header logo image


Page 18«..10..17181920..30..»

Archive for the ‘Molecular Genetics’ Category

No Hydroxychloroquine Benefit in Randomized COVID-19 Trial – Medscape

Saturday, April 18th, 2020

Editor's note: Find the latest COVID-19 news and guidance in Medscape's Coronavirus Resource Center.

Hydroxychloroquine (HCQ) does not help clear the SARS-CoV-2 virus or relieve symptoms for COVID-19 patients more than standard care alone and has more side effects, a randomized controlled trial of 150 hospitalized adults in China suggests.

However, two experts caution that because of confounding, the trial is unable to answer convincingly the question of whether HCQ can benefit COVID-19 patients.

Wei Tang, with the Departments of Pulmonology and Critical Care Medicine at Ruijin Hospital, in Shanghai, China, and colleagues enrolled patients with COVID-19 from 16 treatment centers in China in February. They posted their findings on the medRxiv preprint server, but their paper has not been peer reviewed. A coauthor told Medscape Medical News the work has been submitted to a journal.

The overall 28-day negative conversion rate of SARS-CoV-2, which was the primary endpoint, was similar in the two 75-patient treatment groups. The Kaplan-Meier estimate for negative conversion rate was 85.4% in the HCQ plus standard of care (SOC) arm, vs 81.3% in the SOC-only group (P = .341). Negative conversion rates for the two groups were similar at days 4, 7, 10, 14, and 21.

Adverse events were reported in 8.8% of patients in the control group compared with 30% in the HCQ group. Diarrhea was the most common side effect, occurring in 10% of patients in the HCQ group vs none in the control group. Two patients in the HCQ arm had serious adverse events; one experienced disease progression, and the other experienced upper respiratory tract infection.

Patients in the HCQ group received a high loading dose of 1200 mg daily for 3 days followed by a maintenance dose of 800 mg daily for the remaining days. Total duration was 2 weeks for patients with mild or moderate disease and 3 weeks for those with severe disease.

The two arms were similar in alleviation of symptoms by day 28: 59.9% with HCQ plus SOC vs 66.6% with SOC alone.

However, the researchers said that in a post hoc analysis, they found a significant reduction of symptoms after adjusting for the confounding effects of antiviral agents (hazard ratio, 8.83; 95% confidence interval, 1.09 71.3).

In addition, Tang and colleagues report a significantly greater reduction of C-reactive protein (CRP), a biomarker for inflammation, from baseline to day 28 in the HCQ group in comparison with the control group (6.986 vs 2.723 mg/L).

The authors suggest the alleviation of symptoms may come from HCQ's anti-inflammatory effects.

The mean age of the patients was 46 years, and 55% were male. Almost all patients had mild or moderate disease; two had severe disease.

J. Michelle Kahlenberg, MD, PhD, research professor of rheumatology at the University of Michigan in Ann Arbor, told Medscape Medical News that it's important to note that in the post hoc analysis, 89% of the patients in this trial were receiving other therapy in addition to HCQ.

"When [the researchers] say they saw improvement in symptoms when they removed the confounders, what they actually did was remove the patients from the analysis that got antivirals, and that left 14 patients in each arm," Kahlenberg said.

Moreover, Kahlenberg noted, 20% of patients who received HCQ had mild symptoms, whereas only 9% of those in the SOC group did.

"We don't know how those patients played out in the post hoc analysis whether it was the patients who were really mild that didn't get the antivirals that were left in the hydroxychloroquine group and that's why they had a slightly faster resolution of symptoms," she said.

She said that in this study, the researchers calculated CRP in milligrams per liter, whereas in the United States, it is measured in milligrams per deciliter. The conversion highlights the fact that the reduction in CRP was not terribly noteworthy, she said.

"The patients with COVID who tend to tank and have cytokine storms their CRP is much higher," she said. "So the small improvement in CRP wasn't that exciting.

"I don't think this gets us anywhere closer to an answer. It's another muddy study," she said.

Similarly, Christopher V. Plowe, MD, MPH, director of the Global Health Institute at Duke University in Durham, North Carolina, told Medscape Medical News he sees no convincing answers in this study.

Plowe, professor of medicine, molecular genetics, microbiology, and global health at Duke, also noted differences between the two groups at enrollment.

For example, the HCQ group had more than three times the number of patients with shortness of breath (22.1% vs 5.9%); more with sputum production (16.2 vs 5.9%); and more with cough (51.5% vs 38.2%). In addition, the average age was 4 years higher in the HCQ group.

"It makes me wonder whether the randomization was truly random," Plowe said.

Plowe also questioned the authors' statement that they didn't see cardiac arrhythmia events, such as prolonged QT intervals. "I can't see any evidence that they did an EKG on anybody," he said.

"This study leaves the door open to the possibility that hydroxychloroquine may have a clinical benefit. If there is a benefit, it seems to be related to the drug's anti-inflammatory properties. If that's the case, I'm not sure this particular drug, as opposed to others, would be the way to go," Plowe said.

"Our negative results on the anti-viral efficacy of HCQ obtained in this trial are on the contrary to the encouraging in-vitro results and to the recently reported promising results from a non-randomized trial with 36 COVID-19 patients," the authors write.

However, the 36-patient trial to which they refer has since been called into question, as previously reported by Retraction Watch.

Despite lack of clear evidence of benefit, HCQ is recommended off label for the treatment of COVID-19 by the Chinese National guideline, and the US Food and Drug Administration has issued an emergency-use authorization for the treatment of adult patients with COVID-19.

By contrast, the Infectious Diseases Society of America recently concluded that because of insufficient data, they could not recommend any particular treatment for patients with COVID-19.

The work was supported by the Emergent Projects of National Science and Technology; the National Natural Science Foundation of China; the National Key Research and Development Program of China; the Shanghai Municipal Key Clinical Specialty; the National Innovative Research Team of High-Level Local Universities in Shanghai; the Shanghai Key Discipline for Respiratory Diseases; the National Major Scientific and Technological Special Project for Significant New Drugs Development; and Key Projects in the National Science and Technology Pillar Program. The authors, Kahlenberg, and Plowe have disclosed no relevant financial relationships.

Follow Medscape on Facebook, Twitter, Instagram, and YouTube. Have a tip for us? Contact us.

See the rest here:
No Hydroxychloroquine Benefit in Randomized COVID-19 Trial - Medscape

Read More...

UNM scientists get a better idea of where coronavirus started, how the spread will continue – KRQE News 13

Saturday, April 18th, 2020

ALBUQUERQUE, N.M. (KRQE) Where did a person in New Mexico get the coronavirus? How can we stop this from happening again? University of New Mexico scientists are trying to figure all of that out.

Using samples from people who tested positive for coronavirus, researchers are getting a better idea of where the virus started and how it will continue to spread. We can actually get an estimation on a total number of infections potentially in the state, says Dr. Darrell Dinwiddie, Assistant Professor at the Division of Genetics in the Department of Pediatrics.

UNM scientists are taking a closer look at the coronavirus. Those molecular clues, as we call them, are what gives us the ability to track how the virus is spreading, says Dr. Daryl Domman, Assistant Professor at the Center of Global Health. Those molecular clues are genomes. We compare the genetic sequences. The genome sequence of the virus were seeing in New Mexico, Dinwiddie says.

From that genetic sequence, they can determine how the virus were seeing here compares to stains around that nation, We can see if the early cases in New Mexico, for example, we can see that theyre similar to viruses that are coming out of New York or Europe that gives us the indication that they probably came from those places as well, Dinwiddie says.

They can also determine how the virus is entering and spreading in the community. If we see a cluster of samples that were six or seven samples and they all have the exact same genome sequence of the virus that suggests they all got infected within a week or two of each other potentially through a direct contact or a shared contact, Dinwiddie says.

Dinwiddie and Domman say so far, theyve tested 48 genomes from New Mexico and have 1,400 more ready for testing. Information for those tests tell them how long the virus has been spreading and what it might do next. This gives and empowers our public officials. They can make informed decisions on what needs to be implemented, Domman says.

By the end of the week, the research team expects to have some projections for how many more cases we could see in New Mexico. New Mexico is one of 19 states working with the CDC to generate the genome sequencing data.

Follow this link:
UNM scientists get a better idea of where coronavirus started, how the spread will continue - KRQE News 13

Read More...

Genomic mapping in outbred mice reveals overlap in genetic susceptibility for HZE ion and -rayinduced tumors – Science Advances

Saturday, April 18th, 2020

INTRODUCTION

Interplanetary space is populated by densely ionizing particle radiation not naturally present on Earth (1). Life on Earth has evolved under the protection of a geomagnetic field, which deflects high-charge, high-energy (HZE) ions; however, the constant flux of HZE ions in deep space is essentially impossible to shield, making astronaut exposures inevitable (2).

In the absence of human epidemiological data for exposures to HZE radiation, uncertainties surround the cancer risk estimates for space flight crews that venture beyond low Earth orbit. The current NASA space radiation cancer risk model is built largely upon epidemiological data from the survivors of the Hiroshima and Nagasaki atomic bombings, a cohort of individuals exposed predominantly to -rays (35), a form of photon radiation. One key assumption in this NASA model is that the spectra of tumor types, and their biologic behaviors, will be similar for individuals exposed to ionizing radiation, whether particle or photon. However, notable physical differences exist between ionizing photon and particle radiation, and these physical differences translate to unique ionization and damage patterns at the molecular, cellular, and tissue levels. HZE ion exposures produce spatially clustered DNA double-strand breaks, along with other DNA lesions in close proximity to break sites (6). In contrast, -rays produce sparse ionization events that are random in spatial distribution and less likely to have additional DNA lesions immediately adjacent to the break sites. Other assumptions in the model are that radiogenic tumors are no more lethal than their sporadic counterparts and that females are at greater risk for radiogenic cancers than males (7).

In assessing cancer risks to astronauts, the premise that HZE ion exposures increase the risk for the same types of tumors that arise in human populations exposed to -rays is supported by the few animal studies of HZE ion carcinogenesis conducted to date (8). These studies, conducted on genetically homogeneous animals, have demonstrated that tumor types arising in HZE ionirradiated animals are the same as those that occur spontaneously in these animals or following exposure to photon radiation (8). However, all previous data are from either inbred mice (9, 10) or rats (11), F1 hybrid mice (12, 13), or rat stocks with limited genetic heterogeneity (11, 1416), and the tumor types that arise in inbred rodents are determined, in very large part, by their genetic background. Therefore, the spectrum of tumors that might arise in a genetically diverse population exposed to HZE ions is unknown.

With the emergence of multiparent outbreeding strategies that produce highly recombinant mouse populations with allelic variants from multiple founder strains (1719), it is possible to model the effects of population diversity in carcinogenesis studies by minimizing the overwhelming effects of genetic background and increasing the phenotypic repertoire available within a test population. These populations also allow for high-precision genetic mapping (18, 20). Quantitative trait locus (QTL) mapping is a powerful forward-genetics approach that allows for unbiased testing of genetic variants that may influence gene-environment interactions for radiation effects (21, 22). Highly recombinant populations were constructed for the purpose of mapping complex traits, and QTL can often be resolved to megabase resolution (1820). In addition, complete sequence information can be used on genotyped individuals by imputing the substantial genomic resources available for the founder strains.

Studying tumors that arise in irradiated, genetically diverse mouse populations presents a unique opportunity to test key assumptions of the NASA risk model, particularly whether HZE ions induce the same tumors by the same mechanisms as -rays. If so, the current practice of extrapolating human epidemiological data from individuals exposed to -rays to astronauts exposed to HZE ions would be a valid approach for risk calculation in the space radiation environment.

To study the effects of HZE ion irradiation in a genetically heterogeneous population, 1850 HS/Npt stock mice (23) of both sexes were genotyped for 77,808 single-nucleotide polymorphism (SNPs) and exposed to (i) 0.4 gray (Gy) of 28Si ions (240 MeV/n) [linear energy transfer (LET), 80 keV/m; = 0.031 particles/m2] or (ii) 56Fe ions (600 MeV/n) (LET, 181 keV/m; = 0.014 particles/m2), (iii) 3 Gy of 137Cs -rays, or (iv) sham irradiation. We chose 56Fe ions because of their high abundance in galactic cosmic radiation (GCR) and because their high charge (Z = +26) makes them particularly damaging (24). The 28Si ions were selected because their LET more closely approximates the dose average LET of secondary fragments generated by GCR penetrating an aluminum spacecraft hull (25). The mice were monitored daily until they reached 800 days of age or became moribund. Comprehensive necropsies were performed on each mouse and involved all organ systems. Each detected lesion was characterized histologically by a board-certified veterinary pathologist. Tumors were the predominant cause of morbidity and mortality for both HZE ionirradiated (n = 622) and -rayirradiated (n = 615) populations as well as for the population of unirradiated mice (n = 613). Overall life span was significantly reduced for irradiated populations (Fig. 1A), which can be attributed to the increased incidence and decreased median survival for radiation-induced tumors. For irradiated mice, populations exposed to 0.4-Gy HZE ions had increased survival times compared to mice exposed to 3.0 Gy of -rays (Fig. 1A). Although these doses seem disparate, their selection is based on preliminary dose-response studies (26), which reveal that 0.4 Gy of HZE ions and 3.0-Gy -rays are each maximally tumorigenic.

Overall survival for HS/Npt mice, plotted as Kaplan-Meier survival, is presented for each exposure group (A). The incidence of specific tumor histotypes (B) and median survival times for these tumors (C) are plotted for each exposure group, which demonstrates that certain tumor types occur at an increased frequency following exposures to radiation of specific qualities and survival times in irradiated mice are decreased for some tumor types. The incidence of specific tumor histotypes within HS/Npt families is plotted for unirradiated (D), -rayirradiated (E), and HZE ionirradiated families (F) and demonstrates that specific tumor types often occur at very high incidence within some families and not at all in others, indicating heritability of tumor susceptibility. Furthermore, adjacent families are more closely related, and tumor incidences, for example, family 23 and adjacent families, have a high incidence of B cell lymphoma. The 47 HS/Npt families are arranged along the x axis (D to F).

A wide variety of tumor diagnoses [82 distinct tumor histotypes (table S1)] were observed in HS/Npt mice. Although most of these tumor types were rare, 18 histotypes were observed at incidences greater than 1%. Overall, the spectra of tumor histotypes produced in genetically diverse populations exposed to HZE ions and -rays were similar (Fig. 1B). Furthermore, tumor types induced by radiation were generally similar to those arising spontaneously in HS/Npt mice; however, radiation-exposed populations demonstrated decreased median survival times associated with tumor development (Fig. 1C and figs. S7 to S22) and increased incidences for specific tumor types, such as leukemias and Harderian gland adenocarcinomas, following radiation (Fig. 1B). The structure of the HS/Npt population can be divided into families that consist of mice more closely related to one another. Many tumor histotypes show high incidences within some families but are absent or rare in others (Fig. 1, D to F), which is consistent with genetic susceptibility to certain tumor types. Furthermore, certain tumorsparticularly lymphomas, pulmonary adenocarcinomas, hepatocellular carcinomas, Harderian gland tumors, and myeloid leukemiasdemonstrate a periodicity in tumor incidence (Fig. 1, D to F) where adjacent families often display similar incidences, which could be predicted on the basis of the circular breeding design used to generate HS/Npt, in which adjacent families are more related to one another than families further removed.

Although the tumor spectra are similar for each irradiated population, the different radiation qualities demonstrate varied efficiencies for producing specific tumor histotypes. -rayirradiated mice were at greater risk for myeloid leukemia, T cell lymphoma, pituitary tumors, and ovarian granulosa cell tumors than unirradiated mice; HZE ionirradiated mice demonstrated an intermediate susceptibility to these histotypes (Fig. 1B). For Harderian gland tumors, thyroid tumors, hepatocellular carcinomas, and sarcomas, HZE ion and -rayirradiated mice were at a similarly and significantly increased risk compared to unirradiated controls (fig. S7 to S22).

NASA permissible exposure limits for radiation limit the number of days an astronaut can spend in space based on modeled cancer risk. These limits are different for men and women (27) due primarily to epidemiological data that indicate that women are at greater risk for radiogenic cancers than men due to their longer life spans and susceptibility to specific cancer types, such as lung, ovarian, and breast carcinomas. Female HS/Npt mice have longer life spans than males (P = 2.7 106, log-rank test), with unirradiated females living 43 days longer (686.1 days), on average, than males (643.2 days) (fig. S1A). In contrast, no survival difference is observed between -rayirradiated females and males (P = 0.51) or HZE ionirradiated females and males (P = 0.06), indicating that female HS/Npt mice are more susceptible to radiation-induced morbidities and mortalities than males (fig. S1, B and C). Irradiated female mice had increased incidences of (i) ovarian tumors, (ii) mammary tumors, (iii) central nervous system tumors (pituitary adenomas, choroid plexus tumors, and ependymomas), (iv) diffuse large B cell and lymphoblastic B cell lymphomas, (v) osteosarcomas, and (vi) leiomyosarcomas (fig. S1D). Female mice were at lower risk for radiogenic lung cancer (fig. S1D and table S1), which is a major contributor to limiting flight time for female astronauts. Modeling risk by sex in humans has been confounded by different smoking rates between men and women in the atomic bomb survivor cohort (28).

To determine whether the genetic variants that increase tumor susceptibility following -ray irradiation also increase tumor susceptibility following HZE ion irradiation, genome-wide association mapping was performed for 18 tumor types in which there was an incidence of greater than 1%. Genomes were reconstructed for each mouse using a probabilistic model to predict founder haplotypes from high-density genotype data (18). Reconstructed genomes represent the unique accumulation of meiotic events for each individual and form a scaffold for the imputation of known sequencing information from the eight parental inbred strains. Polygenic covariance among related individuals is of significant concern in multiparent crosses and was corrected for during QTL mapping with a kinship term (18, 29). Mapping was performed for each phenotype using both a generalized linear mixed-effects model and proportional hazards regression model with the aforementioned kinship to adjust for polygenic covariance between related mice. To determine the significance thresholds for a model in which no QTL is present, the phenotypes were permuted, the regression model was run, and the maximum statistic was retained from each permutation (30). The 95% significance threshold was minimally variable between phenotypes with a mean threshold of log(P) > 5.8, and this value was used to identify significant associations. This is consistent with the estimated 0.05 Bonferroni genome-wide corrected threshold of log(P) > 6.0, which is considered overly conservative for QTL mapping (30).

At least one QTL was identified for 13 of the 18 tumor phenotypes examined. For tumor incidence, 35 QTL were identified with an average confidence interval of 3.4 Mb (table S2). For QTL at the 95% confidence threshold, effect sizes average 3.7% of the phenotypic variance with a range of 0.75 to 7.46%. For most of the tumors, the genetic architecture was complex with multiple QTL individually explaining a small proportion of the total variance. Although loci with moderate effects on the phenotype were most common, 11 large effect QTL were observed for seven tumor histotypes, with effect sizes greater than 5% (table S2).

To determine potential effects of genetic variants on tumor latency following irradiation, mapping was also performed using proportional hazards regression model (table S3) and 38 QTL were identified for 12 tumor types. QTL associated with tumor survival times mirrored those identified for tumor incidence, indicating that the genetic variants that control susceptibility to radiation-induced tumors also determine latencies.

Neoplasia is a binomially distributed trait, and therefore, the power to detect significant associations is primarily dependent on tumor incidence and QTL effect size. This leads to important considerations for the ultimate goal of this analysis, which is to determine similarities between QTL for specific neoplasms in populations exposed to different qualities of radiation. For some tumor types, a significant peak was observed in one exposure group with a suggestive peak present at the same locus in the alternative exposure group. We speculate that the reason certain radiation qualities produce only suggestive QTL for certain tumor phenotypes is likely due to decreased mapping power as a result of the variation in incidence between groups. In these cases, if the peak was more significant when combining radiation groups, the QTL was considered significant for all irradiated animals regardless of radiation quality.

Thyroid tumors are a well-known radiation-induced entity for both humans and mice; however, relatively little is known about genetic variants that increase susceptibility to this disease in mice. In HS/Npt mice, spontaneous thyroid adenomas occurred at relatively low frequencies and had a uniformly late onset, with tumors occurring between 700 and 800 days of age (Fig. 2A). In contrast, thyroid tumors arising in HZE ion or -rayexposed mice occur with significantly earlier onsets, with tumors arising as early as 250 days of age (Fig. 2A).

Thyroid follicular adenoma Kaplan-Meier survival estimate (A) along with genome-wide association plots for thyroid adenoma in HZE ionirradiated, -rayirradiated, HZE ion and -rayirradiated, and unirradiated mice (B) and an expanded plot for chromosome 2 (C), which contains the most significant association locus; gray lines indicate 95% (upper line) and 90% confidence (lower line) for log10(P values). Genome-wide association results reveal significant results in HZE ion and -rayirradiated mice that are further bolstered by combining the groups. The top panel of (D) shows strains that contribute the reference allele for the SNPs highlighted in red in the middle panel, indicated by vertical lines (D); the C57BL/6J strain contributes an allele that differs significantly from the other seven strains. The middle panel shows the log10(P value) of each SNP in the interval (D); the most significant SNPs are highlighted in red, and the bottom panel lists genes within the QTL interval. Genes that contain splice site, missense, or stop-related SNPs are colored red (D). Resample model averaging was performed within chromosome 2 to compare the distribution of peak log10(P values) for each exposure group (E); there is broad overlap for HZE- and -rayirradiated mice, and grouping all irradiated mice together further narrows the distribution of peak log10(P values). Mbp, megabase pair.

Association mapping reveals a significant 3.4-Mb interval on chromosome 2 for HZE ionexposed animals (Fig. 2, B and C). The same locus is identified in the -rayirradiated population if the significance threshold is decreased to a level at which 30% of identified QTL will be false positives. Combining both irradiated populations markedly increases the significance of the QTL identified on chromosome 2. The QTL interval (119 to 125 Mb) contains 39,179 SNPs (Sanger Mouse Genomes, REL-1505) and 142 genes (Ensembl version 85) (Fig. 2D). Within the QTL region, the C57BL/6J parental strain contains an introgression from the Mus musculus musculus genome (31); we found that HS/Npt mice carrying the C57BL/6J haplotype at the QTL have increased thyroid tumor incidence regardless of whether they are exposed to HZE ions or -rays.

To further explore the possibility that the QTL identified on chromosome 2 controls susceptibility following -ray and HZE ion exposures, we used a nonparametric resample model averaging procedure (32) across the entire chromosome to identify genomic loci that consistently reappear in resampled populations. Briefly, genome scans are repeated for each new dataset created, in which some individuals may be sampled more than once and some not at all (32). Resample model averaging consistently identifies the same locus for all groups of mice, regardless of radiation exposure (Fig. 2E). Furthermore, the resample model averaging procedure identifies the same locus for tumors arising spontaneously (Fig. 2E). Data from this tumor phenotype indicate that the same inheritable genetic variants contribute to an individuals risk of developing thyroid cancer, regardless of radiation exposure.

Acute myeloid leukemia (AML) is another common radiation-induced tumor in both mice and humans (33, 34). In concordance with previous studies conducted with inbred mice (26), -ray exposures in HS/Npt mice are more efficient at inducing AML than HZE ion exposures. In our -irradiated mice, 15.6% (96 of 615) developed AML compared to 2.9% (18 of 622) of those exposed to HZE ions and 1.6% (10 of 613) of unirradiated mice. AML median survival times were similar for all groups (Fig. 3A). Association mapping revealed a significant QTL for the -irradiated population on chromosome 2 that reached the 95% confidence threshold (Fig. 3, B and C), but no QTL was observed for the HZE ionexposed population, in which the incidence of AML was much lower. However, when grouping HZE ion and -rayirradiated mice together, the same QTL was significantly bolstered (Fig. 3B). If the susceptibility alleles identified at this locus were only contributing to disease following -ray irradiation and were, therefore, randomly distributed among the affected mice in the HZE ionexposed group, then we would expect the log10(P values) to decrease when combining -irradiated mice; however, the log10(P value) for this locus significantly increases when repeating the mapping procedure included all irradiated mice.

(A) Kaplan-Meier plots for myeloid leukemia demonstrate similar median survival estimates for myeloid leukemia between groups. (B) Genome-wide association procedures identify a narrow QTL on chromosome 2; two gray lines indicate 95% (upper line) and 90% confidence (lower line) for log10(P values). Expanded mapping results are depicted in (C) along with contributing strains for the reference allele. The A/J, AKR/J, C57BL/6J, DBA/2J, and LP/J strains contribute alleles that differ from the other strains, indicated by vertical lines in the top panel (C). The middle panel shows the log10(P value) of each SNP in the interval. The most significant SNPs are highlighted in red. The bottom panel shows the genes in the QTL interval. Genes that contain splice site, missense, or stop-related SNPs are indicated in red. Copy number results for Spi1 and Asxl1 in splenic samples from mice diagnosed with myeloid leukemia are plotted by exposure group (D).

Radiation-induced AML is a well-characterized disease in mice (10, 35, 36) and is most commonly the result of a radiation-induced minimally deleted region on chromosome 2 containing the PU.1 gene (current murine nomenclature, Spi1) and a recurrent point mutation that inactivates the remaining Spi1 allele (37). Figure 3C depicts mouse chromosome 2 with the positions of the QTL identified in our irradiated mice and the Spi1 gene. To test the hypothesis that AMLs occurring in HZE ionexposed animals will contain the same molecular aberrations know to occur in AML arising in -rayexposed mice, the copy number for Spi1 was investigated in leukemia samples to assess for deletions. As expected, most of the leukemias occurring in -rayexposed mice had a deletion in one copy of Spi1. In contrast, Spi1 deletions in spontaneously occurring AML were less common (Fig. 3D). Similar to -rayirradiated mice, leukemias that developed in mice exposed to HZE ions, although fewer in number, also have an increased incidence of Spi1 deletion. This finding indicates that AML arises by similar molecular mechanisms following exposures to HZE ions or -rays.

Because the QTL identified on chromosome 2 is approximately 60 Mb from the commonly deleted region containing Spi1 and because radiation-induced deletions can be notoriously large, we considered the possibility that the identified QTL was also deleted in these leukemias, resulting in loss of one copy of the QTL region. To test this hypothesis, we determined the copy number for a gene located at distal to the QTL support interval, Asxl1. As expected, we found that Asxl1 was not deleted in any sample in which Spi1 was not deleted; however, in 69% of cases with a Spi1 deletion, Asxl1and presumably the entire QTL regionwas also deleted (Fig. 3D). This demonstrates that most of the radiation-induced AML cases arose from progenitor cells haploinsufficient for the entire QTL region.

HZE ion and, to a lesser extent, -ray irradiation were particularly effective in inducing Harderian gland tumors at the doses used in this study, which was expected on the basis of extensive published radiation quality data on these tumors (8, 38). In the HZE ionirradiated group, Harderian gland tumors were observed in 22.7% (221 of 622) of mice and 3.2% (20 of 622) were malignant. In the -irradiated group, 15.3% (94 of 615) of mice developed Harderian gland tumors and 2.7% (17 of 615) were malignant. In contrast, spontaneous Harderian gland tumors occurred in only 4.1% (25 of 613) of unirradiated mice and 0.7% (4 of 613) were malignant. Despite the differences in tumor incidences following irradiation, median survival times for Harderian gland adenocarcinoma were similar for all groups (HZE ion, 582 days; -ray, 571 days; and unirradiated mice, 571 days).

Two QTL were observed for Harderian gland adenocarcinomas in HZE ionirradiated mice, one on chromosome 4 and another on chromosome 9 (Fig. 4A). The 1.7-Mb interval identified on chromosome 4 (Fig. 4B) is similar to previously discussed QTL regions in that combining both irradiated populations markedly increases the significance of this locus, which suggests that this QTL is associated with Harderian gland adenocarcinoma susceptibility in both HZE ion and -rayirradiated mice. In contrast, a 2.3-Mb QTL interval on chromosome 9 is observed only in HZE ionirradiated mice, and the locus is absent when combining all irradiated mice and repeating the mapping procedure (Fig. 4C). To further evaluate these QTL, resample model averaging was performed within chromosomes 4 and 9 to determine the distribution of peak log10(P values) along each chromosome. For chromosome 4, there is substantial spatial overlap identified in peak log10(P value) associations in the HZE ionexposed population and the -rayirradiated population, and the HZE ion and -rayirradiated population yields the most consistent identification of the QTL region (Fig. 4D). In contrast, although nearly all identified peak log10(P values) were identified in the 2.3-Mb QTL interval on chromosome 9 for HZE ionirradiated mice, the distributions of peak log10(P values) for other exposure groups do not substantially overlap and are widely distributed along the chromosome (Fig. 4E). The resample model averaging results indicate that while the chromosome 4 QTL contributes to susceptibility to Harderian gland adenocarcinomas in both HZE ion and -rayirradiated populations, the QTL identified on chromosome 9 appears to only be involved in Harderian adenocarcinoma susceptibility following HZE ion exposures.

Genome-wide association plots for Harderian gland adenocarcinoma (A) for HZE ionirradiated, -rayirradiated, HZE ion and -rayirradiated, and unirradiated mice; two gray lines indicate 95% (upper line) and 90% confidence (lower line) for log10(P values). Chromosome 4, which is expanded in (B), reveals a significant QTL associated with HZE ion irradiation, which is further increased significantly when grouping all irradiated mice (HZE ion and -ray irradiated) together, which indicated that the genetic variants in this location are important for Harderian gland adenocarcinoma following exposures to either HZE ion or -ray irradiation. In contrast, chromosome 9, which is expanded in (C), reveals a significant QTL associated only with HZE ion irradiation; this locus is absent when grouping all irradiated mice (HZE ion and -ray irradiated) together, which suggests that the allele(s) present in this region may only play a role for HZE ioninduced tumors. Resample model averaging was performed within chromosomes containing significant QTL. There is significant spatial overlap identified on chromosome 4 for peak log10(P value) associations in the HZE ionexposed population, the -rayirradiated population, and the HZE ion and -rayirradiated population that demonstrates the most consistent identification of the QTL region (D). In contrast, although nearly all identified peak log10(P values) were identified in the chromosome 9 QTL interval for HZE ion irradiated mice, the peak log10(P values) for other exposure groups are widely distributed along the chromosome (E).

In addition to looking for similarities between individual, selected QTL for HZE ion and -rayexposed populations, we also sought a more holistic method in which entire genome-wide association results could be compared between groups in an unsupervised process. We used hierarchical clustering to create cluster dendrograms using entire genome-wide scans for a given phenotype. By considering results from genome-wide associations, rather than individualized peaks observed within genome-wide associations, we submit for comparison not only highly significant QTL regions but also the numerous loci detected with lower confidence.

Unsupervised hierarchical clustering of genome scans creates significant clustering events that often occur for the same histotype regardless of radiation exposure (Fig. 5A). Multiple tumor histotypesincluding mammary adenocarcinoma, thyroid adenoma, and hepatocellular carcinomacluster by histotype, regardless of radiation exposure. To demonstrate and validate the methodology of QTL clustering, genome-wide scans for coat colors in each treatment group are evaluated and coat color genome-wide scans cluster together, as expected (Fig. 5B). These results further support the hypothesis that host genetic factors are highly important in determining risk of radiation carcinogenesis, whether following HZE ion or -ray exposures.

(A) Unsupervised hierarchical clustering of genome-wide association scans for tumor phenotypes reveals that the most significant clustering events often occur for the same histotype regardless of radiation exposure; these include mammary adenocarcinoma, thyroid adenoma, and hepatocellular carcinoma. (B) As expected, clustering genome scans for coat color demonstrates the expected results: that genome scans cluster together despite exposure group. The green line represents the 99% confidence level of the most significant dendrogram heights by permutations (log10 values permuted with genetic markers) to determine a distribution of dendrogram heights under the null hypothesis that no associations exist (C), demonstrating that the observed clusters are highly unlikely to occur randomly.

Permissible exposure limits for astronauts are based on the risk of death from cancer rather than cancer development, and the incidence to mortality conversion used in the risk calculation uses spontaneously occurring cancers in the U.S. population. Thus, there is an assumption that radiogenic tumors are no more lethal than spontaneous tumors. To determine whether tumors that arise following HZE ion exposure are more malignant than their counterparts arising in unirradiated or -rayirradiated mice, metastatic disease was characterized for each group. Pulmonary metastases were consistently observed in cases of hepatocellular carcinoma, Harderian gland adenocarcinoma, osteosarcoma, and ovarian granulosa cell tumor. Metastases were no more frequent in irradiated animals than in controls, and there was no significant difference in metastatic incidence between HZE ionirradiated mice and -rayirradiated mice (fig. S5A), and pulmonary metastatic density is similar between groups (fig. S5, B to D).

Tumor latency following irradiation was compared between exposure groups using survival statistics. Differences in tumor latency in this context indicate a decrease in time for tumor initiation or promotion. Since radiation is efficient at both initiation and promotion, decreased latencies are expected for irradiated population. Tumor progression is not evaluated, and our results therefore do not demonstrate whether tumors arising in irradiated individuals are more likely to progress rapidly than those arising spontaneously. As expected, tumors arising in both HZE ion and -rayirradiated mice show significantly decreased latencies in comparison to the unirradiated population (fig. S7 to S22). However, HZE ions did not further decrease latencies when compared to -rayirradiated mice.

Carcinogenesis as a result of space radiation exposure is considered the primary impediment to human space exploration (2). Compared to forms of radiation found naturally on Earth, including x-rays, -rays, and particles, HZE ions in space are much more difficult to shield (2) and have a distinct ionization pattern that aligns along dense track structures, resulting in clustered damage to chromatin (6). Because HZE ions, a highly penetrating component of GCRs, are not amenable to shielding (28, 29), exposure risks are inherent to manned missions in interplanetary space, but estimating the risk associated with this unique form of particle radiation is complicated by the essential lack of data for human exposures (28). As a substitute, human exposure data from other forms of ionizing radiation, primarily -ray (35) photon radiation, are used in cancer risk models with the assumption that photon and particle radiation have qualitatively comparable biological effects.

Animal models are a vital component in determining the validity of the extrapolation of human terrestrial radiation exposure data to exposures that will occur in astronauts in the space radiation environment. To date, carcinogenesis studies designed to evaluate the effects of HZE ions have used rodents with limited genetic heterogeneity (916). The advantage of removing genetic variability in animal models is the consequent decrease in phenotypic variability, which allows for fewer individuals to detect potential environmental effects on phenotype; the disadvantage is that strain-specific responses in genetically identical populations are significant and can obscure the variability that one might expect in a diverse population, such as humans. By using a genetically diverse population with a wide range of tumor susceptibilities, the spectra of tumors that occur following exposures to particle and photon radiation can be compared. The results of this study indicate that the spectrum of tumor histotypes observed in a genetically diverse population exposed to particle radiation is not unique to that observed in a population exposed to photon radiation or to the tumor spectrum observed in an unirradiated population. Despite the similarities observed in tumor spectra following radiation exposures, the radiation qualities and doses used for this study have unique efficiencies at producing specific tumor types, and while this work demonstrates that the underlying genetics of susceptibility can be similar for tumorigenesis following both high- and low-LET radiation, further work is necessary to define risks for specific tumor histotypes based on exposures.

This study uses a highly recombinant mouse population (HS/Npt stock) that is genetically diverse and designed for genome mapping (1921, 23), a forward-genetics approach that allows for an unbiased search of the entire genome for genetic associations. In contrast, genetically engineered mouse models rely on a reverse-genetics approach in which a given gene is first altered and the resulting phenotypes are then characterized. Studies using forward-genetics are most informative in populations that contain abundant genetic and phenotypic diversity. HS/Npt mice are a multiparent cross derived from eight inbred strains (A/J, AKR/J, BALBc/J, CBA/J, C3H/HeJ, C57BL/6J, DBA/2J, and LP/J); each individual contains a unique mosaic of founder haplotypes and a high degree of heterozygosity, and recombination events become increasingly dense with each generation. Our population of HS/Npt mice was obtained from generation 71 of circular outbreeding. Creating these populations is not trivial and has been a central goal of communities involved in genetics research over the past few decades, resulting in the creation of rodent populations ideal for genome mapping (1820, 3942).

Genome mapping allows the discovery of QTL associated with susceptibility to complex traits, such as radiogenic cancers; this approach is uniquely suited to comparing inheritable risk factors for cancers following exposures to unique carcinogens, such as particle and photon radiation. In broader terms, this work demonstrates the utility of highly recombinant mouse models created for genetic mapping in carcinogenesis studies, an application that has not been previously attempted. Mapping QTL in carcinogenesis studies provides inherent challenges due to the structure of binomial data, potential confounding causes of death following irradiation and aging, the fundamental stochastic nature of radiation tumorigenesis, and incomplete penetrance of potential allelic variants. Despite these challenges, we were able to map QTL for 13 neoplastic subtypes and many of these identified loci are previously unidentified.

At the doses used in this study, HZE ions appear to be less effective than -rays in inducing precursor T cell lymphoblastic lymphoma (pre-T LL) and ovarian tubulostromal adenomas and granulosa cell tumors. This may be due to a combination of dose inhomogeneity in HZE ionirradiated tissues and the major role cell killing plays in the etiology of these specific tumors. pre-T LL can be prevented by transplanting irradiated mice with unirradiated syngeneic bone marrow cells or by shielding some of their bone marrow during irradiation (43, 44). The underlying mechanism by which unirradiated bone marrow cells suppress lymphomagenesis may involve a cell competition process by which older T cell progenitors resident in the thymus are normally replaced by fresh progenitors that immigrate from the bone marrow. Radiation kills these fresh bone marrow cells or reduces their fitness, which, in turn, prolongs the time that older T cell progenitors already in the thymus survive and self-renew. This, along with the increased proliferative cycles of the older T cell progenitors needed to maintain production of mature T cells, results in a corresponding increase in the oncogenic mutations that they accumulate and a concomitant increase in lymphomagenesis (45). Replenishing dead or damaged bone marrow cells by transplantation or preventing their damage through shielding suppresses lymphomagenesis.

At the 3-Gy dose of -rays used in this study, all of the bone marrow cells are uniformly irradiated. This is not the case for HZE particle radiation. The average diameter of a murine bone marrow cell nucleus is around 6 m (46). At the fluence of HZE ions used in this study, the probability that a 6-m-diameter nucleus will be traversed by a 28Si ion and a 56Fe particle is 0.88 and 0.40, respectively. On the basis of a Poisson distribution, the probabilities of a nucleus not being traversed at all are 0.41 and 0.67 for 28Si and 56Fe irradiation, respectively. Thus, many of the T cell progenitors in the bone marrow are not irradiated (although they receive a small dose from -rays). These cells should exert a protective effect similar to transplanting unirradiated bone marrow cells or shielding some of the bone marrow during irradiation, rendering HZE ions less efficient for lymphomagenesis. Given that most of the pre-T LL in the HZE ionirradiated group are likely spontaneous, it is expected that they cluster more closely to spontaneous pre-T LL than to -rayinduced pre-T LL.

The mechanism leading to murine tumors of ovarian surface epithelium origin is well understood. Loss of primordial follicle oocytes by radiation-induced apoptosis results in a decrease in estrogen production, which, in turn, leads to elevated levels of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in the circulation. FSH and LH drive proliferation of ovarian surface epithelium cells (47). Ovarian tumors can be induced in some animal models by artificially manipulating levels of these hormones (4749). Irradiated mice can be protected from tubulostromal adenomas and granulosa cell tumors by shielding one ovary during irradiation or by transplanting the mice with an unirradiated ovary (50, 51); these interventions protect some oocytes and thereby maintain proper regulation of FSH and LH levels.

Assuming that the target cells are primordial follicle oocytes with a diameter of 12 m, the probabilities of no traversals are 0.2 for 56Fe and 0.03 for 28Si at the 0.4-Gy dose used here. The probabilities for one or fewer traversals are 0.52 for 56Fe and 0.14 for 28Si. Whether a sufficient number of follicles survive at 0.4 Gy to account for the observed ovarian tumor sparing is unknown. Mishra and colleagues (52) observed a dose-dependent decrease in primordial stage follicles in C57BL/6 mice 8 weeks after irradiation with 56Fe ions (600 MeV/n). Sixteen percent of the follicles survived at the 0.3-Gy dose, and normal levels of serum FSH and LH were present; at 0.5 Gy, only 1% of the follicles survived and an increase in serum FSH was observed. Caution is needed in using Mishras results in interpreting our own since we used mice with different genetic backgrounds and the FSH and LH levels in the 0.3 Gyirradiated mice may increase relative to unirradiated controls if time points beyond 8 weeks are assayed. In any event, microdosimetric effects should be incorporated into any risk model for tumors in which cell killing plays a prominent role.

The location of the chromosome 2 QTL in a region frequently deleted in radiogenic AMLs may be happenstance, but there are scenarios in which its chromosomal location would be crucial to its function. One possibility is that the polymorphism increases the frequency of AML-associated chromosome 2 deletions in irradiated hematopoietic cells by controlling the spatial confirmation of the chromosome such that the proximal and distal deletion breakpoints are in close proximity to one another (46). This type of proximity mechanism has been evoked to explain recurrent chromosomal rearrangements seen in radiation-induced papillary thyroid carcinoma and some spontaneous cancers (53, 54). In this scenario, the QTL could be a structural polymorphism (e.g., segmental duplication or interstitial telomeric sequence), which would affect chromosomal conformation, yielding a different conformation in susceptible mouse strains than resistant strains. Structural polymorphisms are easily missed in the assembly of the strain-specific genomic sequences used for mapping studies, so we would be unaware of its existence. A second possibility is that the polymorphism is in a gene needed for myeloid progenitor cell survival. Mouse strains resistant to myeloid leukemia would have a hypomorphic allele of this gene. If one copy is lost (i.e., through radiation-induced deletion), then the remaining copy would be insufficient for cell survival. Thus, in mouse strains resistant to radiogenic AML, a chromosome 2 deletion, which is the first step in radiation leukemogenesis, is a lethal event and leukemogenesis is thereby halted. Susceptible strains would have a fully functional allele of the gene, so that if one copy is deleted, the remaining copy maintains cell viability, allowing further leukemogenic events to occur (46). A caveat to both the chromosome conformation and haploinsufficiency scenarios is that the chromosome 2 deletions mapped in radiogenic AMLs from the F1 progeny of AML-susceptible CBA/H mice and AML-resistant C57BL/6 mice do not occur preferentially in the CBA/H origin chromosome (55). However, in that study, only 10 tumors were informative. In addition, susceptibility to radiogenic AML is multigenic, so it is possible that the difference in susceptibility between the CBA/H and C57BL/6 strains is not due to the chromosome 2 QTL.

HZE ions seem particularly effective in inducing Harderian gland tumors at the doses used in this study. This result was expected on the basis of extensive published radiation quality data on these tumors (8, 38). The mechanism responsible for higher tumorigenic efficacy of HZE ions relative to -rays is unknown; however, we have identified a QTL associated with Harderian gland adenocarcinoma following HZE ion exposures that does not appear to lend susceptibility to the same tumor following -ray exposures (Fig. 4C). Furthermore, HZE ioninduced Harderian gland adenomas and adenocarcinomas cluster away from spontaneous and -rayinduced Harderian gland tumors (Fig. 5), indicating non-overlap of some of the susceptibility loci. There are data that suggest that HZE ion irradiation has an effect on tumor promotion that -ray irradiation lacks. The observation is that pituitary isografts, which result in elevated levels of pituitary hormones, enhance the induction of Harderian gland tumors and decrease their latency in mice irradiated with -rays or fission neutrons but do not increase tumor prevalence in mice irradiated with 56Fe ions (600 MeV/n) (12). This would explain the high relative biological effectiveness (RBE) for 56Fe ions. It would also render QTLs that act in the promotion of -ray and spontaneous tumors irrelevant to HZE ioninduced tumors.

The use of unsupervised clustering on genome-wide association results is a novel approach to search for shared tumorigenic mechanisms between radiogenic and spontaneous tumors or between tumors induced by different radiation qualities. Potentially, the results could be used to inform risk modeling. For example, using the 99% confidence interval as a cutoff, thyroid adenomas, pituitary tumors, osteosarcomas, B cell lymphoblastic leukemia, mammary tumors, and hepatocellular carcinomas cluster by histotypes regardless of whether they arose in HZE ionirradiated or -rayirradiated mice. Of these, the incidences of thyroid tumors, pituitary tumors, and osteosarcomas are significantly increased following exposures to either HZE ions or -rays. Taking pituitary adenoma as an example, these findings suggest that it would be reasonable to extrapolate the risk of HZE ioninduced pituitary adenoma as a multiple of -rayinduced pituitary adenoma risk (i.e., using a relative risk model). Because there were too few spontaneous pituitary adenomas to position them on the dendrogram, we cannot determine whether the risk of HZE ioninduced pituitary adenoma could reasonably be modeled on the basis of the incidence of the spontaneous tumor. Another pattern of association is observed for Harderian gland adenoma and follicular B cell lymphoma in which, at the 99% confidence interval, spontaneous tumors cluster with -rayinduced tumors but not with HZE ioninduced tumors. There are a number of ways that this could occur. Three possibilities are as follows: (i) HZE ions act through a tumorigenic mechanism different from that of spontaneous and -rayinduced tumors. (2) HZE ions bypass the need for one or more of the genetically controlled steps required for spontaneous and -rayinduced tumors, and (iii) there are multiple pathways to tumor formation, and HZE ion irradiation forces tumorigenesis through only one (or a subset) of them. Harderian gland tumors may fall into the second possibility. As described earlier, observations on mice receiving pituitary isografts before irradiation suggest that HZE ions may have Harderian gland tumor promotion effects that -rays lack. If so, the QTL controlling those effects would be inconsequential in the tumorigenesis of HZE ioninduced Harderian gland tumors, and those tumors would cluster away from their spontaneous and -rayinduced counterparts. Whether a relative risk model, an absolute risk model, or a combination of the two would be most appropriate in Harderian gland tumor risk calculations would depend on which of the above possibilities is most accurate.

NASA seeks to limit the risk of exposure-induced death (REID) from radiogenic cancer to below 3% (56). For multiple missions aboard the International Space Station (flown in solar minimum conditions), the model projects that males will exceed permissible exposure limits at 24 months and females, at 18 months; women are considered at greater risk for radiogenic cancers than men because of longer life spans and increased susceptibility to specific cancer types, including lung, ovarian, and breast carcinomas. Because the 3% REID is derived from the upper 95% confidence interval for the risk estimate (57), decreasing the uncertainty for space radiationinduced cancers can significantly increase the flight time allowed for astronauts. The 95% confidence interval surrounding the risk estimates not only primarily reflects uncertainties in our understanding of HZE ions but also includes uncertainties surrounding dose-rate effects, transfer of risk between human populations, space dosimetry, and errors in the existing human epidemiology data. Concerning sex predilections, our results also demonstrate a sex difference in carcinogenesis risk, where female mice are at greater risk for radiogenic cancers than males, following either HZE ion or -ray exposures. These results are consistent with the current NASA model to calculate cancer risk from space radiation exposures (5).

Whether genotypic assays of radiosensitivity can improve the precision of risk assessment in humans will depend on a number of factors. One is the extent to which heritable sequence variants determine cancer risk from HZE ion exposures. HZE ion radiation exposures result in more complex molecular lesions that are less amenable to repair (58). Thus, it could be argued that sequence variants that result in subtle differences in DNA repair and damage response pathways would have a lesser impact on HZE ion radiation carcinogenesis. However, this work demonstrates that genetic susceptibility does indeed have a significant role in tumorigenesis following HZE ion exposures. Personalized approaches to cancer risk assessments may eventually allow for greater reductions in uncertainties when generating space radiation cancer risk estimates (28).

There are limitations to a mouse carcinogenesis study comparing acute -ray and HZE ion exposures. First, for cost efficiency and logistics reasons, a single dose was used for each radiation quality: 3.0 Gy for -ray exposures and 0.4 Gy for HZE ion exposures. Preliminary studies have demonstrated that these doses produce the maximum tumor incidence in inbred strains (24). Because tumor susceptibility and association mapping were the primary goals of this study, doses were chosen with the goal of generating the greatest tumor incidences and, therefore, the greatest power to detect significant QTL. However, caution must be taken when comparing the two single-dose groups, as it is impossible to untangle dose responses in such a study. An additional benefit of the selected doses is that 0.4 Gy of HZE ions represents a realistic dose, received over 20 to 30 months, for a flight crew traveling to Mars. Second, the applicability of these findings to human populations is limited, as rodents serve only as models of carcinogenesis.

The results presented here indicate that host genetic factors dictate risk for tumor development following radiation exposures, regardless of radiation quality. Therefore, at a population level, risks can be extrapolated from terrestrial exposures to the space radiation environment and at an individual level, and humans harboring susceptibility alleles for radiation-induced tumors developed on Earth are also likely at increased risk in space.

Male and female HS/Npt mice (n = 1850) were generated from breeding pairs obtained from Oregon Health and Sciences University (Portland, OR). The mice were group-housed (five mice of the same sex per cage) in a climate-controlled facility at 70F (21.1C) with free access to food (Teklad global rodent diet 2918) and sterile water and a 12-hour light cycle. Mice were shipped to Brookhaven National Laboratories (Upton, NY) where they were exposed to accelerator-produced HZE ions at the NASA Space Radiation Laboratory at 7 to 12 weeks of age. HS/Npt stock mice of both sexes were exposed to 0.4 Gy of 28Si ions (240 MeV/n) (n = 308) or 56Fe ions (600 MeV/n) (n = 314), 3 Gy of 137Cs -rays (n = 615), or sham irradiated (n = 622). Following irradiation exposure or sham irradiations, mice were returned to Colorado State University (Fort Collins, CO) and monitored twice daily for the duration of the study. The mice were evaluated for cancer development until they reached 800 days of age or became moribund. All animal procedures were approved by the Colorado State University Institutional Animal Use and Care Committee.

This study uses a highly recombinant mouse population (HS/Npt stock) that is genetically diverse and designed for genome mapping (1921, 23). HS/Npt mice are a multiparent cross derived from eight inbred strains (A/J, AKR/J, BALBc/J, CBA/J, C3H/HeJ, C57BL/6J, DBA/2J, and LP/J); each individual contains a unique mosaic of founder haplotypes and a high degree of heterozygosity, and recombination events become increasingly dense with each generation. Our population of HS/Npt mice was obtained from generation 71 of circular outbreeding.

DNA was isolated from tail biopsies taken from each mouse at 9 to 10 weeks of age. DNA was extracted and purified (QIAGEN, catalog no. 69506) according to the manufacturers instructions. GeneSeek (Lincoln, NE) performed genotyping assays using the Mega Mouse Universal Genotyping Array (MegaMUGA) (59) for a total of 1878 mice (including 28 inbred mice representing the founder strains). The MegaMUGA is built on the Illumina Infinium platform and consists of 77,808 single-nucleotide polymorphic markers that are distributed throughout the genome with an average spacing of 33 kb.

The heterogeneous stock mice are descendants of eight inbred founder strains. For each mouse, allele calls from the MegaMUGA array were used to calculate descent probabilities using a hidden Markov model (HMM), in which the hidden states were the founder strains and the observed data were the genotypes. The HMM generates probabilistic estimates of the diplotype state(s) for each marker locus and produces a unique founder haplotype mosaic for each mouse (18).

For this lifetime carcinogenesis study, all disease states were interpreted within the context of a systematic pathologic evaluation directed by board-certified veterinary pathologists (E.F.E. and D.A.K.). Structured necropsy and tissue collection protocols were followed for each mouse and involved photodocumentation of all gross lesions, collection of frozen tumor material, and preservation of tumor material in RNAlater. All tissues were grossly evaluated for all mice. To evaluate brain tissues and Harderian glands, craniums were decalcified for 48 hours in Formical-4 (StatLab, McKinney, TX 75069, product 1214) and five coronal sections of the skull were reviewed for each mouse. All gross lesions were evaluated microscopically and fixed in 10% neutral-buffered formalin and paraffin-embedded, and 5-m sections were stained with hematoxylin and eosin (H&E) and evaluated by a veterinary pathologist. For mice with solid tumors, all lung fields were examined histologically to detect the presence or absence of micrometastases. Tumor nomenclature was based on consensus statements produced by the Society of Toxicologic Pathology for mouse tumors (www.toxpath.org/inhand.asp). Representative histologic images routinely stained with H&E are presented in figs. S2 (A to E) and S3 (A and B).

Tissue microarrays were constructed to immunophenotype and subcategorize lymphoid neoplasms, which were the most commonly diagnosed tumors in irradiated and unirradiated HS/Npt mice. Identification of tissue sampling regions was performed by a veterinary pathologist. For each case, duplicate cores were taken from multiple anatomic locations (lymph nodes, spleen, thymus, etc.). Thirteen tissue microarrays were created, each of which contained six cores of control tissue at one corner of the array (haired skin, spleen, thymus, or liver); these control tissues were present in a unique combination and allowed for (i) orientation of the resulting sections, (ii) verification that the slide matched the block, and (iii) positive controls for immunohistochemistry. Figure S3D illustrates one tissue microarray as well as the resulting immunohistochemistry results for one thymic lymphoma (fig. S3E) and a core containing normal spleen (fig. S3F). Immunohistochemistry for T cell identification was performed using a rabbit monoclonal, anti-CD3 (SP7) antibody obtained from Abcam (ab16669; 1:300). Immunohistochemistry for B cell identification was performed using two rabbit monoclonal antibodies: an anti-CD45 antibody (ab10558; 1:1000) and an anti-PAX5 antibody (ab140341; 1:50). All immunohistochemistry was performed on a Leica BOND-MAX autostainer with the Leica BOND Polymer Refine Red Detection system (Leica DS9390, Newcastle Upon Tyne, UK). In addition to defining the immunophenotype, lymphomas were characterized according to the Mouse Model of Human Cancer Consortiums Bethesda protocols (60). For these protocols, anatomic location is important for the final diagnosis, and therefore, lymph node involvement was used from necropsy reports when necessary. Additional features included cell size, nuclear size, chromatic organization, and mitotic figure frequency, and the presence or absence of a leukemic phase was defined by bone marrow involvement within the sternum or femur. The most common lymphoma subtypes (fig. S4A) were evaluated for survival (fig. S4B), and pre-T LL typically presented with early-onset and large thymic masses.

Droplet digital polymerase chain reaction (ddPCR) was performed on cases of AML to assess deletion status via copy number variation for two genes: Spi1 and Asxl1. These genes are both located on chromosome 2 at base pair locations 91,082,390 to 91,115,756 for Spi1 and 153,345,845 to 153,404,007 for Asxl1. To establish a reference for normal diploid copy number in each AML sample, the copy number of H2afx was also determined. H2afx is located on chromosome 9, and deletions in this region have not been reported in murine AML. Bio-Rad PrimePCR probes were used for all assays as follows: Asxl1 ddPCR probe (dMmuCPE5100268), Spi1 ddPCR probe (dMmuCPE5094900), and H2afx ddPCR probe (dMmuCPE5104287). Ratios were created between the test gene and the reference gene (Spi1:H2afx and Asxl1:H2afx) to determine copy number with the assumption that the reference gene would not be deleted or amplified. Ideally, ratios of 1:1 represent equal copy numbers for both the test gene and the reference gene, and ratios of 1:2 represent a deletion in one copy of the test gene. However, since the tumor samples contained neoplastic cells as well as stromal cells and other cells, the ideal 1:2 ratio was not commonly observed. This is because stromal cells, which occur at unknown proportions in each tumor and which should not have chromosomal deletions, artificially increase ratios for tumor samples in which a deletion is indeed present. To account for stromal cell contamination, a cutoff ratio of 3:4 was established. Tumor samples with ratios below 3:4 were considered to have a deletion in one copy of the test gene.

For cases in which a solid tumor was identified, a standard section containing all lung lobes was processed and evaluated histologically. In cases where pulmonary metastases were observed, whole-slide scanning was performed at 200 magnification using an Olympus VS120-S5 and the OlyVIA software suite (www.olympusamerica.com/) to generate images for quantification of metastatic density (fig. S5). An analysis software, ImageJ (https://imagej.nih.gov/ij/), was used to quantify the total area of normal lung and the total area of metastatic foci (fig. S5). Metastatic density is reported as a percentage of the total metastasis area divided by the total lung area.

Association mapping was performed using a mixed-effects regression model with sex and cohort as fixed effects and a random-effects term to adjust for relatedness between mice by computing a matrix of expected allele sharing of founder haplotypes for each pair of mice (22). Three statistical models were fit to account for the wide range of trait distributions in this study. A generalized linear regression model was fit for binomial distributions, such as neoplasia. Cox regression analysis was incorporated to model time-to-event distributions to evaluate genetic contributions to tumor latency. Following genome-wide association analyses, resample model averaging methods were used to identify QTL that are consistently reproduced within subsamples of the mapping population.

Thresholds were determined using a permutation procedure in which the genotypes were fixed and the phenotype values were rearranged randomly within each sex. The distribution of the maximum negative log(P value) of association under the null hypothesis that no associations exist (null model) was determined for each genome scan with permuted data. One thousand permutations were performed for each phenotype in each radiation exposure group, simulating effects arising from covariates, the linkage disequilibrium structure of the genome, and effects due to phenotype distribution. A threshold was defined as an estimate of the genome-wide significance for which a type I statistical error will occur at a given frequency (29). Confidence intervals for each QTL were determined by nonparametric resample model averaging procedures using bootstrap aggregation with replacement. In this procedure, the mapping population is sampled to create a new dataset in which some individuals may be omitted and some may appear multiple times (30), and the locus with peak significance is recorded. Resampling is repeated 200 times for each phenotype to determine a 95% confidence interval for a given QTL. Effect sizes were calculated using the Tjur method for association mapping with logistic regression and pseudo-R2 for mapping with Cox proportional hazard regression. Statistical significance for each model was assessed using a permutation strategy to randomize genotypes via resampling without replacement and maintaining covariates. Permutation analysis was performed (1000 tests) for each trait and exposure group to generate estimations of genome-wide significance thresholds. As genome scans with hundreds of thousands of imputed SNPs are computationally intensive, parallel computing was essential and accomplished using spot instances of resizable Elastic Compute Cloud hosting resources.

Comparisons were made between whole-genome scans using Pearson correlations as a similarity measure with clustering based on average linkage. Significance of clustering results was estimated with 10,000 random permutations of the dataset (log10 values permuted with genetic markers) to determine a distribution of dendrogram heights under the null hypothesis that no associations exist. Each permutated dataset simulates a null distribution of the maximally significant clustering based on a randomly assorted set of P values for each genomic locus.

Bootstrap aggregation is a resample model averaging procedure that has been demonstrated to produce highly accurate estimates of QTL in structured populations (32). The procedure is relatively simple: for a genome-wide association study (GWAS) of n individuals, a sampling of n draws is obtained, with replacement, from the observed individuals to form a new dataset in which some individuals are omitted and some appear multiple times. For each new dataset created this way, an estimate of the QTL location is calculated. This process is repeated many times and is the basis for determining a confidence interval for a given result. The use of bootstrap procedures is commonly used this way to estimate QTL support intervals in experimental crosses; however, this statistical method can potentially be applied to other areas of QTL research, including comparative QTL mapping.

When an identical QTL is observed for two distinct traits, one explanation is that a single gene is involved for two distinct biologic processes, also known as pleiotropy. This was sometimes assumed in early mouse QTL studies that resulted in coincident loci for distinct traits. Another possibility, however, is that two distinct genetic variants are present in close proximity, each independently contributing to the two phenotypes. Because the two hypothetical genetic variants happen to be in close proximity, they are difficult to distinguish in low-resolution mapping studies. Using resample model averaging in highly recombinant mice is proposed to best differentiate precise locations of the QTL; if the same markers were repeatedly identified, then the case for pleiotropy was strengthened. For comparative QTL mapping in tumorigenesis studies, nonparametric resample model averaging could similarly be leveraged to identify whether the same QTL renders an individual susceptible to distinct environmental carcinogens. One significant advantage to using bootstrap procedures to detect potential coincident loci is that comparisons can be made between groups based on the identification of a highly significant QTL identified in only one exposure group (e.g., at a false-positive rate of 1 per 20 scans). This QTL may be present in the alternative exposure group, but at lower confidence (e.g., at a false-positive rate of 1 per 10 scans), and therefore discarded in a typical GWAS. A diagrammatic representation of the comparative QTL bootstrap procedure is presented in fig. S6. Because the resultant genetic positions derived from bootstrapping are composed of the most significant locus for each resampling regardless of the significance level for the mapping procedure, comparisons can be drawn between QTL that might have been discarded on the basis of the stringent statistical demands of an assay involving hundreds of thousands of independent tests. Using this procedure on thyroid tumors demonstrates that the same loci are consistently identified whether exposed to particle or photon irradiation (Fig. 2E). Using the comparative QTL procedure described, it can be determined whether an individuals cancer risk from one carcinogen will be predictive of that individuals cancer risk to another carcinogen. The application of this procedure is well illustrated by the space radiation problem, where much is known about -ray exposures and little is known about space radiation exposures.

In addition to looking for similarities between individual selected QTL for HZE ion and -rayexposed populations, we also sought a more holistic method in which entire genome scans could be compared between groups in an unsupervised process. By using entire genome scans, we submit for comparison not only highly significant regions but also the numerous loci detected with lower confidence. To determine similarity of genetic association profiles for all phenotypes and to detect possible coincident QTL, clustering procedures were used to compare genome-wide association scans between different radiation exposure groups. To demonstrate and validate the methodology of QTL clustering, genome-wide scans for coat colors in each treatment group are evaluated (Fig. 5B). As expected, genome-wide scans for coat color are unaffected by radiation exposures, and therefore, clustering is based entirely on coat phenotype rather than radiation exposure group. Using the same procedure for neoplasia indicates that tumor types often clustered together as well, regardless of radiation exposure (Fig. 5A). Genome scans for thyroid tumors and mammary adenocarcinomas in radiation-exposed groups and all hepatocellular carcinoma genome scans cluster together. This finding supports the hypothesis that host genetic factors are more important in determining neoplasm incidence than radiation exposure type. Unlike other statistic procedures, such as regression models, clustering lacks a response variable and is not routinely performed as a formal hypothesis test. Therefore, determining the significance of a clustering result can be problematic, as no consensus method exists for cluster validation. Permutation analysis provides the distribution of clustering results that will randomly occur from a given dataset; this can then be used as a baseline from which to determine a significance level on a given dendrogram tree [green line in Fig. 5 (A to C)]. While the overall validity of a given cluster can be accomplished by cluster permutation analysis, no method is identified to estimate the number of clusters that should be present in a dataset. Furthermore, methods to determine the significance of specific subset of objects clustering together do not exist; in such cases, the permutation threshold is likely overly stringent.

See more here:
Genomic mapping in outbred mice reveals overlap in genetic susceptibility for HZE ion and -rayinduced tumors - Science Advances

Read More...

Article on COVID-19 in Iceland in New England Journal of Medicine – Iceland Monitor

Saturday, April 18th, 2020

An article on the spread of the novel coronavirus in Iceland was published by the The New England Journal of Medicine yesterday. The authors are scientist at deCode Genetics and their colleagues at the Directorate of Health and at Landsptali National University Hospital, mbl.is reports.

At the website of deCode, the aim of the study is explained:

The aim of the study was to provide as comprehensive a view as possible of how the virus spreads in a population, in this case one of 360,000 and implementing early and aggressive testing, tracking and isolation measures to contain the epidemic.

The results show that roughly 0.8 percent of the population at large is infected with several strains or clades of the virus supporting the concern that silent carriers spread the disease.

This suggests that while the efforts of the public health system have been effective so far in mitigating the spread to date, more data, including massive population screening, will be key to informing efforts to contain the virus in Iceland in the long run.

In attempting to carefully map the molecular epidemiology of COVID-19 in Iceland we hope to provide the entire world with data to use in the collective global effort to curb the spread of the disease, Kri Stefnsson, CEO of deCode Genetics is quoted as saying.

Kri states that widespread screening (ten percent of the nation has already been tested for the virus) and measures taken by the Directorate of Health to slow the spread of the virus give a good example of how this dangerous pandemic can be fought.

General screening in Iceland by deCode began March 13, where anyone who wanted could sign up for screening. By March 31, 10,797 individuals had been tested, 87 of whom tested positive for the virus. Subsequently, 2,283 peoople, randomly chosen, were tested, with a similar result.

After that, a sequence analysis of 643 positive tests was done, showing that the first cases originated in Italy and Austria, but later on, transmissions were traced to other countries.

Currently, 291 mutations of the virus have been found in the country that have not been identified elsewhere, according to decode.com.

See the rest here:
Article on COVID-19 in Iceland in New England Journal of Medicine - Iceland Monitor

Read More...

Solving the Puzzle of Mitchell Disease – Technology Networks

Saturday, April 18th, 2020

When a patient with puzzling neurological symptoms enrolled in the Undiagnosed Diseases Network, researchers led by Dr. Hugo J. Bellen were set on solving the mystery. The patient presented with an unidentified late-onset neurodegenerative disorder. The team named this new syndrome "Mitchell disease" in reference to the first patient to be diagnosed with this disorder and looked to identify its genetic basis."On comparing the patient's and his parents' DNA, the team identified a mutation in the patient that resulted in a single amino acid substitution (N237S) in the ACOX1 protein. This change was seen only in the patient and was not present in either of his parents' DNA, indicating that the patient had a de novo, or new, mutation on this gene,' said Bellen, professor at Baylor College of Medicine and investigator at the Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital and also a Howard Hughes Medical Institute investigator. "With the help of the online gene-matching tool GeneMatcher, we found two more patients who had the same new mutation in the ACOX1 gene."

All three patients, who ranged from 3 to 12 years old at the time of disease onset, had remarkably similar clinical features, including degeneration of peripheral nerves that caused a progressive loss of mobility and hearing. The three individuals had identical gene variants, a clear indication that ACOX1 dysfunction likely was the cause of the symptoms.

"The brain has large amounts of lipids, which are critical for the proper functioning of the nervous system. Abnormal breakdown of lipids in the brain and peripheral nervous system is associated with several neurodegenerative diseases," Bellen said.

The gene ACOX1 is involved in lipid breakdown. It produces an enzyme called Acyl-CoA oxidase 1 that initiates a series of reactions that break down very-long-chain-fatty acids in small intracellular organelles called peroxisomes.

To understand how ACOX1 variants affect the function of glia, they generated two mutant fly lines, the first one lacked both the copies of ACOX1 gene and the second, carried the substitution mutation (N237S) found in one of the ACOX1 genes in the Mitchell disease patients.

"Flies lacking ACOX1 mimicked the symptoms of ACOX1 deficiency in humans, including elevated levels of very-long-chain-fatty acids along with dramatic loss of glia and neurons and progressively impaired neuronal function. When we reduced the synthesis of very-long-chain-fatty acids in these flies by administering the drug bezafibrate, we observed significant improvement in lifespan, vision, motor coordination and neuronal function, implicating elevated levels of these lipids and their excessive accumulation in glia as an important contributor," said Chung, postdoctoral fellow in the Bellen lab.

"It is remarkable how well bezafibrate suppressed the symptoms of ACOX1 deficiency, suggesting a new therapeutic avenue for patients with this condition," Bellen said.

In contrast to the loss of ACOX1, the introduction of the single amino acid substitution (N237S) in ACOX1 gene resulted in a hyperactive ACOX1 protein. Typically, breakdown of very-long-chain-fatty acids by the enzymatic action of ACOX1 produces small amounts of highly reactive oxygen species, but glial cells quickly neutralize them. However, in Mitchell's disease, hyperactive ACOX1 produces copious amounts of toxic reactive oxygen species, leading to the destruction of glia and their neighboring neurons.

The harmful effects due to hyperactive ACOX1 were potently reversed with the antioxidant N-acetyl cysteine amide (NACA). However, NACA did not suppress the lethality or toxic effects in flies that lacked ACOX1, a clear indication that the two diseases act via entirely different pathways and would need to be treated with two distinct therapeutic strategies.

"This study is a prime example of how combining UDN's unique team science approach with power of fruit fly genetics is facilitating rapid and phenomenal progress in rare diseases research. We take on cases of patients with conditions never described before, uncover new diseases and find definitive molecular diagnosis for them. We make significant progress in unraveling the causes of these novel diseases and rapidly identify and test promising new treatment options," Bellen said. "We have successfully identified more than 25 disease-causing genes within the past three years - a task that typically takes many years."ReferenceChung et al. (2020) Loss- or Gain-of-Function Mutations in ACOX1 Cause Axonal Loss via Different Mechanisms. Neuron. DOI: https://doi.org/10.1016/j.neuron.2020.02.021

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

See the article here:
Solving the Puzzle of Mitchell Disease - Technology Networks

Read More...

Bill Of The Month: Pricey Genetic Test For Essential Thrombocythemia : Shots – Health News – NPR

Wednesday, April 1st, 2020

Michelle Kuppersmith's doctor recommended a bone marrow biopsy after suspecting she had a rare blood disorder. Though the biopsy was done by an in-network provider at an in-network hospital, Kuppersmith learned she was on the hook for $2,400 for out-of-network genetic profiling. Shelby Knowles for KHN hide caption

Michelle Kuppersmith's doctor recommended a bone marrow biopsy after suspecting she had a rare blood disorder. Though the biopsy was done by an in-network provider at an in-network hospital, Kuppersmith learned she was on the hook for $2,400 for out-of-network genetic profiling.

Michelle Kuppersmith feels great, works full time and exercises three to four times a week. So she was surprised when a routine blood test found that her body was making too many platelets, which help control bleeding.

Kuppersmith's doctor suspected the 32-year-old Manhattanite had a rare blood disorder called essential thrombocythemia, which can lead to blood clots, strokes and, in rare cases, leukemia.

Her doctor suggested a bone marrow biopsy, in which a large needle is used to suck out a sample of the spongy tissue at the center of the patient's hip bone.

Doctors examine the bone marrow under a microscope and analyze the DNA. The procedure allows doctors to judge a patient's prognosis and select treatment, if needed. Kuppersmith had heard the procedure can be intensely painful, so she put it off for months.

The biopsy performed by a provider in her insurance network, at a hospital in her network lasted only a few minutes, and Kuppersmith received relatively good news.

While a genetic analysis of her bone marrow confirmed her doctor's suspicions, it showed that the only treatment she needs, for now, is a daily, low-dose aspirin. She will check in with her doctor every three to four months to make sure the disease isn't getting worse.

All in all, Kuppersmith felt relieved.

Then she got a notice saying her insurer refused to pay for the genetic analysis, leaving her responsible for a $2,400 payment.

The patient: New York resident Michelle Kuppersmith, 32, who is insured by Maryland-based CareFirst Blue Cross Blue Shield. She works as director of special projects at a Washington-based watchdog group. Because she was treated in New York, Empire Blue Cross Blue Shield which covers that region handled part of her claim.

Total amount owed: $2,400 for out-of-network genetic profiling

The providers: Kuppersmith had her bone marrow removed at the Mount Sinai Ruttenberg Treatment Center in New York City, which sent her biopsy sample to a California lab, Genoptix, for testing.

Medical services: Bone marrow biopsy and molecular profiling, which involves looking for genetic mutations

What gives: The field of molecular diagnostics, which includes a variety of gene-based testing, is undergoing explosive growth, said Gillian Hooker, president of the National Society of Genetic Counselors and vice president of clinical development for Concert Genetics, a health IT company in Nashville, Tennessee.

A report from Concert Genetics, a company that helps clients manage genetic testing, found there are more than 140,000 molecular diagnostic products on the market, with 10 to 15 added each day.

The field is growing so quickly that even doctors are struggling to develop a common vocabulary, Hooker said.

Kuppersmith underwent a type of testing known as molecular profiling, which looks for DNA biomarkers to predict whether patients will benefit from new, targeted therapies. These mutations aren't inherited; they develop over the course of a patient's life, Hooker said.

Medicare spending on molecular diagnostics more than doubled from 2016 to 2018, increasing from $493 million to $1.1 billion, according to Laboratory Economics, a lab industry newsletter.

Charges range from hundreds to thousands of dollars, depending on how many genes are involved and which billing codes insurers use, Hooker said.

Based on Medicare data, at least 1,500 independent labs perform molecular testing, along with more than 500 hospital-based labs, said Jondavid Klipp, the newsletter's publisher.

In a fast-evolving field with lots of money at stake, tests that a doctor or lab may regard as state-of-the-art an insurer might view as experimental.

Worse still, many of the commercial labs that perform the novel tests are out-of-network, as was Genoptix.

Stephanie Bywater, chief compliance officer at NeoGenomics Laboratories, which owns Genoptix, said that insurance policies governing approval have not kept up with the rapid pace of scientific advances. Kuppersmith's doctor ordered a test that has been available since 2014 and was updated in 2017, Bywater said.

Although experts agree that molecular diagnostics is an essential part of care for patients like Kuppersmith, doctors and insurance companies may not agree on which specific test is best, said Dr. Gwen Nichols, chief medical officer of the Leukemia & Lymphoma Society.

Tests "can be performed a number of different ways by a number of different laboratories who charge different amounts," Nichols said.

Insurance plans are much more likely to refuse to pay for molecular diagnostics than other lab tests. Laboratory Economics found Medicare contractors denied almost half of all molecular diagnostics claims over the past five years, compared with 5-10% of routine lab tests.

With so many insurance plans, so many new tests and so many new companies, it is difficult for a doctor to know which labs are in a patient's network and which specific tests are covered, Nichols said.

"Different providers have contracts with different diagnostic companies," which can affect a patient's out-of-pocket costs, Nichols said. "It is incredibly complex and really difficult to determine the best, least expensive path."

Kuppersmith said she has always been careful to check that her doctors accept her insurance. She made sure Mount Sinai was in her insurance network, too. But it never occurred to her that the biopsy would be sent to an outside lab or that it would undergo genetic analysis.

She added: "The looming threat of a $2,400 bill has caused me, in many ways, more anxiety than the illness ever has."

The resolution: Despite making dozens of phone calls, Kuppersmith got nothing but confusing and contradictory answers when she tried to sort out the unexpected charge.

An agent for her insurer told her that her doctor hadn't gotten preauthorization for the testing. But in an email to Kuppersmith, a Genoptix employee told her the insurance company had denied the claim because molecular profiling was viewed as experimental.

A spokesperson for New York-based Empire Blue Cross Blue Shield, which handled part of Kuppersmith's claim, said her health plan "covers medically necessary genetic testing."

New York, one of 28 states with laws against surprise billing, requires hospitals to inform patients in writing if their care may include out-of-network providers, said attorney Elisabeth Benjamin, vice president of health initiatives at the Community Service Society, which provides free help with insurance problems.

A spokesperson for Mount Sinai said the hospital complies with that law, noting that Kuppersmith was given such a document in 2018 nearly one year before her bone marrow biopsy and signed it.

Benjamin said that's not OK, explaining: "I think a one-year-old, vague form like the one she signed would not comply with the state law and certainly not the spirit of it."

Instead of sending Kuppersmith a bill, Genoptix offered to help her appeal the denied coverage to CareFirst. At first, Genoptix asked Kuppersmith to designate the company as her personal health care representative. She was uncomfortable signing over what sounded like sweeping legal rights to strangers. Instead, she wrote an email granting the company permission to negotiate on her behalf. It was sufficient.

A few days after being contacted by KHN, Kuppersmith's insurer said it would pay Genoptix at the in-network rate, covering $1,200 of the $2,400 charge. Genoptix said it has no plans to bill Kuppersmith for the other half of the charge.

The takeaway: Kuppersmith is relieved her insurer changed its mind about her bill. But, she said: "I'm a relatively young, savvy person with a college degree. There are a lot of people who don't have the time or wherewithal to do this kind of fighting."

Patients should ask their health care providers if any outside contractors will be involved in their care, including pathologists, anesthesiologists, clinical labs or radiologists, experts said. And check if those involved are in-network.

"Try your best to ask in advance," said Jack Hoadley, a research professor emeritus at Georgetown University. "Ask, 'Do I have a choice about where [a blood or tissue sample] is sent?'"

Ask, too, if the sample will undergo molecular diagnostics. Since the testing is still relatively new and expensive most insurers require patients to obtain "prior authorization," or special permission, said Dr. Debra Regier, a medical geneticist at Children's National Hospital in Washington and an associate with NORD, the National Organization of Rare Diseases. Getting this permission in advance can prevent many headaches.

Finally, be wary of signing blanket consent forms telling you that some components of your care may be out-of-network. Tell your provider that you want to be informed on a case-by-case basis when an out-of-network provider is involved and to consent to their participation.

Bill of the Month is a crowdsourced investigation by Kaiser Health News and NPR that dissects and explains medical bills. Do you have a perplexing medical bill you want to share with us? Tell us about it here.

Read this article:
Bill Of The Month: Pricey Genetic Test For Essential Thrombocythemia : Shots - Health News - NPR

Read More...

Researchers at U of T developing antibodies to ‘neutralize’ novel coronavirus before it invades cells – News@UofT

Wednesday, April 1st, 2020

Universityof Toronto researcherSachdev Sidhuand his collaborators are engineering antibody molecules that can neutralize the novel coronavirus in the body before it invades cells.

Sidhu (left) already leads a differentteam that received supportin the first round of federal funding. The goal of that project is to design antiviral medicines that block viral replication.

With our two funded projects, we are working to develop molecules that can target the virus both inside human cells and on the outside to prevent it from getting in, says Sidhu, who is a professor of molecular genetics in the Faculty of Medicine.

Rini has previously helped to determine how antibodies bind to and inactivate the SARS virus, the coronavirus that caused the outbreak in Asia more than 15 years ago. Also on the team isAlan Cochrane, a professor in the department of molecular genetics and an HIV virologist with expertise in viral RNA processing.

The antibodies will be engineered to block the so-called S-protein that forms spikes on the virus's surface. The spikes lock on to a protein called ACE2 on the surface of human cells to gain entry. Coating viral particles with synthetic antibodies should prevent the spikes from binding to ACE2.

Sidhu and Rini will also engineer antibodies that bind ACE2 to make it inaccessible to the virus. This type of engineered immunity surpasses the capacity of the bodys natural immune system since antibodies that react against self-proteins have been filtered out. If successful, the approach may obviate worries about viral mutations that can render drugs ineffective to new emerging viral strains becausethe host protein ACE2 does not change over time.

Sidhus team has advanced a technology called phage display to rapidly create and select human antibodies with desired biological properties, including blocking the virussspike protein. Over the last decade, his team has created hundreds of antibodies with therapeutic potential some of which are in clinical development through spin-off companiesand large pharmaceutical firms.

The group has demonstrated success with both approaches for inhibiting viral entry, having developed neutralizing antibodies that target the Ebola virus as well as antibodies that target the human host receptor of hantavirus or hepatitis C. Moreover, other research has shown that antibodies targeting SARS, a related virus whose genetic material is over 80 per cent identical to the one causing COVID-19, can clear infection in cells and mice.

Using phage display, in which tiny bacterial viruses called phages are instructed to create vast libraries of diverse antibodies, the team will select the antibodies that can kill the virus in human cells before testing them on mice and, eventually, patients. Experiments on mice could start within three to six months, Sidhu says.

In addition to creating antibodies tailored to the new virus from scratch, the researchers will also modify existing SARS-blocking antibodies so that they attack COVID-19 and provide an additional route to the development of a therapeutic.

Given the global spread of the virus, its possible that it will become endemic and circulate in the population like seasonal flu. And, like the flu, it could mutate into new strains that will evade acquired immunity and the vaccines that are being developed. By generating a panel of different antibodies, the researchers aim to stay one step ahead of the virus.

Our advances in antibody engineering technologiesand access to the complete genomes of the COVID-19 virus and its relatives provides us with an opportunity to create tailored therapeutic antibodies at a scale and speed that was not possible even a few years ago, says Sidhu.

Ultimately, we aim to optimize methods to the point where the evolution of new drugs will keep pace with the evolution of the virus itself, providing new and effective drugs in response to new outbreaks.

The rest is here:
Researchers at U of T developing antibodies to 'neutralize' novel coronavirus before it invades cells - News@UofT

Read More...

2020 World Molecular Diagnostic Analyzer and Reagent Forecasts for 100 Tests: Americas, EMEA, APAC–A 68-Country Analysis–Infectious and Genetic…

Wednesday, April 1st, 2020

NEW YORK, March 31, 2020 /PRNewswire/ -- 2020 World Molecular Diagnostic Analyzer and Reagent Forecasts for 100 Tests: Americas, EMEA, APAC--A 68-Country Analysis--Infectious and Genetic Diseases, Cancer, Forensic and Paternity Testing

Read the full report: https://www.reportlinker.com/p05876987/?utm_source=PRN

This new 68-country survey provides granular data and analysis not available from any other source. The report is designed to help current suppliers and potential market entrants identify and evaluate major business opportunities emerging in the molecular diagnostics market during the next five years.

Highlights

- Supplier sales and market shares in major countries

- Five-year test volume and sales forecasts

- Strategic profiles of market players and start-up firms developing innovative technologies and products

- Emerging technologies

- Review of molecular diagnostic analyzers

- Specific product and business opportunities for instrument and consumable suppliers

Rationale

The molecular diagnostics market is unquestionably the most rapidly growing segment of the in vitro diagnostics industry. The next five years will witness significant developments in reagent systems and automation, as well as introduction of a wide range of new products that will require innovative marketing approaches. The rate of market penetration into routine clinical laboratories, however, will depend on the introduction of cost-effective and automated systems with amplification methods.

In order to successfully capitalize on the opportunities presented by the molecular diagnostics market, many companies are already exploiting new molecular technologies as corporate strategic assets, managed in support of business and marketing strategies. Integrating new technology planning with business and corporate strategies will be one of the most challenging tasks for diagnostic companies during the next five years.

Geographic Regions

Asia-Pacific, Europe, Latin America, Middle East, North America

Country Analyses

Argentina, Australia, Austria, Bahrain, Bangladesh, Belgium, Brazil, Bulgaria, Canada, Chile, China, Colombia, Croatia, Cyprus, Czech Republic, Denmark, Estonia, Finland, France, Germany, Greece, Hong Kong, Hungary, Iceland, India, Indonesia, Iran, Iraq, Ireland, Israel, Italy, Japan, Jordan, Kuwait, Latvia, Lebanon, Lithuania, Malaysia, Malta, Mexico, Myanmar, Netherlands, New Zealand, Norway, Oman, Pakistan, Peru, Philippines, Poland, Portugal, Qatar, Romania, Saudi Arabia, Serbia, Singapore, Slovakia, Slovenia, South Korea, Spain, Sweden, Switzerland, Taiwan, Thailand, UK, United Arab Emirates, USA, Venezuela, Vietnam

Market Segmentation Analysis

- Sales and market shares of key suppliers of molecular diagnostic reagent kits and components in major markets.

Five-year test volume and sales forecasts for major applications, including:

- Infectious Diseases - Forensic Testing- Cancer - Paternity Testing/HLA Typing- Genetic Diseases - Others

- Five-year test volume and sales projections for over 30 NAT assays.

- A comprehensive analysis of the sequencing market, by country and laboratory segment,including:

- Industrial - Academic- Government- Commercial

- Market segmentation analysis, including review of the market dynamics, trends, structure, size, growth and suppliers in major countries.

Product/Technology Review

- Comparison of leading molecular diagnosticanalyzers marketed by Abbott, Beckman Coulter, BD, Bio-Rad, Gen-Probe, Roche, Tecan and other suppliers.

- Extensive review of molecular diagnostic technologies, test formats, detection methodologies, trends in testing automation and over 30 target/signal amplification methods, including:

- PCR - bDNA - SDA - NASBA - TMA - SSSR, and others - LCR

- Universities and research centers developing new molecular diagnostictechnologies and products.

Competitive Assessments

- Extensive strategic assessments of major suppliers and emerging market entrants, including their sales, product portfolios, marketing tactics, collaborative arrangements and new technologies/products in RandD.

- Companies developing and marketing molecular diagnostics products, by test and application.

Opportunities and Strategic Recommendations

Story continues

See the original post:
2020 World Molecular Diagnostic Analyzer and Reagent Forecasts for 100 Tests: Americas, EMEA, APAC--A 68-Country Analysis--Infectious and Genetic...

Read More...

UVA Finds Way to Improve Cancer Outcomes by Examining Patients’ Genes – University of Virginia

Wednesday, April 1st, 2020

By mining a vast trove of genetic data,researchers at theUniversity of Virginia School of Medicineare enhancing doctors ability to treat cancer, predict patient outcomes and determine which treatments will work best for individual patients.

The researchers have identified inherited variations in our genes that affect how well a patient will do after diagnosis and during treatment. With that information in hand, doctors will be able to examine a patients genetic makeup to provide truly personalized medicine.

Oncologists can estimate how a patient will do based on the grade of the tumor, the stage, the age of the patient, the type of tumor, etc. We found [adding a single genetic predictor] can improve our predictive ability by 5% to 10%, said UVAs Anindya Dutta. Many of the cancers had multiple inherited genetic change that were predictive of outcome, so if we add those in, instead of a 10% increase we might get a 30% increase in our ability to predict accurately how patients will do with our current therapy. Thats amazing.

Dutta, the chair of UVAs Department of Biochemistry and Molecular Genetics, believes reviewing the inherited genetic makeup of a patient can provide similar benefits for predicting outcome and choosing therapy for many, many other conditions, from diabetes to cardiac problems. As such, the approach represents a major step forward in doctors efforts to tailor treatments specifically to the individuals needs and genetic makeup.

The research offers answers to questions that have long perplexed doctors.Every clinician has this experience: Two patients come in with exactly the same cancersame grade, same stage, received the same treatment. One of them does very well, and the other one doesnt, Dutta said.The assumption has always been that there is something about the two that we didnt understand, like maybe there are some tumor-specific mutations that one patient had but the other did not. But it occurred to us that with all this genomic data, there is another hypothesis that we could test.

Instead of a 10% increase we might get a 30% increase in our ability to predict accurately how patients will do with our current therapy. Thats amazing.

- Anindya Dutta

To determine if genetic differences in the patients could be the answer, Dutta and his colleagues did a deep dive into the Cancer Genome Atlas, an enormous repository of genetic information assembled by the National Institutes of Healths National Cancer Institute. The researchers sought to correlate inherited genetic variations with patient outcomes.

This incredibly smart M.D.-Ph.D. student in the lab, Mr.Ajay Chatrath,decided that this was a perfect time to explore this, Dutta recalled. With the help of cloud computing services at UVA, we managed to download all this genomic sequencing data and identify what are known as germline variants not just tumor-specific mutations, but the mutations that were inherited from the parents and are present in all cells of the patient.

The researchers started small, but soon realized how quickly the work could be done and how big the benefits could be. Once we realized this was a very easy thing to do, we went on to do all 33 cancers and all 10,000 patients, and that took another six months, Dutta said. All of this came together beautifully. It was very exciting that every single member in the lab contributed to the analysis.

Dutta is eager to share his findings in hopes of finding collaborators and inspiring researchers and private industry to begin mining the data for other conditions. This is very low-hanging fruit, he said. Germline variants predicting outcome can be applicable to all types of diseases and not just cancer, and [they can predict] responsiveness to all types of therapy, and thats why Im particularly excited.

The researchers have published their findings in the scientific journal Genome Medicine. The studys authors were Chatrath, Roza Przanowska, Shashi Kiran, Zhangli Su, Shekhar Saha, Briana Wilson, Takaaki Tsunematsu,Ji-Hye Ahn, Kyung Yong Lee, Teressa Paulsen, Ewelina Sobierajska, Manjari Kiran, Xiwei Tang, Tianxi Li, Pankaj Kumar, Aakrosh Ratan and Dutta.

The research was supported by the National Institutes of Health, grants R01 CA166054, R01 1094 CA60499, T32 GM007267, AHA 18PRE33990261; and a Cancer 1095 Genomics Cloud Collaborative Support grant. The Seven Bridges Cancer 1096 Genomics Cloud has been funded by the National Cancer Institute, National Institutes of Health.

To keep up with the latest medical research news from UVA, subscribe to theMaking of Medicineblog.

Read this article:
UVA Finds Way to Improve Cancer Outcomes by Examining Patients' Genes - University of Virginia

Read More...

Her Genetic Test Revealed A Microscopic Problem And A Jumbo Price Tag – Bryan-College Station Eagle

Wednesday, April 1st, 2020

Michelle Kuppersmith, 32, feels great, works full time and exercises three to four times a week. So she was surprised when a routine blood test found that her body was making too many platelets, which help control bleeding. Kuppersmiths doctor suspected she had a rare blood disorder called essential thrombocythemia, which can lead to blood clots, strokes and, in rare cases, leukemia.

[khnslabs slabs="813866"]

Her doctor suggested a bone marrow biopsy, in which a large needle is used to suck out a sample of the spongy tissue at the center of the patients hip bone. Doctors examine the bone marrow under a microscope and analyze the DNA. The procedure allows doctors to judge a patients prognosis and select treatment, if needed. Kuppersmith had heard the procedure can be intensely painful, so she put it off for months.

The biopsy performed by a provider in her insurance network, at a hospital in her network lasted only a few minutes, and Kuppersmith received relatively good news. While a genetic analysis of her bone marrow confirmed her doctors suspicions, it showed that the only treatment she needs, for now, is a daily, low-dose aspirin. She will check in with her doctor every three to four months to make sure the disease isnt getting worse.

All in all, Kuppersmith felt relieved.

Then she got a notice saying her insurer refused to pay for the genetic analysis, leaving her responsible for a $2,400 payment.

The Patient: New York resident Michelle Kuppersmith, 32, who is insured by Maryland-based CareFirst Blue Cross Blue Shield. She works as director of special projects at a Washington-based, nonpartisan watchdog group. Because she was treated in New York, Empire Blue Cross Blue Shield which covers that region handled part of her claim.

Total Amount Owed: $2,400 for out-of-network genetic profiling

[khnslabs slabs="790331" view="inline" /]

The Providers: Kuppersmith had her bone marrow removed at the Mount Sinai Ruttenberg Treatment Center in New York City, which sent her biopsy sample to a California lab, Genoptix, for testing.

Medical Services: Bone marrow biopsy and molecular profiling, which involves looking for genetic mutations

What Gives: The field of molecular diagnostics, which includes a variety of gene-based testing, is undergoing explosive growth, said Gillian Hooker, president of the National Society of Genetic Counselors and vice president of clinical development for Concert Genetics, a health IT company in Nashville, Tennessee.

A Concert Genetics report found there are more than 140,000 molecular diagnostic products on the market, with 10 to 15 added each day.

The field is growing so quickly that even doctors are struggling to develop a common vocabulary, Hooker said.

Kuppersmith underwent a type of testing known as molecular profiling, which looks for DNA biomarkers to predict whether patients will benefit from new, targeted therapies. These mutations arent inherited; they develop over the course of a patients life, Hooker said.

[documentcloud url="http://www.documentcloud.org/documents/6815388-BOTM-March2020.html" responsive=true]

Medicare spending on molecular diagnostics more than doubled from 2016 to 2018, increasing from $493 million to $1.1 billion, according to Laboratory Economics, a lab industry newsletter.

Charges range from hundreds to thousands of dollars, depending on how many genes are involved and which billing codes laboratories use, Hooker said.

Based on Medicare data, at least 1,500 independent labs perform molecular testing, along with more than 500 hospital-based labs, said Jondavid Klipp, the newsletters publisher.

In a fast-evolving field with lots of money at stake, tests that a doctor or lab may regard as state-of-the-art an insurer might view as experimental.

Worse still, many of the commercial labs that perform the novel tests are out-of-network, as was Genoptix.

After lining up an in-network provider at an in-network hospital, Kuppersmith pushed back when she got a $2,400 charge for an out-of-network lab. She appealed and won but says, There are a lot of people who dont have the time or wherewithal to do this kind of fighting.

Stephanie Bywater, chief compliance officer at NeoGenomics Laboratories, which owns Genoptix, said that insurance policies governing approval have not kept up with the rapid pace of scientific advances. Kuppersmiths doctor ordered a test that has been available since 2014 and was updated in 2017, Bywater said.

Although experts agree that molecular diagnostics is an essential part of care for patients like Kuppersmith, doctors and insurance companies may not agree on which specific test is best, said Dr. Gwen Nichols, chief medical officer of the Leukemia & Lymphoma Society.

Tests can be performed a number of different ways by a number of different laboratories who charge different amounts, Nichols said.

Insurance plans are much more likely to refuse to pay for molecular diagnostics than other lab tests. Laboratory Economics found Medicare contractors denied almost half of all molecular diagnostics claims over the past five years, compared with 5-10% of routine lab tests.

With so many insurance plans, so many new tests and so many new companies, it is difficult for a doctor to know which labs are in a patients network and which specific tests are covered, Nichols said.

Different providers have contracts with different diagnostic companies, which can affect a patients out-of-pocket costs, Nichols said. It is incredibly complex and really difficult to determine the best, least expensive path.

Kuppersmith said she has always been careful to check that her doctors accept her insurance. She made sure Mount Sinai was in her insurance network, too. But it never occurred to her that the biopsy would be sent to an outside lab or that it would undergo genetic analysis.

She added: The looming threat of a $2,400 bill has caused me, in many ways, more anxiety than the illness ever has.

Kuppersmiths doctor recommended a bone marrow biopsy after suspecting she had a rare blood disorder. Though the biopsy was done by an in-network provider at an in-network hospital, Kuppersmith learned she was on the hook for $2,400 for out-of-network genetic profiling.

The Resolution: Despite making dozens of phone calls, Kuppersmith got nothing but confusing and contradictory answers when she tried to sort out the unexpected charge.

An agent for her insurer told her that her doctor hadnt gotten preauthorization for the testing. But in an email to Kuppersmith, a Genoptix employee told her the insurance company had denied the claim because molecular profiling was viewed as experimental.

[khnslabs slabs="822742"]

A spokesperson for New York-based Empire Blue Cross Blue Shield, which handled part of Kuppersmiths claim, said her health plan covers medically necessary genetic testing.

New York, one of 28 states with laws against surprise billing, requires hospitals to inform patients in writing if their care may include out-of-network providers, said attorney Elisabeth Benjamin, vice president of health initiatives at the Community Service Society, which provides free help with insurance problems.

A spokesperson for Mount Sinai said the hospital complies with that law, noting that Kuppersmith was given such a document in 2018 nearly one year before her bone marrow biopsy and signed it.

Benjamin said thats not OK, explaining: I think a one-year-old, vague form like the one she signed would not comply with the state law and certainly not the spirit of it.

Instead of sending Kuppersmith a bill, Genoptix offered to help her appeal the denied coverage to CareFirst. At first, Genoptix asked Kuppersmith to designate the company as her personal health care representative. She was uncomfortable signing over what sounded like sweeping legal rights to strangers. Instead, she wrote an email granting the company permission to negotiate on her behalf. It was sufficient.

A few days after being contacted by KHN, Kuppersmiths insurer said it would pay Genoptix at the in-network rate, covering $1,200 of the $2,400 charge. Genoptix said it has no plans to bill Kuppersmith for the other half of the charge.

The Takeaway: Kuppersmith is relieved her insurer changed its mind about her bill. But, she said: Im a relatively young, savvy person with a college degree. There are a lot of people who dont have the time or wherewithal to do this kind of fighting.

[khnslabs slabs="975218"]

Patients should ask their health care providers if any outside contractors will be involved in their care, including pathologists, anesthesiologists, clinical labs or radiologists, experts said. And check if those involved are in-network.

Try your best to ask in advance, said Jack Hoadley, a research professor emeritus at Georgetown University. Ask, Do I have a choice about where [a blood or tissue sample] is sent?

Ask, too, if the sample will undergo molecular diagnostics. Since the testing is still relatively new and expensive most insurers require patients to obtain prior authorization, or special permission, said Dr. Debra Regier, a medical geneticist at Childrens National Hospital in Washington and an associate with NORD, the National Organization of Rare Diseases. Getting this permission in advance can prevent many headaches.

Finally, be wary of signing blanket consent forms telling you that some components of your care may be out-of-network. Tell your provider that you want to be informed on a case-by-case basis when an out-of-network provider is involved and to consent to their participation.

Visit link:
Her Genetic Test Revealed A Microscopic Problem And A Jumbo Price Tag - Bryan-College Station Eagle

Read More...

The human DNA is littered with fossils of viruses past that attacked us. And lost – Telegraph India

Monday, March 30th, 2020

English naturalist Charles Robert Darwins seminal book On the Origin of Species by Means of Natural Selection went to press in 1859. That was 40 years before the concept of viruses was introduced to the world of science by Russian botanist Dmitri Ivanovsk. It took another century for viral researchers to decipher the genetic make-up of these infectious agents, to find out how they replicate and spread disease.

Darwin shunned and ridiculed after the publication of his book would probably be amazed to know that viruses such as the Covid-19 are considered living evidence of his theory of evolution.

If Charles Darwin reappeared today, he might be surprised to learn that humans are descended from viruses as well as from apes, British microbiologist Robin Weiss wrote in the journal Retrovirology. He was referring to fragments of retroviruses close cousins of the coronavirus found in the human genome.

These fragments are the fossils of a number of killer viruses, including several novel reassortments of influenza or coronaviruses that ravaged humans on a large scale in the past. These so-called endogeneous retroviruses (ERVs) are actually trophies of ancient molecular battles between viruses and their human hosts one which the humans eventually won. Some 8 per cent of human DNA represents fossil retroviral genomes, pointed out Weiss in his seminal research paper in 2010.

When competing groups of scientists in different parts of the world fully mapped the human genome in 2003, they found something they had never anticipated: bits that have no known function. Many scientists termed these seemingly inert shards junk DNA that littered our bodies. In the following decade, however, geneticists realised that some of those bits were actually endogenous retroviruses, fossils of defeated viruses that managed to invade our bodies but were disabled by our immune system. In Darwinian terms, in the struggle for existence our immune system got better of them and there was survival of the fittest through natural selection. Instead of getting buried as mineralised relics, these viruses reside within our DNA as bits of genetic code carrying records of millions of years but forever disabled, with no power to make us sick.

The discovery of the human genome as a living document of ancient and now extinct viruses prompted the emergence of a new field called palaeovirology. Two of its proponents, Michael Emerman and Harmit S. Malik, at the Fred Hutchinson Cancer Research Center in Seattle, US, define palaeovirology as the study of extinct viruses (called palaeoviruses) and the effects these agents have had on the evolution of their hosts. In other words, indirect evidence of these viral fossils can help reconstruct the past and offer clues on how to fight emerging viral epidemics or pandemics.

Malik grew up in Bombay and studied chemical engineering at the Indian Institute of Technology there. He studies evolving proteins and the genetics of evolutionary conflict embedded in the molecules, which has helped him uncover previously unrecognised sources of conflict.

As a pioneer palaeovirologist, Malik is fascinated by the constant battle being waged between humans and viruses for hundreds of thousands of years. In the course of his study, he found telltale imprints in our genome that narrate the story of how viruses infected our cells, how sometimes we have fought back by changing our protein and how sometimes viruses evaded them to get an upper hand. This evolutionary cat-and-mouse game has shaped our defence against viruses.

Palaeovirologists have also studied how similar viruses have attacked our close relatives, the primates chimpanzees and gorillas and compared how we have fared in these battles. For instance, the virus that leads to to the killer disease Aids in humans does not have much of an effect on chimps. What makes chimps relatively immune to this scary virus? Malik and Emerman found the clue to this mystery in an endogenous retrovirus called Pterv found in chimps (and other apes) but not in humans. They surmised that the retrovirus may have infected both humans and chimps about 4 million years ago. We learnt to stave off the virus while the chimps were hit by an epidemic. A gene called Trim 5 alpha is believed to have helped humans make a protein to purge the virus.

Malik and his colleagues reconstructed a part of the Pterv through computer modelling and found that while Trim 5 alpha helped us prevent the entry of the virus, it made us vulnerable to the HIV virus that causes Aids. However, the monkey version of the gene helps protect the apes from HIV and Aids. They concluded that if we can develop a therapy based on the Trim 5 alpha protein, it could defeat HIV. Research on drugs based on such evolutionary principles, however, is few and far between

Scientists have been studying several resurrected palaeoviruses like Pterv through evolution-guided reconstruction procedures. In 2005, researchers at the US Centers for Disease Control and Prevention (CDC), reconstructed the influenza virus that caused the 1918-19 flu pandemic, which killed as many as 50 million people worldwide. According to CDC, the research provides new information about the properties that contributed to its exceptional virulence. The natural emergence of another pandemic virus is considered highly likely by many experts, and therefore insights into pathogenic mechanisms can and are contributing to the development of prophylactic and therapeutic interventions needed to prepare for future pandemic viruses, says a CDC release.

The rapid evolution of viruses and emergence of the new coronavirus Covid-19 has once again shown that viruses evolve by the same means as humans. Many of these viruses hop from animals to humans and evolve, swapping genetic material in and out of respective genomes. Thats why we can have immunity to a virus weve had in the past, but get seriously affected by one our body has never seen before.

Somewhere, Darwin must be feeling vindicated that his theory is so starkly exposed in a viral machinery.

Follow this link:
The human DNA is littered with fossils of viruses past that attacked us. And lost - Telegraph India

Read More...

The distorted idea of ‘cool’ brain research is stifling psychotherapy – The Next Web

Monday, March 30th, 2020

There is always a well-known solution to every human problem neat, plausible, and wrong.From Prejudices (1920) by H L Mencken

There has never been a problem facing mankind more complex than understanding our own human nature. And no shortage of neat, plausible, and wrong answers purporting to plumb its depths.

Having treated many thousands of psychiatric patients in my career, and having worked on the American Psychiatric Associations efforts to classify psychiatric symptoms (published as the Diagnostic and Statistical Manual of Mental Disorders, or DSM-IV and DSM-5), I can affirm confidently that there are no neat answers in psychiatry. The best we can do is embrace an ecumenical four-dimensional model that includes all possible contributors to human functioning: the biological, the psychological, the social, and the spiritual. Reducing people to just one element their brain functioning, or their psychological tendencies, or their social context, or their struggle for meaning results in a flat, distorted image that leaves out more than it can capture.

The National Institute of Mental Health (NIMH) was established in 1949 by the federal government in the United States with the practical goal of providing an objective, thorough, nationwide analysis and reevaluation of the human and economic problems of mental health. Until 30 years ago, the NIMH appreciated the need for this well-rounded approach and maintained a balanced research budget that covered an extraordinarily wide range of topics and techniques.

But in 1990, the NIMH suddenly and radically switched course, embarking on what it tellingly named the Decade of the Brain. Ever since, the NIMH has increasingly narrowed its focus almost exclusively to brain biology leaving out everything else that makes us human, both in sickness and in health. Having largely lost interest in the plight of real people, the NIMH could now more accurately be renamed the National Institute of Brain Research.

Read: [How tech is helping brain-injured patients with decision-making]

This misplaced reductionism arose from the availability of spectacular research tools (eg, the Human Genome Project, functional magnetic resonance imaging, molecular biology, and machine learning) combined with the naive belief that brain biology could eventually explain all aspects of mental functioning. The results have been a grand intellectual adventure, but a colossal clinical flop. We have acquired a fantastic window into gene and brain functioning, but little to help clinical practice.

The more we learn about genetics and the brain, the more impossibly complicated both reveal themselves to be. We have picked no low-hanging fruit after three decades and $50 billion because there simply is no low-hanging fruit to pick. The human brain has around 86 billion neurons, each communicating with thousands of others via hundreds of chemical modulators, leading to trillions of potential connections. No wonder it reveals its secrets only very gradually and in a piecemeal fashion.

Genetics offers the same baffling complexity. For instance, variation in more than 100 genes contributes to vulnerability to schizophrenia, with each gene contributing just the tiniest bit, and interacting in the most impossibly complicated ways with other genes, and also with the physical and social environment. Even more discouraging, the same genes are often implicated in vulnerability to multiple mental disorders defeating any effort to establish specificity. The almost endless permutations will defeat any easy genetic answers, no matter how many decades and billions we invest.

The NIMH has boxed itself into a badly unbalanced research portfolio. Playing with cool brain and gene research toys trumps the much harder and less intellectually rewarding task of helping real people.

Contrast this current NIMH failure with a great success story from NIMHs distant past. One of the high points of my career was sitting on the NIMH granting committee that funded psychotherapy studies in the 1980s. We helped to support the US psychologist Marsha Linehans research that led her to develop dialectical behavior therapy; the US psychiatrist Aaron T Becks development of cognitive therapy; along with numerous other investigators and themes. Subsequent studies have established that psychotherapy is as effective as medications for mild-to-moderate depression, anxiety, and other psychiatric problems, and avoids the burden of medication side-effects and complications. Many millions of people around the world have already been helped by NIMH psychotherapy research.

In a rational world, the NIMH would continue to fund a robust psychotherapy research budget and promote its use as a public-health initiative to reduce the current massive overprescription of psychiatric medication in the US. Brief psychotherapy would be the first-line treatment of most psychiatric problems that require intervention. Drug treatments would be reserved for severe psychiatric problems and for those people who havent responded sufficiently to watchful waiting or psychotherapy.

Unfortunately, we dont live in a rational world. Drug companies spend hundreds of millions of dollars every year influencing politicians, marketing misleadingly to doctors, and pushing pharmaceutical treatments on the public. They successfully sold the fake marketing jingle that all emotional symptoms are due to a chemical imbalance in the brain and therefore all require a pill solution. The result: 20% of US citizens use psychotropic drugs, most of which are no more than expensive placebos, all of which can produce harmful side-effects.

Drug companies are commercial Goliath with enormous political and economic power. Psychotherapy is a tiny David with no marketing budget; no salespeople mobbing doctors offices; no TV ads; no internet pop-ups; no influence with politicians or insurance companies. No surprise then that the NIMHs neglect of psychotherapy research has been accompanied by its neglect in clinical practice. And the NIMHs embrace of biological reductionism provides an unintended and unwarranted legitimization of the drug-company promotion that there is a pill for every problem.

A balanced NIMH budget would go a long way toward correcting the two biggest mental-health catastrophes of today. Studies comparing psychotherapy versus medication for a wide variety of mild to moderate mental disorders would help to level the playing field for the two, and eventually reduce our massive overdependence on drug treatments for nonexistent chemical imbalances. Health service research is desperately needed to determine best practices to help people with severe mental illness avoid incarceration and homelessness, and also escape from them.

The NIMH is entitled to keep an eye on the future, but not at the expense of the desperate needs of the present. Brain research should remain an important part of a balanced NIMH agenda, not its sole preoccupation. After 30 years of running down a bio-reductionistic blind alley, it is long past time for the NIMH to consider a biopsychosocial reset, and to rebalance its badly uneven research portfolio.

This article was originally published at Aeonby Allen Frances and has been republished under Creative Commons.

Read next: What will we do when diseases reach space?

Read our daily coverage on how the tech industry is responding to the coronavirus and subscribe to our weekly newsletter Coronavirus in Context.

For tips and tricks on working remotely, check out our Growth Quarters articles here or follow us on Twitter.

Continue reading here:
The distorted idea of 'cool' brain research is stifling psychotherapy - The Next Web

Read More...

CMSS: 800000 physicians across 40 specialties continue to support travel and gathering restrictions – YubaNet

Monday, March 30th, 2020

March 27, 2020

Dear President Trump, Vice President Pence, and Ambassador Birx:

Advertisement

Thank you for actively engaging the health care communityparticularly the nations physicians and the organizations that represent themin addressing the COVID-19 pandemic in the United States.

As more than 800,000 physicians across more than 40 specialties in medicine, we continue to support travel and gathering restrictions to slow the transmission of coronavirus disease 2019 (COVID-19). Physicians, other health professionals, and staff are putting themselves in harms way without adequate personal protective equipment or necessary tools (such as ventilators) as well as with no proven treatment or vaccine for the virus.

Significant COVID-19 transmission continues across the United States, and we need your leadership in supporting science-based recommendations on social distancing that can slow the virus. Our societies have closely adhered to these measures by moving our staff to fulltime telework and canceling in-person meetings (including annual meetings). These actions have helped to keep physicians and other health professionals in health care facilities, including hospitals, and reduce the risk of spreading COVID-19.

Statewide efforts alone will not sufficiently control this public health crisis. A strong nationwide plan that supports and enforces social distancingand recognizes that our health and our economy are inextricably linkedshould remain in place until public health and medical experts indicate it can be lifted.

Federal, state, and local governments should only set a date for lifting nationwide social distancing restrictions consistent with assessments by public health and medical experts. Lifting restrictions sooner will gravely jeopardize the health of all Americans and extend the devastation of the COVID-19 pandemic.

Again, thank you for actively engaging the nations physicians and the organizations that represent them. We appreciate your considering these comments and welcome an opportunity for further dialogue.

Sincerely,

Council of Medical Specialty Societies

American Academy of Allergy, Asthma & ImmunologyAmerican Academy of DermatologyAmerican Academy of Family PhysiciansAmerican Academy of Hospice and Palliative MedicineAmerican Academy of NeurologyAmerican Academy of OphthalmologyAmerican Academy of Orthopaedic SurgeonsAmerican Academy of PediatricsAmerican Academy of Physical Medicine and RehabilitationAmerican Association of Clinical EndocrinologistsAmerican College of CardiologyAmerican College of Emergency PhysiciansAmerican College of Medical Genetics and GenomicsAmerican College of Obstetricians and GynecologistsAmerican College of Occupational and Environmental MedicineAmerican College of PhysiciansAmerican College of Preventive MedicineAmerican College of RadiologyAmerican College of RheumatologyAmerican College of SurgeonsAmerican Epilepsy SocietyAmerican Gastroenterological AssociationAmerican Geriatrics SocietyAmerican Medical Informatics AssociationAmerican Psychiatric AssociationAmerican Society of AnesthesiologistsAmerican Society for Clinical PathologyAmerican Society of Colon and Rectal SurgeonsAmerican Society of HematologyAmerican Society of NephrologyAmerican Society for Radiation OncologyAmerican Society for Reproductive MedicineAmerican Thoracic SocietyAmerican Urological AssociationAssociation for Clinical OncologyInfectious Diseases Society of AmericaNorth American Spine SocietySociety of Critical Care MedicineSociety of Gynecologic OncologySociety of Hospital MedicineSociety of Interventional RadiologySociety of Nuclear Medicine and Molecular ImagingSociety of Thoracic SurgeonsSociety for Vascular Surgery

Founded in 1965 as the Tri-College Council, CMSS was created to provide an independent forum for the discussion by medical specialists of issues of national interest and mutual concern.Founding members were the American College of Obstetricians and Gynecologists, the American College of Physicians, and the American College of Surgeons.In 1967, as other specialty societies joined, CMSS adopted its current name.Today,43 societies with more than 790,000 U.S. physician membersare members of CMSS. CMSS is a 501(c)(3) not-for-profit association incorporated in the state of Illinois. http://www.cmss.org

Go here to read the rest:
CMSS: 800000 physicians across 40 specialties continue to support travel and gathering restrictions - YubaNet

Read More...

How is the COVID-19 Virus Detected using Real Time RT-PCR? – International Atomic Energy Agency

Saturday, March 28th, 2020

A virus is a microscopic package of genetic material surrounded by a molecular envelope. The genetic material can be either DNA or RNA.

DNA is a two-strand molecule that is found in all organisms, such as animals, plants, and viruses, and it holds the genetic code, or blueprint, for how these organisms are made and develop.

RNA is generally a one-strand molecule that copies, transcribes and transmits parts of the genetic code to proteins so they can synthetize and carry out functions that keep organisms alive and developing. There are different variations of RNA that do the copying, transcribing and transmitting.

Some viruses such as the coronavirus (SARS-Cov2) only contain RNA, which means they rely on infiltrating healthy cells to multiply and survive. Once inside the cell, the virus uses its own genetic code RNA in the case of the coronavirus to take control of and reprogramme the cells so that they become virus-making factories.

In order for a virus like the coronavirus to be detected early in the body using real time RT-PCR, scientists need to convert the RNA to DNA. This is a process called reverse transcription. They do this because only DNA can be copied or amplified which is a key part of the real time RT-PCR process for detecting viruses.

Scientists amplify a specific part of the transcribed viral DNA hundreds of thousands of times. Amplification is important so that instead of trying to spot a minuscule amount of the virus among millions of strands of genetic information, scientists have a large enough quantity of the target sections of viral DNA to accurately confirm that the virus is present.

Link:
How is the COVID-19 Virus Detected using Real Time RT-PCR? - International Atomic Energy Agency

Read More...

How sick will the coronavirus make you? The answer may be in your genes – Science Magazine

Saturday, March 28th, 2020

A patient in Italy receives intensive care for COVID-19. Human geneticists are coming together to look for genes that make some people more vulnerable to the disease.

By Jocelyn KaiserMar. 27, 2020 , 3:25 PM

Sciences COVID-19 reporting is supported by the Pulitzer Center.

COVID-19, caused by the new pandemic coronavirus, is strangelyand tragicallyselective. Only some infected people get sick, and although most of the critically ill are elderly or have complicating problems such as heart disease, some killed by the disease are previously healthy and even relatively young. Researchers are now gearing up to scour the patients genomes for DNA variations that explain this mystery. The findings could be used to identify those most at risk of serious illness and those who might be protected, and they might also guide the search for new treatments.

The projects range from ongoing studies with DNA for many thousands of participants, some now getting infected with the coronavirus, to new efforts that are collecting DNA from COVID-19 patients in hard-hit places such as Italy. The goal is to compare the DNA of people who have serious cases of COVID-19 (which stands for coronavirus disease 2019)but no underlying disease like diabetes, heart or lung diseasewith those with mild or no disease. We see huge differences in clinical outcomes and across countries. How much of that is explained by genetic susceptibility is a very open question, says geneticist Andrea Ganna of the University of Helsinkis Institute for Molecular Medicine Finland (FIMM).

Its hard to predict what will pop out from these gene hunts, some researchers say. But there are obvious suspects, such as the gene coding for the cell surface protein angiotensin-converting enzyme 2 (ACE2), which the coronavirus uses to enter airway cells. Variations in the ACE2 gene that alter the receptor could make it easier or harder for the virus to get into cells, says immunologist Philip Murphy of the National Institute of Allergy and Infectious Diseases, whose lab identified a relatively common mutation in another human cell surface protein, CCR5, that makes some people highly resistant to HIV.

Ganna heads up a major effort to pool COVID-19 patients genetic data from around the world. The idea came quite spontaneously about 2 weeks ago when everyone was sitting at their computers watching this crisis, says Ganna, who is also affiliated with the Broad Institute, a U.S. genomic powerhouse.

He and FIMM Director Mark Daly quickly created a website for their project, the COVID-19 Host Genetics Initiative, and reached out to colleagues who run large biobank studies that follow thousands of volunteers for years to look for links between their DNA and health. At least a dozen biobanks, mostly in Europe and the United States, have expressed interest in contributing COVID-19 data from participants who agreed to this. Among them are FinnGen, which has DNA samples and health data for 5% of the 5 millionperson Finnish population, and the 50,000-participant biobank at the Icahn School of Medicine at Mount Sinai.

The UK Biobank, one of worlds largest with DNA data for 500,000 participants, also plans to add COVID-19 health data from participants to its data set, the project tweeted this month. And the Icelandic company deCODE Genetics, which is helping test much of the nations population to see who is infected with the new coronavirus, has received government permission to add these data and any subsequent COVID-19 symptoms to its database, which contains genome and health data on half of Icelands 364,000 inhabitants, says its CEO Kri Stefnsson. We will do our best to contribute to figuring this out, Stefnsson says.

Another effort to identify protective or susceptibility DNA variants is the Personal Genome Project led by Harvard Universitys George Church, which recruits people willing to share their full genome, tissue samples, and health data for research. Earlier this month, it sent questionnaires to its thousands of participants, asking about their COVID-19 status. More than 600 in the United States responded within 48 hours. It seems that most people want to do their part, says Church, whose group isnt yet part of Gannas collaboration.

Other researchers working with Gannas initiative are recruiting COVID-19 patients directly within hospitals for such genomics studies. Italian geneticist Alessandra Renieri of the University of Siena expects at least 11 hospitals in the nation to give ethics approval for her team to collect DNA samples from willing patients. It is my opinion that [host] genetic differences are a key factor for susceptibility to severe acute pneumonia, Renieri says.

Pediatrics researcher Jean-Laurent Casanova at the Rockefeller University, who specializes in identifying rare genes that can make healthy young people susceptible to certain serious diseases, is drawing on a network of pediatricians around the world to look for the relatively few young people who develop COVID-19 serious enough to get admitted to intensive care. We study exclusively patients who were previously healthy and under 50, as their serious COVID-19 illness is more likely to have a genetic basis, he explains.

In addition to genetic variants of the ACE2 receptor, scientists want to see whether differences in the human leukocyte antigen genes, which influence the immune systems response to viruses and bacteria, affect disease severity. And some investigators want to follow up a finding, which a Chinese team reported in a preprint: that people with type O blood may be protected from the virus. Were trying to figure out if those findings are robust, says Stanford University human geneticist Manuel Rivas, who is contributing to Gannas initiative.

The catastrophic spread of the coronavirus should soon increase the number of COVID-19 patients available to these gene hunts. And that could speed findings. Ganna expects the first susceptibility genes could be identified within a couple of months.

With reporting by Elizabeth Pennisi.

Follow this link:
How sick will the coronavirus make you? The answer may be in your genes - Science Magazine

Read More...

Universities Issue Hiring Freezes in Response to COVID-19 – The Scientist

Saturday, March 28th, 2020

Heather Ray, a developmental biology postdoc at the University of Alabama at Birmingham who studies the genetics of birth defects, received an email on March 24 from the search committee for a faculty job to which she had applied. Due to the COVID-19 pandemic and a resulting hiring freeze at the university, Ray says the email read, the search and position had been canceled.

I hadnt even thought that this might happen, she says.

A few minutes later, Ray received an email from a search committee in her own department that had been interviewing applicants for two open positions. Three candidates had completed in-person interviews for those positions, and all three had been invited back to campus for a second round of interviews. The University of Alabama at Birmingham had recently announced that it too was suspending hiring for individuals who did not have a documented offer, which meant that the search in Rays department was also called off.

All of the work that they did, thats just gone away, she says, because when the job is posted again in the fall, or the following year, the search committee will have to start all over.

A day earlier, on March 23, Brown University announced the suspension of all new hiring for the current year and for the fiscal year starting on July 1. At the same time that supporting our communitys health and safety remain a top priority, reads the universitys press release, the financial consequences resulting from the increased expenditures and the dramatic reductions in revenue provoked by the current pandemic demand prompt attention.

Numerous other universities have made similar announcements about the suspension of hiring, including Miami University of Ohio, Wright State University, Villanova University, the University of Nevada, Reno, Indiana University, and the University of Auckland in New Zealand, which cites the travel ban affecting Chinese students as one of the main causes of its freeze. The Professor Is In, a website and consulting service for applicants on the academic job market, posted yesterday an unofficial list of 58 universities that had issued some form of a hiring freeze. Comments in response to that post point to additional universities that have also scaled back hiring to various degrees.

James McGrath, a postdoc in digital public humanities at Brown University, tweeted about Browns announcement on Tuesday, eliciting responses from students and academics in various disciplines.

The worry is with an institution like Brown that has the endowment that it has and the resources that it has, McGrath tells The Scientist. If theyre making this decision, you can only wonder . . . what kinds of decisions [institutions with fewer resources] are going to be making. He says the hiring freezes in response to the coronavirus pandemic are a sign that the whole higher ed job market, which is already in bad shape, is heading for darker times.

A summary from the Senate Appropriations Committee released yesterday specified that the $2 trillion stimulus package passed by the Senate Wednesday (March 25) and currently being considered by the House would include at least $1.25 billion for federal research agencies working on the coronavirus, as well as $14.25 billion for universities that have been forced to shut down during the pandemic, which may funnel aid to labs whose work has been disrupted, Science reports.

But how that stimulus package may affect those currently searching for academic positions remains unclear.

Its definitely scary as someone who is looking for another postdoc or potentially applying to faculty jobs to have this looming situation, says Kishana Taylor, a postdoc in microbiology and molecular genetics at the University of California, Davis. If universities are going along the same lines as Brown, Taylor says, essentially, there wont be any applications to put in in the fall. She notes that the job market is always competitive, especially in the biomedical sciences, and that the hiring freezes will only increase that competition.

Ray agrees, adding that if the current situation persists, universities may experience a bottleneck where two or three years worth of applicants [are] all trying for the same number of positions at one time.

Julie Craft Van De Weghe, a cellular biology postdoc at the University of Washington whose research focuses on primary cilia and Joubert syndrome, says she wonders whether the terms of her K99 Pathway to Independence Award from the National Institutes of Health will be adjusted if the current lack of hiring in academia persists. The grant is supposed to cover two years of a postdoc and the first three years of a faculty position, according to Craft Van De Weghe, but she is unsure what would happen if shes unable to secure an academic job in the coming year. I will be reevaluating my contingency plans, she says.

The hiring freezes and their effects on the larger scientific community also concern Craft Van De Weghe. Although universities are producing researchers well-trained to contribute to humanitys knowledge of how science works, she says, well lose a ton of good people who could have made substantial contributions.

Roughly two weeks ago, Ray signed an offer letter from a university, completing her extended search for a faculty position that began in June of last year, and had pulled her application from the university that later notified her of its hiring freeze. The timing had been perfect, she says, because she was able to travel for an in-person interview and negotiate her contract before universities starting closing their campuses. But on the morning of March 25, a day after she had received the emails about hiring freezes and canceled searches, Ray learned that the university where she had accepted an offer had issued its own hiring freeze. She immediately emailed the committee chair asking whether her position was under threat.

I wasnt left waiting too long, Ray says, but it was a brief moment of almost panic. Her new job, she reports, is secure.

Amy Schleunes is an intern atThe Scientist. Email her ataschleunes@the-scientist.com.

View post:
Universities Issue Hiring Freezes in Response to COVID-19 - The Scientist

Read More...

In the start-up nation – The Hindu

Saturday, March 28th, 2020

When I first arrived at the Weizmann Institute of Science (WIS), Tel Aviv, in May 2013 as a summer trainee, little did I know that I will be spending a good part of my life in Israel. There is some inexplicable aura of scientific aptitude in the beautiful campus nestled just outside the bustling city of Tel Aviv. It was rather an easy decision to return for a Ph.D. in Molecular Genetics at the institute after graduating with an Integrated BS-MS in Biology from the Indian Institute of Science Education and Research (IISER), Kolkata.

Unparalleled quality

WIS is a rather unusual destination for graduate school, as compared to other major schools in the U.S. and Europe as it offers only MSc and Ph.D. programmes in natural sciences (Physics, Chemistry, Mathematics/CS, Biology). Quite naturally, the student/post-doc body is compact leading to thought-provoking scientific conversations, often leading to interesting scientific collaborations. In addition to roughly 250 professors across 18 departments, there are numerous staff scientists, many of whom serve as an immediate source of guidance and mentorship.

Weizmann supports services for almost all major advanced scientific tools and techniques. This enables the students and other researchers to answer tough questions. For instance, discoveries at the institute have enabled the treatment of diseases such as multiple sclerosis, lung cancer, prostate cancer, and lymphoma. Not only is the quality of science unparalleled, but also, Weizmann Institute is a great place to work. The continual support of the mostly English-speaking administrative staff makes it probably the best place to be for an international student in Israel.

Co-curricular activities

Outside the lab, the institute provides ample opportunities for personal and professional growth. There are multiple student clubs such as Entrepreneurship, Debate and LGBTQ clubs that train students with various hard and soft skills such as innovation, management and persuasive speaking. In fact, I have been involved in establishing the Weizmann Biotech Club, which is a forum for Ph.D. students and post-docs to interact with life-science industry leaders, learn about the industry trends and provide career development support.

Finally, Israel is a great place to begin ones professional career. It is known to be the start up nation and provides a plethora of opportunities to engage with the booming innovation ecosystem of the country for aspiring technology entrepreneurs.

Israel is opening its doors to foreign entrepreneurs with the recent launch of an innovation visa, that comes with generous support from the Israel Innovation Authority.

The writer is a Ph.D. student at the Dept. of Molecular Genetics, Weizmann Institute of Science, Israel

You have reached your limit for free articles this month.

Register to The Hindu for free and get unlimited access for 30 days.

Find mobile-friendly version of articles from the day's newspaper in one easy-to-read list.

Enjoy reading as many articles as you wish without any limitations.

A select list of articles that match your interests and tastes.

Move smoothly between articles as our pages load instantly.

A one-stop-shop for seeing the latest updates, and managing your preferences.

We brief you on the latest and most important developments, three times a day.

Not convinced? Know why you should pay for news.

*Our Digital Subscription plans do not currently include the e-paper ,crossword, iPhone, iPad mobile applications and print. Our plans enhance your reading experience.

Go here to read the rest:
In the start-up nation - The Hindu

Read More...

The First Genetic Map of the Cerebral Cortex – Technology Networks

Saturday, March 28th, 2020

The cerebral cortex is the relatively thin, folded, outer gray matter layer of the brain crucial for thinking, information processing, memory, and attention. Not much has been revealed about the genetic underpinnings that influence the size of the cortexs surface area and its thickness, both of which have previously been linked to various psychiatric traits, including schizophrenia, bipolar disorder, depression, attention deficit hyperactivity disorder (ADHD), and autism.Now, for the first time, more 360 scientists from 184 different institutions including UNC-Chapel Hill have contributed to a global effort to find more than 200 regions of the genome and more than 300 specific genetic variations that affect the structure of the cerebral cortex and likely play important roles in psychiatric and neurological conditions.

The study was led by co-senior authors Jason Stein, PhD, assistant professor in the Department of Genetics at the UNC School of Medicine; Sarah Medland, PhD, senior research fellow at the QIMR Berghofer Medical Research Institute in Australia; and Paul Thompson, PhD, associate director of the Mark and Mary Stevens Neuroimaging and Informatics Institute at the University of Southern California. Ten years ago, these scientists cofounded the ENIGMA Consortium, an international research network that has brought together hundreds of imaging genomics researchers to understand brain structure, function, and disease based on brain imaging and genetic data.

This study was only possible due to a huge scientific collaboration of more than 60 sites involved in MRI scanning and genotyping participants, Stein said. This study is the crown jewel of the ENIGMA Consortium, so far.

The researchers studied MRI scans and DNA from more than 50,000 people to identify 306 genetic variants that influence brain structure in order to shed light on how genetics contribute to differences in the cerebral cortex of individuals. Genetic variants or variations are simply the slight genetic differences that make us unique. Generally speaking, some variants contribute to differences such as hair color or blood type. Some are involved in diseases. Most of the millions of genetic variants, though, have no known significance. This is why pinpointing genetic variants associated with cortex size and structure is a big deal. Stein and colleagues consider their new genetic roadmap of the brain a sort of Rosetta stone that will help translate how some genes impact physical brain structure and neurological consequences for individuals.

Among the findings of the research:

Most of our previous understanding of genes affecting the brain are from model systems, like mice, Stein said. With mice, we can find genes, knock out genes, or over express genes to see how they influence the structure or function of the brain. But there are a couple of problems with this.One problem is, quite simply, a mouse is not a human. There are many human-specific features that scientists can only study in the human brain.

The genetic basis for a mouse is very different than the genetic basis for humans, Stein said, especially in in the noncoding regions of the genome.

Genes contain DNA, the basic human code that, when translated into action, creates proteins that do things, such as help your finger muscles type or your heart beat or your liver process toxins. But only about 3 percent of the human genome codes for proteins. The vast majority of the human genome is called the noncoding genome. Much of this region is not shared between mice and humans. This noncoding genome consists of tiny molecular switches that can modulate the expression of other genes. These switches dont directly alter the function of a protein, but they can affect the amounts of a protein that is expressed. Turns out, most genetic variants associated with psychiatric disorders are found in the noncoding region of the genome.

These findings can now be a resource for scientists to help answer important questions about the genetic influences on the brain and how they relate to numerous conditions.ReferenceGrasby et al. (2020) The genetic architecture of the human cerebral cortex. Science. DOI: https://doi.org/10.1126/science.aay6690

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

View original post here:
The First Genetic Map of the Cerebral Cortex - Technology Networks

Read More...

Removing EGR4 "Brake" on Immune Activation May Be Viable I/O Therapeutic Approach – Clinical OMICs News

Saturday, March 28th, 2020

Scientists at the Lewis Katz School of Medicine at Temple University (LKSOM) and Fox Chase Cancer Center show that EGR4, known mainly for its role in male fertility, serves as a critical brake on immune activation. The new study Suppression of Ca2+signals by EGR4 controls Th1 differentiation and anticancer immunityin vivo,published online inEMBO Reports, demonstrates that taking EGR4 away, thus effectively releasing the brake, promotes the activation of killer T cells, which infiltrate and attack tumors and thereby boost anticancer immunity.

While the zinc finger transcription factors EGR1, EGR2, and EGR3 are recognized as critical for Tcell function, the role of EGR4 remains unstudied. Here, we show that EGR4 is rapidly upregulated upon TCR engagement, serving as a critical brake on Tcell activation. Hence, TCR engagement of EGR4/T cells leads to enhanced Ca2+responses, driving sustained NFAT activation and hyperproliferation. This causes profound increases in IFN production under resting and diverse polarizing conditions that could be reversed by pharmacological attenuation of Ca2+ entry, wrote the investigators.

Finally, aninvivomelanoma lung colonization assay reveals enhanced antitumor immunity in EGR4/mice, attributable to Th1 bias, Treg loss, and increased CTL generation in the tumor microenvironment. Overall, these observations reveal for the first time that EGR4 is a key regulator of Tcell differentiation and function.

Other early growth response proteins, or EGRs, are important to T cell activity, but whether EGR4 also has a role in immunity has been largely overlooked, explained Jonathan Soboloff, Ph.D., professor of Medical Genetics and Molecular Biochemistry at the Fels Institute for Cancer Research and Molecular Biology at LKSOM. Our study reveals a new side to the importance of EGR4.

Soboloffs team examined the influence of EGR4 expression in immune cells in collaboration with Dietmar J. Kappes, Ph.D., professor of Blood Cell Development and Cancer at Fox Chase Cancer Center. In initial experiments, the researchers found that T cell activation is associated with EGR4 upregulation. They then showed that knocking-out, or eliminating, EGR4 from immune cells results in a dramatic increase in calcium signaling and expansion of T helper type 1 (Th1) cell populations. Th1 cells, in response to the presence of foreign entities, including tumor cells, activate cytotoxic, or killer, T cells, which then wipe out the invader.

We know from our previous work that T cells control calcium signaling and that when intracellular calcium levels are elevated, calcium signaling can drive T cell activation, Soboloff said.

The Soboloff and Kappes labs next studied the functional importance of EGR4 in cancer immunity by utilizing an adoptive mouse model of melanoma in which some host animals lacked EGR4 expression. Compared to mice with typical EGR4 levels, EGR4 knockout animals showed evidence of expanded populations of Th1 cells and enhanced anticancer immunity. In particular, EGR4 knockout mice had reduced lung tumor burden and fewer metastases than mice with normal EGR4 expression.

In future work, the Soboloff and Kappes groups plan to further explore strategies for EGR4 targeting. The development of an agent to target EGR4 specifically may be difficult, due to the diverse actions of EGR pathways. But eliminating EGR4 specifically from a patients T cells, and then putting those cells back into the patient, may be a viable immunotherapeutic approach, Kappes said.

More:
Removing EGR4 "Brake" on Immune Activation May Be Viable I/O Therapeutic Approach - Clinical OMICs News

Read More...

More Hints of Order in the Genome – Discovery Institute

Saturday, March 28th, 2020

Genomics has come a long way since the central dogma (the notion that DNA is the master controller that calls all the shots) and junk DNA (the expectation that much of the genome is non-functional). If scientists ditch those old dogmas and approach the genome expecting to find reasons for things, they often do.

To-may-to or to-mah-to? The British write flavour; the Americans write flavor, but generally each understands the other without too much difficulty. Genomes, too, have alternate ways of spelling things: GGU and GGC in messenger RNA both spell glycine. No big deal, thought geneticists; these silent mutations cause no change in the resulting protein. At the University of Notre Dame, however, biochemists are finding that the differences in spelling are not just background noise; they alter the proteins folding. Is that good or bad?

Synonymous mutations were long considered to be genomic background noise, but we found they do indeed lead to altered protein folding, and in turn impair cell function, said Patricia Clark, the Rev. John Cardinal OHara professor of biochemistry at the University of Notre Dame, and lead author of the study. Our results show that synonymous variations in our DNA sequences which account for most of our genetic variation can have a significant impact on shaping the fitness level of cellular proteins.

Surely many of these mutations are harmful, as are random mutations in humans that cause genetic disease. But E. coli has been around for a long time. Wouldnt the species have gone extinct by now with the accumulation of defective spellings if they are always deleterious? Other work has suggested a secret code in synonymous variations that fine-tunes expression rates or regulates the supply of a given protein based on environmental conditions. The news release only mentions impairments caused by synonymous variations, but Notre Dame teams paper in PNAS suggests some possible advantages:

Synonymous codon substitutions alter the mRNA coding sequence but preserve the encoded amino acid sequence. For this reason, these substitutions were historically considered to be phenotypically silent and often disregarded in studies of human genetic variation. In recent years, however, it has become clear that synonymous substitutions can significantly alter protein function in vivo through a wide variety of mechanisms that can change protein level, translational accuracy, secretion efficiency, the final folded structure and posttranslational modifications. The full range of synonymous codon effects on protein production is, however, still emerging, and much remains to be learned regarding the precise mechanisms that regulate these effects. [Emphasis added.]

A design perspective would consider every possible function before rendering a judgment that all synonymous variations reduce fitness.

Keeping the genome accurate to a high degree preserves it from collapsing due to error catastrophe. At the time of cell division, proofreading enzymes (what a concept!) perform this vital function. Chelsea R. Bulock et al., writing in PNAS, have found one duplication enzyme that proofreads itself while proofreading its partner! DNA polymerase proofreads errors made by DNA polymerase , the paper is titled.

Pol and Pol are the two major replicative polymerases in eukaryotes, but their precise roles at the replication fork remain a subject of debate. A bulk of data supports a model where Pol and Pol synthesize leading and lagging DNA strands, respectively. However, this model has been difficult to reconcile with the fact that mutations in Pol have much stronger consequences for genome stability than equivalent mutations in Pol. We provide direct evidence for a long-entertained idea that Pol can proofread errors made by Pol in addition to its own errors, thus, making a more prominent contribution to mutation avoidance. This paper provides an essential advance in the understanding of the mechanism of eukaryotic DNA replication.

In other words, Pol is a proofreader of a proofreader. The paper says that Pol is a versatile extrinsic proofreading enzyme. One could think of it as a supervisor checking the work of a subordinate, or better yet, as an auditor or inspector able to fix errors before they cause harm to the product. Why would this be necessary during replication? The authors see a seniority system:

Thus, the high efficiency of Pol at correcting errors made by Pol may result from a combination of two factors: the high proclivity of Pol to yield to another polymerase and the greater flexibility and robustness of Pol when associating with new primer termini.

One proofreader is amazing to consider evolving by a Darwinian mechanism. A proofreader of a proofreader is astonishing. Consider, too, that this proofreading operation occurs in the dark by feel, automatically, without eyes to see.

Now that genetics is long past the heady days of finding that DNA forms a code that is translated, additional discoveries continue to show additional codes and factors that contribute to genomic function. One factor is the high-order structure of DNA. Researchers at South Koreas UNIST center have explored further into the formation of this structure, which involves chromatin wrapping around histone proteins so that long strands of DNA can fit within the compact space of the cell nucleus. As with everything else in genomics, the structure doesnt just happen. It requires a lot of help.

Regulation of histone proteins allows the DNA strands become more tightly or loosely coiled during the processes of DNA replication and gene expression. However, problems may arise when histones clump together or when DNA strands intertwine. Indeed, the misregulation of chromatin structures could result in aberrant gene expression and can ultimately lead to developmental disorders or cancers.

Histone chaperones are those proteins, responsible for adding and removing specific histones [found] at the wrong time and place during the DNA packaging process. Thus, they also play a key role in the assembly and disassembly of chromatin.

Cryo-EM imaging allowed the team to envision the molecular structure of some of these chaperone proteins. Their paper in Nature Communications begins, The fundamental unit of chromatin, the nucleosome, is an intricate structure that requires histone chaperones for assembly. Their cryo-EM images of one particular chaperone named Abo1 reveals a six-fold symmetry with precise locations for docking to histones, its hexameric ring thus creating a unique pocket where histones could bind with energy from ATP. Not only is Abo1 distinct as a histone chaperone, they write, but Abo1 is also unique compared to other canonical AAA+protein structures. Like Lego blocks, Abo1 features tight knob-and-hole packing of individual subunits plus linkers and other binding sites, such as for ATP. And unlike static blocks, these blocks undergo conformational changes as they work.

Such sophistication is far beyond the old picture of DNA as a master molecule directing all the work. It couldnt work without the help of many precision machines like this.

These stories are mere samples from a vast and growing literature indicating higher order in the genome than expected. Here are some more samples readers may wish to investigate:

Researchers at the University of Seville found additional factors involved in the repair of DNA strand breaks. These repairs are essential for the maintenance of genome integrity. The factors they discovered help maintain the right tension in cohesin molecules that hold the chromosomes together until the right time to separate. The news was relayed by EurekAlert!and published in Nature Communications.

Remember Paleys Watch? Researchers at the University of Basel discovered that Inner clockwork sets the time for cell division in bacteria. In PNAS and in Nature Communications, the Basel team elucidates the structure and function of a small signaling molecule that starts the clock, which then informs the cell about the right time to reproduce. They report in the news release:

A team at the Biozentrum of the University of Basel, led by Prof. Urs Jenal has now identified a central switch for reproduction in the model bacteriumCaulobacter crescentus: the signaling molecule c-di-GMP. In their current study,published in the journalNature Communications,they report that this molecule initiates a clock-like mechanism, which determines whether individual bacteria reproduce.

Proteins must fold properly to perform their functions. Small proteins usually fold successfully on their own, but large ones can fall into several misfolding traps that are equally likely as the canonical fold. It appears that the sequence of the sequence in a gene has something to do with this. Interestingly, many of these proteins sequences contain conserved rare codons that may slow down synthesis at this optimal window, explain Amir Bitran et al. in a January 21 paper in PNAS, discovering that Cotranslational folding (i.e., folding that begins as the polypeptide exits the ribosome) allows misfolding-prone proteins to circumvent deep kinetic traps.

Design advocates and evolutionists need to fathom what they are dealing with when discussing origins. Theres nothing like some low-level detail to put the challenge in perspective.

Image credit: Caulobacter crescentus, by University of Basel, Swiss Nanoscience Institute/Biozentrum, via EurekAlert!

Continued here:
More Hints of Order in the Genome - Discovery Institute

Read More...

Page 18«..10..17181920..30..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick