increased tail to a maximum dimension of 180 (Fig. 5F), consistent with it containing the same SYCE1core-SIX6OS1N structure with an extended and potentially unstructured C terminus to amino acid 240. We conclude that SYCE1core mediates a direct interaction with SIX6OS1N that imposes a conformational change to a 1:1 complex that adopts a shorter and wider coiled-coil conformation than the isolated SYCE1core antiparallel homodimer.

Does the SYCE1core-SIX6OS1N complex represent the sole means by which SYCE1 interacts with SIX6OS1? We were unable to obtain soluble biochemical complexes containing SIX6OS1 sequences beyond its N terminus and so used yeast two-hybrid (Y2H) to test SYCE1 binding by full-length SIX6OS1. Having confirmed direct binding of SYCE1core to full-length SIX6OS1, we used C-terminal truncation to dissect its minimal binding site to amino acids 1 to 75, in keeping with our biochemical findings, and identified an additional interaction between SYCE1 177305 and full-length SIX6OS1 (Fig. 6A).

(A) Y2H analysis of interactions between SYCE1 and SIX6OS1 in which positive reactions are indicated by the growth of blue colonies. These data are representative of three repeats. (B) Schematic of the SYCE1-SIX6OS1 interaction based on the Y2H data in (A), with the two binding sites highlighted in red and green. The SYCE1 POF mutation blocks the second binding interface between SYCE1 177305 and SIX6OS1 downstream sequence within region 1262, whereas the SIX6OS1 1021 deletion blocks the first binding interface between SYCE1core (25179) and SIX6OS1n (167). (C) COS7 cells were transfected with mouse Six6os1 1021 alone or in combination with mouse Syce1. SIX6OS1 1021 showed nuclear localization with some cytoplasmatic signal and colocalized in cytoplasmic foci with SYCE1; the percentage of cells exhibiting colocalization is shown. Scale bars, 20 m. (D) Coimmunoprecipitation of SIX6OS1 1021 and Flag-SYCE1 from cotransfected HEK293T cells using anti-Myc or anti-EGFP antibodies, or mouse IgG as a negative control. SIX6OS1 1021 coimmunoprecipitated SYCE1, indicating that the second SYCE1 binding interface is retained. The untransfected lanes confirm the absence of SIX6OS1 1021 and SYCE1 in total protein extracts of untransfected 293T cells.

To establish whether SYCE1core and 177305 bind to the same or distinct sites within SIX6OS1, we established an internal deletion of SIX6OS1 amino acids 10 to 21 (1021) that blocks formation of the SYCE1core-SIX6OS1N biochemical complex (Fig. 5A). SIX6OS1 122 did not interact with any SYCE1 construct (Fig. 6A), indicating that amino acids 10 to 21 are necessary but not sufficient for SYCE1core binding. While 1021 completely abrogated the Y2H interaction of full-length SIX6OS1 with SYCE1core (25179), it retained a robust interaction with SYCE1 177305, suggesting distinct SIX6OS1-binding sites (Fig. 6A). Furthermore, 1021 blocked the ability of SIX6OS1 1262 to interact with SYCE1core and SYCE1pof (amino acids 25 to 240) while retaining its binding to full-length and 25315 SYCE1 (Fig. 6A). Thus, SYCE1 undergoes multivalent interactions with SIX6OS1, with the first binding interface mediated by SYCE1core and SIX6OS1N (167), and the second interface mediated by SYCE1 177305 and downstream sequence within SIX6OS1 1262. Furthermore, the first and second binding interfaces are specifically disrupted by SIX6OS1 deletion 1021 and the SYCE1 POF mutation, respectively, and in both cases, an SYCE1-SIX6OS1 complex is retained through the unaffected alternative site (Fig. 6B).

Our biochemical and Y2H analyses concluded that SIX6OS1 1021 would disrupt the first SYCE1-SIX6OS1 binding interface while retaining complex formation through the second interface. In support of this, we found that SIX6OS1 1021 retained its ability to form intense colocalized foci with SYCE1 upon coexpression in COS7 cells (98% of the cells; Fig. 6C), similar to our previous observations for the SYCE1 POF mutation (Fig. 4A). Similarly, SIX6OS1 1021 retained its ability to coimmunoprecipitate SYCE1 upon coexpression in HEK293 cells (Fig. 6D). Thus, localization and coimmunoprecipitation data from heterologous systems support our Y2H findings that the second SYCE1-SIX6OS1 binding interface is retained in SIX6OS1 1021, mirroring the retention of only the second binding interface that is predicted for the 126155 deletion of the SYCE1 c.375-2A>G NOA mutation (40).

Having established that the severe phenotype of the SYCE1 POF mutation likely results from the disruption of the second SYCE1-SIX6OS1 binding interface and its interaction with SYCE3, we wondered whether a similar phenotype would result from the sole disruption of the first SYCE1-SIX6OS1 binding interface. To test this, we generated mice harboring mutations of Six6os1 alleles encoding internal in-frame deletions of amino acids 10 to 21 (equivalent numbering to the human protein) (fig. S8, A and B). While heterozygotes (designated Six6os11021/WT) were fertile, both male and female homozygotes (designated Six6os11021/1021) were infertile, similar to the SYCE1 POF mutation. In males, we observed reduced testis size (Fig. 7A) and a zygotene-like arrest similar to that observed in the Six6os1 and Syce1 knockouts (11, 16). The mutant spermatocytes were defective in synapsis and SC assembly, with reduced staining for SC proteins SYCP1 (Fig. 7B) and SYCE3 (Fig. 7C) and no staining for SYCE2-TEX12 (Fig. 7, F and G). In contrast with their complete absence in the SYCE1 POF mutation, we observed some residual staining for SYCE1 (Fig. 7D) and SIX6OS1 (Fig. 7E) even though the levels of transcription of Six6os11021 appeared to be increased in the mutant testis (fig. S9 and table S1B). We detected -H2AX (fig. S10A) and DMC1/RAD51 foci (fig. S10, B and C) on aligned axial elements but no MLH1 foci (fig. S10D), indicating the proper induction of DSBs with their failed repair and absence of COs. Thus, SIX6OS1 1021 leads to infertility with a phenotype of failed DSB repair and SC assembly, similar to the SYCE1 POF mutation and those reported for disruption of structural components of the CE (10, 11, 1618).

(A) Genetic deletion of amino acids 10 to 21 of SIX6OS1 led to a reduction of the testis size compared to the WT (mice of 3 months of age). (B) Double immunolabeling of WT pachytene and Six6os1/ zygotene-like spermatocytes with SYCP3 (red) and SYCP1 (green). AEs failed to synapse in Six6os1/ spermatocytes despite partial alignment, with reduced loading of SYCP1 along the AEs. (C to G) Double immunolabeling of spermatocyte spreads with SYCP3 (red) and all CE components (green). Six6os1/ zygotene-like spermatocytes showed reduced signals of (C) SYCE3, (D) SYCE1, and (E) SIX6OS1, and the absence of (F) SYCE2 and (G) TEX12 from the AEs. Scale bars, 10 m. Plots represent the quantification of fluorescence intensity levels in Six6os1/ zygotene-like and WT pachytene spermatocytes (B to E). Welchs t test analysis: ***P < 0.0001. (H) Schematic of how the SYCE1 antiparallel dimer (yellow) undergoes conformational change upon interaction with SIX6OS1 (blue) to form a possible 1:1 complex through consecutive binding interfaces mediated by SYCE1core-SIX6OS1n (site 1) and SYCE1 177305 and downstream sequence within SIX6OS1 1262 (site 2). The consequence of SYCE1 mutations associated with POF (c.613C>T) and NOA (c.375-2A>G) and SIX6OS1 1021 on the integrity, predicted stoichiometry, and conformation of resultant SYCE1-SIX6OS1 complexes is illustrated. Photo credit (A): Laura Gmez-H, Instituto de Biologa Celular y Molecular del Cncer.

Thus, we conclude that both first and second SYCE1-SIX6OS1 binding interfaces are essential for SC assembly and meiotic progression. Furthermore, these findings explain how the sole disruption of individual SYCE1-SIX6OS1 binding interfaces by SYCE1 NOA (c.375-2A>G) and POF (c.613C>T) mutations result in the reported familial cases of human infertility.

The structural and functional integrity of the SC is contingent on the structure and assembly of is constituent protein components. Here, we report that SC assembly depends on multivalent interactions between CE components SYCE1 and SIX6OS1 that are disrupted by infertility-associated mutations of SYCE1. The first binding interface is formed by the structural core of SYCE1 (SYCE1core; amino acids 25 to 179), which undergoes conformational change from an antiparallel homodimer to a 1:1 complex upon interaction with SIX6OS1s N terminus (SIX6OS1N; amino acids 1 to 67). The second binding interface is formed by downstream sequence within SIX6OS1 1262 interacting directly with SYCE1 177305. Through the generation of mice harboring an internal deletion of SIX6OS1s N terminus (1021) and the SYCE1 POF mutation (murine p.Gln243*), which specifically block the first and second binding interfaces, respectively, we find that integrity of both SYCE1-SIX6OS1 binding interfaces is essential for SC assembly and meiotic progression in vivo.

What is the structure of the SYCE1-SIX6OS1 complex? SEC-SAXS analysis revealed that the SYCE1core-SIX6OS1N 1:1 complex formed by the first binding interface has a length and cross-sectional radius of 138 and 11 , in comparison with 186 and 9 for the SYCE1core dimer. We previously reported a model for SYCE1core in which amino acids 52 to 179 form an antiparallel dimeric coiled coil containing a midline kink, with helices of amino acids 25 to 50 packing against this structural core (fig. S11A) (28). A maximum dimension of 138 for SYCE1core-SIX6OS1N suggests a coiled-coil length of approximately 92 amino acids, given a helical rise of 1.5 per amino acid (44). This could be explained by the 52179 region forming a helix-turn-helix structure through exaggeration of the kink to a full turn, which may combine with the helix formed by amino acids 25 to 50 and an helix from SIX6OS1N to form a four-helical coiled coil, consistent with its 11- cross-sectional radius (fig. S11B). The second binding interface between SYCE1 177305 and downstream sequence within SIX6OS1 1262 suggests that SYCE1core-SIX6OS1N likely adopts a parallel configuration to form a single SYCE1-SIX6OS1 1:1 complex of consecutive first and second binding interfaces (Fig. 7H).

Our analysis of the SYCE1-SIX6OS1 complex reveals how the three reported clinical mutations of SYCE1 differentially affect its interaction with SIX6OS1. The SYCE1 NOA mutation c.197-2A>G is predicted to result in a truncated product of amino acids 1 to 65 (39), which would disrupt both binding sites and so likely abrogates SYCE1-SIX6OS1 complex formation and thus works as a null mutation. The SYCE1 NOA mutation c.375-2A>G is predicted to result in internal deletion of amino acids 126 to 155 (40), which would disrupt the first binding interface while retaining the second binding interface, and so is likely to result in a conformationally altered 1:1 complex (Fig. 7H). In contrast, while 1021 SIX6OS1 similarly disrupts the first binding interface and retains the second binding interface, the SYCE1core remains unaffected and so is predicted to enable formation of a head-to-head 2:2 complex (Fig. 7H). The SYCE1 POF mutation c.613C>T generates a premature stop codon (p.Gln241*) that gives a truncated product of amino acids 1 to 240 (41), which we have demonstrated disrupts the second binding interface while retaining the first binding interface (Fig. 7H). Thus, the latter two infertility-associated mutations of SYCE1 specifically disrupt one SYCE1-SIX6OS1 interface while retaining the other, which combine with our mouse genetic studies to confirm that both interfaces are essential for the structural assembly of the SC and its function in meiosis.

What are the structural roles of SYCE1 and SYCE1-SIX6OS1 within the SC? Our analyses of Syce1POF/POF and Six6os11021/ 1021 mouse strains revealed similar phenotypes with retention of some SYCP1 and SYCE3 recruitment to chromosome axes, with absence or substantial reduction of SYCE1 and SIX6OS1, and lack of recruitment of SYCE2-TEX12. This pattern suggests a hierarchical model of SC assembly in which SYCE1 and SYCE1-SIX6OS1 lie downstream of SYCP1 and SYCE3, and upstream of SYCE2-TEX12 (Fig. 1A), which is consistent with existing knockout data (10, 11, 1518). The disruption of SYCE3 binding by the POF mutation suggests that its SYCE1-SIX6OS1 complex would be defective for SC recruitment, whereas the SYCE1-SYCE3 interaction, and hence SC recruitment, should be retained for the SYCE1-SIX6OS1 complex of the SIX6OS1 1021 internal deletion. This explains the greater severity of the CE loading defect in Syce1POF/POF than Six6os11021/ 1021, in which SYCE1 and SIX6OS1 staining was substantially reduced in the latter (83.77% of SYCE1 reduction, 0.12 0.02 in the Six6os11021/ 1021 versus 0.73 0.21 in the WT; 68.27% of SIX6OS1 reduction, 0.24 0.02 in the Six6os11021/ 1021 versus 0.76 0.15 in the WT) but completely absent in the former. Thus, we conclude that the first and second SYCE1-SIX6OS1 interfaces are essential for initiation of SC CE formation and likely function by stabilizing a local three-dimensional SC structure that mediates recruitment and self-assembly of SYCE2-TEX12 into fibers that mediate SC elongation along the chromosome axis. Furthermore, the SYCE1 POF mutation is likely worsened by its additional disruption of SYCE3 binding that removes the residual SYCE1-SIX6OS1 SC recruitment observed for the SIX6OS1 1021 internal deletion.

The existence of SYCE1core as an isolated antiparallel homodimer and in a 1:1 complex with SIX6OS1N raises the question of which is the biologically relevant conformation. It is important to highlight that the CD melting temperatures of SYCE1-SIX6OS1N complexes and isolated SYCE1 dimers are very similar, ranging between 38 and 41C. In contrast, highly stable SC components SYCE2-TEX12 and SYCP3 have melting temperatures of approximately 65C (24, 29). Thus, the relatively low melting temperatures of SYCE1-SIX6OS1N complexes and SYCE1 suggest that they may undergo conformational change in vivo, with each conformation functioning at different stages of meiosis and/or at different locations within the SC. Furthermore, our analysis of SYCE1 infertility-associated mutations and a targeted internal deletion of SIX6OS1 revealed at least four possible conformations of SYCE1 and SYCE1-SIX6OS1 complexes (Fig. 7H). Owing to the direct competition between SIX6OS1N binding and SYCE1core dimerization, these conformations could be achieved in the absence of mutations, through alterations of protein levels, local concentrations, allosteric changes, and posttranslational modifications. Hence, alterative conformations of SYCE1 and SYCE1-SIX6OS1 are intriguing candidates for local structural heterogeneity and the propagation of signals along the length of the SC, which could function in roles such as crossover enforcement and interference. Thus, as we progress toward a full molecular understanding of the mammalian SC, the multivalent SYCE1-SIX6OS1 interactions described herein provide tantalizing possibilities for a dynamic role of SC structure in its enigmatic functions in the mechanics of meiosis.

Human SYCE1 sequences were cloned into pHAT4 and pMAT11 vectors (45) for bacterial expression as His- and His-MBP (Maltose-Binding Protein) fusions with TEV (Tobacco Etch Virus) cleavage sites for fusion protein removal. Human SIX6OS1 was cloned into pRSF-Duet1 vectors with a TEV-cleavable N-terminal MBP fusion for coexpression with SYCE1. Proteins were expressed in BL21(DE3) Escherichia coli cells (Novagen), in 2xYT (Yeast Extract Tryptone) media. Expression was induced with addition of 0.5 mM isopropyl--d-thiogalactopyranoside with the cells incubated at 25C for 16 hours. Cells were lysed via sonication in 20 mM tris (pH 8.0) and 500 mM KCl, followed by centrifugation. Supernatant was applied to an amylose (New England Biolabs) affinity chromatography column, followed by HiTrap Q HP (GE Healthcare) anion exchange chromatography. His- and His-MBP/MBP tags were removed by incubation with TEV protease at 4C for 16 hours. The cleaved proteins were further purified by HiTrap Q HP (GE Healthcare) anion exchange chromatography followed by size exclusion chromatography (HiLoad 16/600 Superdex 200, GE Healthcare). The purified proteins/complexes were concentrated using Microsep Advance 3 kDa (PALL) centrifugal filter units and stored at 80C. Protein samples were analyzed for purity using Coomassie-stained SDSpolyacrylamide gel electrophoresis. Protein molecular weights and extinction coefficients were calculated using ExPASY ProtParam (http://web.expasy.org/protparam/) with protein concentrations determined using a Cary 60 ultraviolet (UV) spectrophotometer (Agilent).

Far-UV CD spectra were collected using a Jasco J-810 spectropolarimeter (Institute for Cell and Molecular Biosciences, Newcastle University). Wavelength scans were recorded at 4C from 260 to 185 nm at 0.2-nm intervals using a 0.2-mm path length quartz cuvette (Hellma). Protein samples were measured at 0.2 to 0.4 mg/ml in 10 mM Na2HPO4 (pH 7.5) and 150 mM NaF. Nine measurements were taken for each sample, averaged, buffer-corrected and converted to mean residue ellipticity (MRE) ([]) (1000 degcm2dmol1 per residue). Spectral deconvolutions were carried out using the Dichroweb CDSSTR algorithm (http://dichroweb.cryst.bbk.ac.uk). CD thermal melts were recorded at 222 nm between 5 and 95C, at intervals of 0.5C with a 1C/min ramping rate. Protein samples were measured at 0.1 mg/ml in 20 mM tris (pH 8.0), 150 mM KCl, and 2 mM dithiothreitol (DTT), using a 1-mm path length quartz cuvette (Hellma). The data were plotted as % unfolded after conversion to MRE ([]222,x-[]222,5)/([]222,95-[]222,5). The melting temperature was determined as the temperature at which the proteins are 50% unfolded.

SEC-MALS analysis of protein samples was carried out at concentrations of 5 to 20 mg/ml in 20 mM tris (pH 8.0), 150 mM KCl, and 2 mM DTT. Samples were loaded onto a Superdex 200 Increase 10/300 GL (GE Healthcare) column at 0.5 ml/min using an KTA Pure (GE Healthcare) system. The eluate was fed into a DAWN HELEOS II MALS detector (Wyatt Technology), followed by an Optilab T-rEX differential refractometer (Wyatt Technology). SEC-MALS data were collected and analyzed using ASTRA 6 software (Wyatt Technology), using Zimm plot extrapolation with a 0.185 ml/g dn/dc value to determine absolute protein molecular weights.

SEC-SAXS experiments were carried out on beamline B21 at the Diamond Light Source synchrotron facility (Oxfordshire, UK). Protein samples at concentrations 6 to 20 mg/ml were loaded onto a Superdex 200 Increase 10/300 GL size exclusion chromatography column (GE Healthcare) in 20 mM tris (pH 8.0) and 150 mM KCl at 0.5 ml/min using an Agilent 1200 high-performance liquid chromatography system. The eluate was fed through the experimental cell, with SAXS data recorded at 12.4 keV, in 3.0-s frames with a detector distance of 4.014 m. Sctter 3.0 (www.bioisis.net) was used to subtract and average the frames and carry out the Guinier analysis for the Rg and cross-sectional Rg (Rc). P(r) distributions were fitted using PRIMUS. Ab initio modeling was performed using DAMMIF (46) imposing P1 symmetry. Twenty independent runs were averaged. The PyMOL Molecular Graphics System, Version 2.0 Schrdinger, LLC was used to generate images of the SAXS ab initio models.

Constructs of human SYCE1 and SIX6OS1 were cloned into pGBKT7 and pGADT7 vectors (Clontech). Y2H experiments were carried out using the Matchmaker Gold system (Clontech) according to the manufacturers guidelines. Y187 yeast strain was transformed with pGBKT7 vectors, while the Y2H gold strain was transformed with pGADT7 vectors. Yeast transformations were carried out using standard lithium acetate methods. Mating of the two strains was carried out in 0.5 ml 2 YPDA (Yeast Peptone Dextrose Adenine) at 30C, 40 rpm, by mixing respective colonies. After 24 hours, the cultures were centrifuged and pellets were resuspended in 0.5xYPDA. These were then plated onto SD/Trp/Leu to select for mated colonies and onto SD/Trp/Leu/Ade/His with X--gal to detect mated colonies through ADE1, HIS3, and MEL1 reporter gene activation. Plates were then incubated for 5 days at 30C.

For developing the Syce1POF/POF model, Syce1single-guide RNA (sgRNA) 5-TGACTTCTTTCCACACTATC-3 targeting the intron 10 was predicted at https://eu.idtdna.com/site/order/designtool/index/CRISPR_SEQUENCE. This crRNA (CRISPR RNA), the tracrRNA (trans-activating CRISPR RNA), and the ssODN (single-stranded donor oligonucleotides) (5-GGGACTCTTCCTCCGAAGCCATGAGGCAGCTGCAGCAATGTAAGATGCAGGGTGGGGCAGGAGGAGGAAATGTCTAGCACTGACTTCTTTCCACACCCCCAGGTAGATCTTCAAGGATGAGAACAAGAAAGCTGAGG

AGTTCCTAGAGGCTGCAGCTCAGCAGCACGAGCAGCTGCAGCAGAGGTGCCACCAGCTACAG-3) were produced by chemical synthesis at IDT. The ssODN contains the mutated base (C>T, p.Gln241*) and the peptidyl-glycine -amidating monooxygenase (PAM) was mutated by substituting it by the human intron sequence (ACTATCAG > CCCCCAG). The crRNA and tracrRNA were annealed to obtain the mature sgRNA. A mixture containing the sgRNAs, recombinant Cas9 protein (IDT), and the ssODN [Cas9 (30 ng/l), annealed sgRNA (20 ng/l each), and ssODN (10 ng/l)] were microinjected into B6/CBA F2 zygotes (hybrids between strains C57BL/6 J and CBA/J) (47) at the Transgenic Facility of the University of Salamanca. Edited founders were identified by polymerase chain reaction (PCR) amplification (Taq polymerase, NZYTech) with primers flanking the exon 11 (primer F 5-CTGTAGAGAAACTGATGAAAGT-3 and R 5-CAAGAAAATATGAAGAGACATAC-3) producing an amplicon of 398 base pairs (bp) for both edited and WT alleles, and either direct sequenced or subcloned into pBlueScript (Stratagene) followed by Sanger sequencing, selecting the point mutation in the targeted region of Syce1 (fig. S2). For generating the Six6os11021/ 1021 (named as Six6os1/), Six6os1-crRNA G68 5-ATCTGTTTGTCAGTTTGGAC-3 and Six6os1-crRNA G75 5-TACTTATGTCTTGCTCATAC-3 targeting exons 2 and 3 and the ssODN (5-GTTCTTACTTTATGTATGCTCTTTTATATATGGCTTCTGAAAGTTTTATTATTTATTTTACACAGTGTCCAAGATGAATGATAATCTGTTTGTCAGTTTGCAAGACATAAGTATTAAAGAAGATACGATTCAAAGAATTAATAGTAAGTAGTTTTGCATGAAATAAATATTTTAGTCTTTTGGTTTTATCTTATATAGCA-3) were predicted, produced, and microinjected, as previously described. Edited founders with the predicted deletion were identified through PCR using primers flanking this region (primer F 5-CACTTACATTTTCCTTTTAAGAATGC-3 and R 5-CCCCTCTCATACATACAAGTTGC-3). The 1021 allele was 285 bp long versus 413 bp of the WT allele (fig. S8, A and B). The founders were crossed with WT C57BL/6 J to eliminate possible unwanted off-targets. Heterozygous mice were resequenced and crossed to give rise to edited homozygous. Genotyping was performed by analysis of the PCR products of genomic DNA with primers F and R.

Histology. For histological analysis of ovaries, after the necropsy of the mice, their ovaries were removed and fixed in formol 10%. They were processed into serial paraffin sections and stained with hematoxylin and eosin. The samples were analyzed using a microscope OLYMPUS BX51, and images were taken with a digital camera OLYMPUS DP70.

Immunocytology. Testes were detunicated and processed for spreading using a conventional dry-down technique. Oocytes from fetal ovaries (E17.5 embryos) were digested with collagenase, incubated in hypotonic buffer, disaggregated, and fixed in paraformaldehyde. Both meiocyte preparations were incubated with the following primary antibodies for immunofluorescence (IF): rabbit SIX6OS1 R1 and R2 [1:100, Proteogenix (11)], rabbit SYCE1 17406-1-AP (1:50, Proteintech), guinea pig SYCE1 (1:100, provided by C. Hg), mouse SYCP3 immunoglobulin G (IgG) sc-74569 (1:1000, Santa Cruz Biotechnology), rabbit serum SYCP3 K921 (1:500), rabbit SYCP1 IgG ab15090 (1:200), guinea pig SYCE3(1:20, provided by R. Benavente), guinea pig SYCE2 (1:100, provided by C. Hg), rabbit TEX12 IgG (1:100, provided by R. Benavente), rabbit anti--H2AX (ser139) IgG #07-164 (1:200) (Millipore), mouse MLH1 51-1327GR (1:5, BD Biosciences), rabbit RAD51 PC130 (1:50, Calbiochem), and rabbit DMC1 R1 and R2 (1:500, Proteogenix). The secondary antibodies used were goat Alexa 555 -mouse A-32727, goat Alexa 488 -mouse A-11001, donkey Alexa 555 -rabbit A-31572 (1:200, Thermo Fisher Scientific), goat Alexa 488Fab -rabbit 111-547-003, and donkey fluorescein isothiocyanate guinea pig 706-095-148 (1:100, Jackson Immunoresearch). Slides were visualized at room temperature using a microscope (Axioplan 2; Carl Zeiss Inc.) with 63 objectives with an aperture of 1.4 (Carl Zeiss Inc.). Images were taken with a digital camera (ORCA-ER; Hamamatsu) and processed with OPENLAB 4.0.3 and Photoshop (Adobe). Quantification of fluorescence signals was performed using ImageJ software.

HEK293T and COS7 cell lines were and obtained from the American Type Culture Collection (ATCC). Cell lines were tested for mycoplasma contamination (Mycoplasma PCR ELISA, Sigma-Aldrich). They were transfected with Jetpei (PolyPlus) according to the manufacturers protocol.

Immunoprecipitation and Western blotting. HEK293T cells were transiently transfected, and whole-cell extracts were prepared and cleared with protein G Sepharose beads (GE Healthcare) for 1 hour. The antibody was added for 2 hours, and immunocomplexes were isolated by adsorption to protein G Sepharose beads overnight. After washing, the proteins were eluted from the beads with 2 SDS gel-loading buffer 100 mM tris-HCl (pH 7), 4% SDS, 0.2% bromophenol blue, 200 mM -mercaptoethanol, and 20% glycerol and loaded onto reducing polyacrylamide SDS gels. The proteins were detected by Western blotting with the indicated antibodies. Immunoprecipitations were performed using mouse -Flag IgG (5 g; F1804, Sigma-Aldrich), mouse green fluorescent protein (-GFP) IgG (4 g; CSB-MA000051M0m, Cusabio), mouse -Myc obtained from hybridoma cell myc-1-9E10.2 ATCC (4 g), and ChromPure mouse IgG (5 g/1 mg protein; 015-000-003). Primary antibodies used for Western blotting were rabbit -Flag IgG (1:2000; F7425 Sigma-Aldrich), goat -GFP IgG (sc-5385, Santa Cruz Biotechnology) (1:3000), and rabbit -Myc Tag IgG (1:3000; #06-549, Millipore). Secondary horseradish peroxidaseconjugated -mouse (715-035-150, Jackson ImmunoResearch), -rabbit (711-035-152, Jackson ImmunoResearch), or -goat (705-035-147, Jackson ImmunoResearch) antibodies were used at 1:5000 dilution. Antibodies were detected by using Immobilon Western Chemiluminescent HRP Substrate from Millipore. Both Syce1POF and Six6os1 1021 complementary DNAs (cDNAs) used for IF and coimmunoprecipitation experiments were reverse transcription PCRamplified (the primers used for it were Syce1 S 5-GAGCAGTATGGCCACCAGACC-3 and Syce AS 5-GAGGAGGGTATTAGGTCCTGC-3; Six6os1 S 5-AGTGTCCAAGATGAATGATAATCTG-3 and Six6os1 AS 5-GTTCAAAAATAATAACTCAAAAAAAC-3) from total RNA extracted from Syce1POF/POF and Six6os11021/ 1021 mice, respectively. PCR-amplified fragments were cloned in pcDNA3-based mammalian expression vectors with different tags (enhanced GFP or Flag) and verified by Sanger sequencing.

Total RNA was isolated from testis of WT and mutant mice. To analyze the expression of Syce1 and Six6os1 mRNAs, equal amounts of cDNA were synthesized using SuperScript II Reverse Transcriptase (Invitrogen, Life Technologies) and Oligo (dT). Quantitative PCR (qPCR) was performed using FastStart Universal SYBR Green Master Mix (ROX) (Roche) and specific forward and reverse primers: qSYCE1_F 5-GGACATGGTGAAAAAGTTGCAG-3 and qSYCE1_R 5-CAGTTCCTTCTGCAGGTTGTC-3 for Syce1, and qSIX6OS1_F 5-GCTGAATGTGGAGATAAAGAG-3 and qSIX6OS1_R 5-AGGAGTTTCAGGAGTTTGAGG-3 for Six6os1. All qPCR reactions were performed at 95C for 10 min and then 40 cycles of 95C for 15 s and 62C for 1 min on the iQ5 Thermal Cycler (Bio-Rad). -Actin was amplified as a housekeeping gene with the primers q-actin_F 5-GGCACCACACCTTCTACAATG-3and q-actin_R 5-GTGGTGGTGAAGCTGTAGCC-3.

Statistics. To compare counts between genotypes, we used the Welchs t test (unequal variances t test), which was appropriate as the count data were not highly skewed (i.e., were reasonably approximated by a normal distribution) and, in most cases, showed unequal variance. We applied a two-sided test in all the cases. Asterisks denote statistical significance: *P < 0.01, **P < 0.001, and ***P < 0.0001.

Acknowledgments: We thank Diamond Light Source and the staff of beamline B21 (proposals sm15836, sm21777, and sm23510). We thank H. Waller for assistance with CD data collection. Funding: O.R.D. is a Sir Henry Dale Fellow jointly funded by the Wellcome Trust and Royal Society (grant number 104158/Z/14/Z). This work was supported by MINECO (BFU2017-89408-R) and by Junta de Castilla y Leon (CSI239P18). F.S.-S., L.G.-H., and N.F.-M. are supported by European Social Fund/JCyLe grants (EDU/556/2019, EDU/1083/2013, and EDU/310/2015). CIC-IBMCC is supported by the Programa de Apoyo a Planes Estratgicos de Investigacin de Estructuras de Investigacin de Excelencia cofunded by the CastillaLen autonomous government and the European Regional Development Fund (CLC201701). The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. Ethics statement: Mice were housed in a temperature-controlled facility (specific pathogen free) using individually ventilated cages, standard diet, and a 12-hour light/dark cycle, according to European Union laws at the Servicio de Experimentacion Animal, SEA. Mouse protocols were approved by the Ethics Committee for Animal Experimentation of the University of Salamanca (USAL). We made every effort to minimize suffering and to improve animal welfare. Blinded experiments were not possible since the phenotype was obvious between WT and mutant mice for all of the experimental procedures used. No randomization methods were applied since the animals were not divided in groups or treatments. The minimum size used for each analysis was two animals per genotype. Author contributions: F.S.-S., L.G.-H., O.M.D., N.F.-M., C.G.-P., M.S.-M., and O.R.D. performed experiments. O.R.D. and A.M.P. designed experiments, analyzed data, and wrote the manuscript. A.M.P., E.L., and O.R.D. supervised and designed the work. Competing financial interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

Originally posted here:

Meiotic chromosome synapsis depends on multivalent SYCE1-SIX6OS1 interactions that are disrupted in cases of human infertility - Science Advances

Thought to Be Extinct, New Guinea’s Singing Dogs Found Alive in the Wild – Smithsonian Magazine

The New Guinea singing dog was thought to be extinct in the wild, but new genetic research suggests their distinctive howl still echoes in the highlands of the Oceanic islands, reports James Gorman for the New York Times.

Not seen in the wild by scientists since the 1970s, conservation biologists thought the only New Guinea singing dogs left on Earth were the 200 to 300 captive animals residing in zoos and sanctuaries, reports Michael Price for Science.

But anecdotal reports and a pair of photographs suggested a similarly tan-colored, medium-sized wild dog was roaming the mountainous terrain near a gold mine on Papua, the western, Indonesian half of the large island north of Australia.

The locals called them the highland wild dog, James McIntyre, president of the New Guinea Highland Wild Dog Foundation and co-author of the paper, tells the Times. The New Guinea singing dog was the name developed by caucasians. Because I didnt know what they were, I just called them the highland wild dogs.

To find out what these highland wild dogs really were, McIntyre trekked into the rugged terrain surrounding the Grasberg Mine, one of the worlds largest gold and copper mines, in 2016. The expedition produced 149 photographs of 15 individual dogs as well as an array of fecal samples. Per Science, if one were trying to cast a pooch for the role of the New Guinea singing dog, the wild dogs looked, acted and sounded the part.

However, the fecal samples didnt have enough genetic material for a proper analysis, so in 2018 the researchers returned and collected blood samples from three of the animals, according to the paper which was published this week in the journal Proceedings of the National Academy of Sciences.

These three samples were used to sequence the highland wild dogs genomes. The researchers then compared the dogs nuclear DNA with 16 captive New Guinea singing dogs, 25 dingoes as well as more than 1,000 individuals from 161 additional breeds.

The genetic analysis suggests that these highland wild dogs are in fact part of a wild population of New Guinea singing dogs. Crucially, the newly revealed wild population is much more genetically diverse than captive singing dogs, which descended from just eight individuals and are severely inbred, reports Katie Hunt for CNN.

Assuming these highland wild dogs are the original New Guinea singing dogs, so to speak, that really gives us a fantastic opportunity for conservation biology, Elaine Ostrander, a geneticist at the U.S. National Human Genome Research Institute and co-author of the study, tells Ed Cara of Gizmodo. Itll give us a chance to reintroduce the original genetics of these dogs into this conservation population.

Both the wild dogs and the captive singing dogs are close relatives of the Australian dingo, and relatively distant relatives of domestic dogs. The New Guinea singing dogs closest domesticated relatives are East Asian breeds including the chow chow, Akita and shiba inu, according to Science. This connection suggests that the singing dog may have split off from the ancestors of these Asian breeds some 3,500 years ago when humans and a few canine companions migrated to Oceania, per the Times.

Ostrander tells the Times that the genome of the wild singing dogs offers researchers a missing piece that we didnt really have before, that may help clarify the history of dog domestication.

Follow this link:

Thought to Be Extinct, New Guinea's Singing Dogs Found Alive in the Wild - Smithsonian Magazine

Humans have had mystery DNA for 300,000 yearsand now we might finally know what it is – SYFY WIRE

Humans getting into interspecies dating? Not on this planet oh wait, that already happened.While there was really no such thing as dating hundreds of thousands of years ago, when it was more of a find your mate and dont get eaten sort of thing, there is evidence that Homo sapiens interbred with other proto-human species in the distant past.

Neanderthals were one of those species (and also the butt of endless caveman jokes). Denisovans were another. Geneticist and computer scientistAdam Siepel developed an algorithm to trace human genetics and recently published a study in PLOS Genetics. He andhis research teamhave now found that these groups gave us more of their DNA than we thought, and thatsome of us have genes from a mysterious ancestral hominid, possibly Homo erectus. There was only one way for that to happen. But why did Homo sapiens end up taking over while Neanderthals and Denisovans eventually vanished?

"That is a great matter of speculation among both archaelogists and geneticistscould be disease, conquest, out-competition for scarce resources, or perhaps the modern humans simply absorbed them.There is little hard evidence,"Siepeltold SYFY WIRE."But the one thing that we can see, as geneticists, is that these Neanderthal and Denisovan populations had relatively low levels of genetic diversity, suggesting they may have been prone to genetic diseases and/or particularly susceptible to infectious diseases."

Hybridization of a species results in introgression, or the genetic exchange which occurs in interbreeding species. Humans migrated out of Africa to Eurasia about 50,000 years ago and interbred with the Neanderthal population there. This is the migration and subsequent genetic merging that is the most recognized example of such a phenomenon. What Siepel found, using an updated ancestral recombination graph (ARG) algorithm called ARGweaver-D, is that they were already headed elsewhere much earlier, around 200,000-300,000 years ago. The algorithm also revealed interbreeding between mystery super-archaic ancestors with both Neanderthals and Denisovans before either of those species interbred with ancient Homo sapiens.

Homo erectus is the most likely ancient relative of humans to be that ancestor. Now extinct except for fragments of DNA that show up in some modern human samples, these proto-humans were the first Homo sapiensrelatives that showed body proportions similar to what you see when you look in the mirror. Unlike earlier hominids, the arms and legs of Homo erectus had evolved to be shorter than its torso. They were also the first hominids believed to have migrated out of Africa. This strengthens the case for interbreeding with Denisovans and Neanderthals, especially Denisovans.

That has to make you wonder. If you've ever taken a commercial DNA test and your results came back with a small percentage labeled "unknown", could that be a connection to the mystery ancestor?

"Some of the commercial tests specifically look for Neanderthal ancestry, but yes, it is possible that superarchaic ancestry, or ancestry from a highly divergent branch of Neanderthals or Denisovans, would be labeled 'unknown'by a commercial test," Siepel said.

The most common genetic transfers happened between Neanderthals and Denisovans, Neanderthals and ancient Homo sapiens, super-archaic ancestors and ancient Homo sapiens who stayed in Africa, and super-archaic ancestors and Denisovans. Alleles, or alternate versions of genes, shared by Denisovans and the mystery ancestor support super-archaic DNA making its way into the modern gene pool when that species interbred with Denisovans. Unfortunately, so did mutations.

"It appears that Neanderthals and Denisovansintroduced deleterious mutations into modern human populations when they interbred with them," Siepel explained."Many of these mutations gradually fadedover time, but some undoubtedly persist.Interestingly, however, we could not find clear evidence of the reverse effectof modern humans introducing deleterious mutations into Neanderthals through this interbreeding.It is possible, though, that we do not yet have enough sensitivity to detect this phenomenon."

Even with an advanced algorithm, it still proved more difficult to identify when and where super-archaic human ancestors interbred with Denisovans than it was to find the same information about Neanderthal or Denisovan interbreeding with Homo sapiens. This is probably because no sequence exists for the genes of the super-archaic ancestor yet, and also because they have been broken over and over again by recombining with the genes of ancient humans and the other two hominid groups so many times. Will we ever really know who the super-archaic ghosts of our ancestors were?

"That is the big question we are all wondering about," said Siepel."It is conceivable that it could be done, at least in principle,if very well-preserved remains were recovered from permafrost or from a cave that is well protected from the environment. But I do not know of any promising leads at present."

So dont be offended if someone calls you a Neanderthal. Just tell them science says you probably are to some extent, and so are they.

Here is the original post:

Humans have had mystery DNA for 300,000 yearsand now we might finally know what it is - SYFY WIRE

The Secret to a Long, Healthy Life Is in the Genes of the Oldest Humans Alive – Singularity Hub

The first time I heard nematode worms can teach us something about human longevity, I balked at the idea. How the hell can a worm with an average lifespan of only 15 days have much in common with a human who lives decades?

The answer is in their genesespecially those that encode for basic life functions, such as metabolism. Thanks to the lowly C. elegans worm, weve uncovered genes and molecular pathways, such as insulin-like growth factor 1 (IGF-1) signaling that extends healthy longevity in yeast, flies, and mice (and maybe us). Too nerdy? Those pathways also inspired massive scientific and popular interest in metformin, hormones, intermittent fasting, and even the ketogenic diet. To restate: worms have inspired the search for our own fountain of youth.

Still, thats just one success story. How relevant, exactly, are those genes for humans? Were rather a freak of nature. Our aging process extends for years, during which we experience a slew of age-related disorders. Diabetes. Heart disease. Dementia. Surprisingly, many of these dont ever occur in worms and other animals. Something is obviously amiss.

In this months Nature Metabolism, a global team of scientists argued that its high time we turn from worm to human. The key to human longevity, they say, lies in the genes of centenarians. These individuals not only live over 100 years, they also rarely suffer from common age-related diseases. That is, theyre healthy up to their last minute. If evolution was a scientist, then centenarians, and the rest of us, are two experimental groups in action.

Nature has already given us a genetic blueprint for healthy longevity. We just need to decode it.

Long-lived individuals, through their very existence, have established the physiological feasibility of living beyond the ninth decade in relatively good health and ending life without a period of protracted illness, the authors wrote. From this rare but valuable population, we can gain insight into the physiology of healthy aging and the development of new therapies to extend the human healthspan.

While it may seem obvious now, whether genes played a role in longevity was disputed for over a century. After all, rather than genes, wouldnt access to health care, socioeconomic status, diet, smoking, drinking, exercise, or many other environmental and lifestyle factors play a much larger role? Similar to height or intelligence (however the latter is assessed), the genetics of longevity is an enormously complicated and sensitive issue for unbiased studying.

Yet after only a few genetic studies of longevity, a trend quickly emerged.

The natural lifespan in humans, even under optimal conditions in modern societies, varies considerably, the authors said. One study, for example, found that centenarians lived much longer than people born around the same time in the same environment. The offspring of centenarians also have lower chances of age-related diseases and exhibit a more youthful profile of metabolism and age-related inflammation than others of the same age and gender.

Together, about 25 to 35 percent of the variability in how long people live is determined by their genesregardless of environment. In other words, rather than looking at nematode worm genes, we have a discrete population of humans whove already won the genetic lottery when it comes to aging. We just need to parse what winning means in terms of biology. Genes in hand, we could perhaps tap those biological phonelines and cut the wires leading to aging.

Identification of the genetic factors that underlie extreme human lifespan should provide insights into the mechanisms of human longevity and disease resistance, the authors said.

Once scientists discovered that genes play a large role in aging, the next question was which ones are they?

They turned to genome-wide association studies, or GWAS. This big data approach scans existing genomic databases for variations in DNA coding that could lead to differences in some outcomefor example, long versus short life. The differences dont even have to be in so-called coding genes (that is, genes that make proteins). They can be anywhere in the genome.

Its a powerful approach, but not that specific. Think of GWAS as rudimentary debugging software for biological code: it only looks for differences between different DNA letter variants, but doesnt care which specific DNA letter swap most likely impacts the final biological program (aging, in this case).

Thats a huge problem. For one, GWAS often finds dozens of single DNA letter changes, none powerful enough to change the trajectory of aging by itself. The technique highlights a village of DNA variants, that together may have an effect on aging by controlling the cells course over a lifetime, without indicating which are most important. Its also hard to say that a DNA letter change causally leads to (or protects against) aging. Finally, GWAS studies are generally performed on populations of European ancestry, which leaves out a huge chunk of humansfor example, the Japanese, who tend to produce an outsized percentage of centenarians.

So what needs to change?

Rather than focusing on the general population, the key is to home in on centenarians of different cultures, socioeconomic status, and upbringing. If GWAS are like fishing for a rare species in several large oceans, then the authors point is to focus on pondsdistributed across the worldwhich are small, but packed with those rare species.

Extremely long-lived individuals, such as centenarians, compose only a tiny proportion (~0.01 percent to 0.02 percent) of the United States population, but their genes contain a biological blueprint for healthy aging and longevity, the authors said. Theyre spared from usual age-related diseases, and this extreme and extremely rare phenotype is ideal for the study of genetic variants that regulate healthspan and lifespan.

Its an idea that would usually make geneticists flinch. Its generally thought that the larger the study population, the better the result. Here, the recommendation is to narrow our focus.

And thats the point, the authors argue.

Whatever comes out of these studies will likely have a much larger impact on aging than a GWAS fishing experiment. Smaller (genomic) pond; larger (pro-youth) fish. Whats more, a pro-youth gene identified in one European-based long-living population can be verified in another group of centenarianssay, Japaneseensuring that the gene candidates reflect something fundamental about human aging, regardless of race, culture, upbringing, and wealth.

A genomic screen of centenarians can easily be done these days on the cheap. But thats only the first step.

The next step is to validate promising anti-aging genetic differences, similar to how scientists validated such differences in nematode worms during classic longevity studies. For example, a promising pro-youth gene variant can be genetically edited into mice using CRISPR or some other tool. Scientists can then examine how the mice grow up and grow old, compared to their non-edited peers. Does the gene make these mice more resilient to dementia? What about muscle wasting? Or heart troubles? Or hair greying and obesity?

From these observations, scientists can then use an enormous selection of molecular tools to further dissect the molecular pathways underlying these pro-youth genetic changes.

The final step? Guided by centenarian genes and validated by animal models of aging, we can design powerful drugs that sever the connection between the genes and proteins that drive aging and its associated diseases. Metformin is an experimental pill that came out of aging studies in nematode wormsimagine what studies in human centenarians will yield.

Despite enormous improvements in human health over the past century, we remain far from a situation in which living to 100 years of age in fairly good health is the norm, the authors said.

But as centenarians obviously prove, this is possible. By digging into their genes, scientists may find a path towards healthy longevitynot just for the genetically fortunate, but for all of us.

Image credit:Cristian Newman / Unsplash

Follow this link:

The Secret to a Long, Healthy Life Is in the Genes of the Oldest Humans Alive - Singularity Hub

LogicBio Therapeutics Reports Second Quarter 2020 Financial Results and Provides Business UpdatesFDA Clears IND Application for LB-001 for the…

LEXINGTON, Mass., Aug. 10, 2020 (GLOBE NEWSWIRE) -- LogicBio Therapeutics, Inc. (Nasdaq:LOGC) (LogicBio or the Company), a company dedicated to extending the reach of genetic medicine with pioneering targeted delivery platforms, today reported financial results for the quarter ended June 30, 2020, provided a business update and announced the U.S. Food and Drug Administration (FDA) has cleared the Companys Investigational New Drug (IND) application for LB-001 for the treatment of methylmalonic acidemia in pediatric patients. LogicBio released a separate press release this morning providing further details on the planned Phase 1/2 clinical design for LB-001.

We are thrilled to have received clearance to move forward with this first-in-human clinical trial with our lead product candidate, LB-001, for the treatment of methylmalonic acidemia, a life-threatening congenital genetic disease with no current therapeutic treatment options. This represents a significant milestone in our goal of bringing a treatment to MMA patients as well as for our GeneRide platform. We have maintained continuous dialogue with the centers of excellence that are planned to participate in the Phase 1/2 clinical trial, and we look forward to activating these sites as quickly as possible, said Fred Chereau, CEO of LogicBio. We have instituted systems attempting to mitigate COVID-19 dynamics on our study start-up process and, based on our best estimates, we plan to enroll our first patient in early 2021.

Commenting on the Next Generation Capsid Program, Mr. Chereau said, We are very excited about the recent advances in our novel capsid program, which has generated liver-tropic capsids intended for use in gene editing technologies such as GeneRide and other gene therapy approaches. We are focused on executing across all of our programs and look forward to sharing further details on our novel capsids in early 2021.

Appointment of Daniel Gruskin, M.D. to SVP, Head of Clinical Development

Daniel Gruskin, M.D. was appointed as SVP, head of clinical development in August 2020. Dr. Gruskin has served as interim head of clinical development of LogicBio since June 2020. In April 2020, Dr. Gruskin started consulting with the Company as a special advisor. Previously, Dr. Gruskin served in roles of increasing responsibility at Sanofi Genzyme, most recently as vice president, head of global medical affairs, rare disease, in which capacity he oversaw medical affairs, life cycle management, scientific affairs and other medical and development activities related to metabolic, rare and/or genetic diseases. Prior to his role at Sanofi Genzyme, Dr. Gruskin served as assistant professor, human genetics and pediatrics at Emory University School of Medicine, where he was also the chief of the genetics section at Childrens Healthcare of Atlanta.

Daniel has been instrumental in leading LB-001 clinical development efforts including getting the IND cleared. His deep experience in genetic medicines and metabolic diseases will serve LogicBio well as we look to execute on our goals for both the GeneRide and Next Generation Capsid platforms in search of transformative medicines, said Mr. Chereau.

Anticipated Milestones for 2020 and 2021:

Second Quarter 2020 Financial Results

Three Months Ended June 30, 2020 and 2019

About LogicBio Therapeutics

LogicBio Therapeuticsis dedicated to extending the reach of genetic medicine with pioneering targeted delivery platforms.

LogicBios proprietary genome editing technology platform, GeneRide, enables the site-specific integration of a therapeutic transgene without nucleases or exogenous promoters by harnessing the native process of homologous recombination. LogicBio has received FDA clearance for the first-in-human clinical trial of LB-001, a wholly owned genome editing program leveraging GeneRide for the treatment of methylmalonic acidemia. Patient enrollment is expected to begin in early 2021. In addition, LogicBio has a collaboration with Takeda to research and develop LB-301, an investigational therapy leveraging GeneRide for the treatment of the rare pediatric disease Crigler-Najjar syndrome.

LogicBio is also developing a Next Generation Capsid platform for use in gene editing and gene therapies. Data presented have shown that the capsids deliver highly efficient functional transduction of human hepatocytes with improved manufacturability with low levels of pre-existing neutralizing antibodies in human samples. Top-tier capsid candidates from this effort demonstrated significant improvements over benchmark AAVs currently in clinical development. LogicBio is developing these highly potent vectors for internal development candidates and potentially for business development collaborations.

LogicBio is headquartered inLexington, Mass. For more information, please visitwww.logicbio.com.

Forward Looking Statements

This press release contains forward-looking statements within the meaning of the federal securities laws, including those related to the Companys plans to initiate, advance and complete its planned SUNRISE Phase 1/2 clinical trial of LB-001 in MMA; the timing, progress and results of the Companys research and development activities, including those related to the GeneRide technology platform and Next Generation Capsid Program; its plans for LB-301 in Crigler-Najjar; and the sufficiency of its cash and cash equivalents to fund operating expenses and capital expenditure requirements. These are not statements of historical facts and are based on managements beliefs and assumptions and on information currently available. They are subject to risks and uncertainties that could cause the actual results and the implementation of the Companys plans to vary materially, including the risks associated with the initiation, cost, timing, progress and results of the Companys current and future research and development activities and preclinical studies and potential future clinical trials. In particular, the impact of the COVID-19 pandemic on the Companys ability to progress with its research, development, manufacturing and regulatory efforts, including the Companys plans to initiate, advance and complete its Phase 1/2 clinical trial for LB-001 in MMA, and the value of and market for the Companys common stock, will depend on future developments that are highly uncertain and cannot be predicted with confidence at this time, such as the ultimate duration of the pandemic, travel restrictions, quarantines, social distancing and business closure requirements in the United States and in other countries, and the effectiveness of actions taken globally to contain and treat the disease. These risks are discussed in the Companys filings with the U.S. Securities and Exchange Commission (SEC), including, without limitation, the Companys Annual Report on Form 10-K filed on March 16, 2020 with the SEC, the Companys Quarterly Report on Form 10-Q filed on May 11, 2020, and the Companys subsequent Quarterly Reports on Form 10-Q and other filings with the SEC. Except as required by law, the Company assumes no obligation to update these forward-looking statements publicly, even if new information becomes available in the future.

Contacts:

Investors:Brian LuqueAssociate Director, Investor Relationsbluque@logicbio.com951-206-1200

Media:Stephanie SimonTen Bridge CommunicationsStephanie@tenbridgecommunications.com617-581-9333

Read more here:

LogicBio Therapeutics Reports Second Quarter 2020 Financial Results and Provides Business UpdatesFDA Clears IND Application for LB-001 for the...

Research on Tasmanian devil offers new insights into tackling human cancer – National Science Foundation

Findings offer hope for the Tasmanian devil, at the brink of extinction

This Tasmanian devil's future hangs in the balance, and with it, perhaps human health discoveries.

August 10, 2020

A rare, transmissible tumor has brought the iconic Tasmanian devil to the brink of extinction, but new research by scientists at Washington State University and the Fred Hutchinson Cancer Research Center in Seattle shows hope for the animals' survival -- and possible new treatments for human cancers.

The U.S. National Science Foundation-funded study, published in Genetics, found a single genetic mutation that leads to reduced growth of a transmissible cancer in Tasmanian devils in the wild.

"This gene is also implicated in human prostate and colon cancers," said biologist Andrew Storfer. "The findings hold the promise of helping save the world's few remaining Tasmanian devils, and could also translate to human health."

The research team studied the genomes of cases of devil facial tumor disease that regressed spontaneously -- that is, the cancer began disappearing on its own.

The scientists were surprised to find that the mutation contributing to tumor regression doesn't change the gene function but instead turns on a gene that slows cell growth in the tumor. At least, it behaves that way in the lab.

While infections cause up to 20% of all human cancers -- such as gastric cancer from Helicobacter pylori and cervical cancer from human papillomavirus -- for Tasmanian devils, the cancer is the infection.

The devil facial tumor disease spreads between the animals when they bite each other during common social behaviors. Since the mid-1990s, the disease has decimated the population of these carnivorous marsupials, which are now found only on the island state of Tasmania, off the southeastern coast of Australia.

"Although this disease is largely fatal, we're seeing tumors disappear from an increasing number of individual animals," Storfer said. "We hope to learn something that could be applied to understanding and possibly treating a number of human cancers in the future."

Added Sam Scheiner, a program director in NSF's Division of Environmental Biology, "These results show that basic research can produce totally unexpected results that are important for a completely different problem."

See the original post here:

Research on Tasmanian devil offers new insights into tackling human cancer - National Science Foundation