scispace - formally typeset
Search or ask a question
Author

Ayshwarya Subramanian

Bio: Ayshwarya Subramanian is an academic researcher from Broad Institute. The author has contributed to research in topics: Biology & Tumor progression. The author has an hindex of 17, co-authored 43 publications receiving 1555 citations. Previous affiliations of Ayshwarya Subramanian include Harvard University & Birla Institute of Technology and Science.


Papers
More filters
Journal ArticleDOI
13 Jun 2019-Cell
TL;DR: The data suggest that tissue stroma responds to malignant cells by disadvantaging normal parenchymal cells, and provides a comprehensive bone marrow cell census and experimental support for cancer cell crosstalk with specific stromal elements to impair normal tissue function and thereby enable emergent cancer.

532 citations

Journal ArticleDOI
Toni Delorey1, Carly G. K. Ziegler, Graham Heimberg1, Rachelly Normand, Yiming Yang1, Yiming Yang2, Asa Segerstolpe1, Domenic Abbondanza1, Stephen J. Fleming1, Ayshwarya Subramanian1, Daniel T. Montoro1, Karthik A. Jagadeesh1, Kushal K. Dey2, Pritha Sen, Michal Slyper1, Yered Pita-Juárez, Devan Phillips1, Jana Biermann3, Zohar Bloom-Ackermann1, Nikolaos Barkas1, Andrea Ganna4, Andrea Ganna2, James Gomez1, Johannes C. Melms3, Igor Katsyv3, Erica Normandin2, Erica Normandin1, Pourya Naderi5, Pourya Naderi2, Yury Popov5, Yury Popov2, Siddharth S. Raju1, Siddharth S. Raju2, Sebastian Niezen2, Sebastian Niezen5, Linus T.-Y. Tsai, Katherine J. Siddle1, Katherine J. Siddle2, Malika Sud1, Victoria M. Tran1, Shamsudheen K. Vellarikkal6, Shamsudheen K. Vellarikkal1, Yiping Wang3, Liat Amir-Zilberstein1, Deepak Atri6, Deepak Atri1, Joseph M. Beechem7, Olga R. Brook5, Jonathan H. Chen1, Jonathan H. Chen2, Prajan Divakar7, Phylicia Dorceus1, Jesse M. Engreitz8, Jesse M. Engreitz1, Adam Essene5, Donna M. Fitzgerald2, Robin Fropf7, Steven Gazal9, Joshua Gould1, John Grzyb6, Tyler Harvey1, Jonathan L. Hecht2, Jonathan L. Hecht5, Tyler Hether7, Judit Jané-Valbuena1, Michael Leney-Greene1, Hui Ma2, Hui Ma1, Cristin McCabe1, Daniel E. McLoughlin2, Eric M. Miller7, Christoph Muus2, Christoph Muus1, Mari Niemi4, Robert F. Padera6, Robert F. Padera10, Robert F. Padera2, Liuliu Pan7, Deepti Pant5, Carmel Pe’er1, Jenna Pfiffner-Borges1, Christopher J. Pinto2, Jacob Plaisted6, Jason Reeves7, Marty Ross7, Melissa Rudy1, Erroll H. Rueckert7, Michelle Siciliano6, Alexander Sturm1, Ellen Todres1, Avinash Waghray2, Sarah Warren7, Shuting Zhang1, Daniel R. Zollinger7, Lisa A. Cosimi6, Rajat M. Gupta6, Rajat M. Gupta1, Nir Hacohen1, Nir Hacohen2, Hanina Hibshoosh3, Winston Hide, Alkes L. Price2, Jayaraj Rajagopal2, Purushothama Rao Tata11, Stefan Riedel2, Stefan Riedel5, Gyongyi Szabo2, Gyongyi Szabo1, Gyongyi Szabo5, Timothy L. Tickle1, Patrick T. Ellinor1, Deborah T. Hung2, Deborah T. Hung1, Pardis C. Sabeti, Richard M. Novak12, Robert S. Rogers5, Robert S. Rogers2, Donald E. Ingber13, Donald E. Ingber2, Donald E. Ingber12, Z. Gordon Jiang2, Z. Gordon Jiang5, Dejan Juric2, Mehrtash Babadi1, Samouil L. Farhi1, Benjamin Izar, James R. Stone2, Ioannis S. Vlachos, Isaac H. Solomon6, Orr Ashenberg1, Caroline B. M. Porter1, Bo Li2, Bo Li1, Alex K. Shalek, Alexandra-Chloé Villani, Orit Rozenblatt-Rosen14, Orit Rozenblatt-Rosen1, Aviv Regev 
29 Apr 2021-Nature
TL;DR: In this article, single-cell analysis of lung, heart, kidney and liver autopsy samples shows the molecular and cellular changes and immune response resulting from severe SARS-CoV-2 infection.
Abstract: COVID-19, which is caused by SARS-CoV-2, can result in acute respiratory distress syndrome and multiple organ failure1–4, but little is known about its pathophysiology. Here we generated single-cell atlases of 24 lung, 16 kidney, 16 liver and 19 heart autopsy tissue samples and spatial atlases of 14 lung samples from donors who died of COVID-19. Integrated computational analysis uncovered substantial remodelling in the lung epithelial, immune and stromal compartments, with evidence of multiple paths of failed tissue regeneration, including defective alveolar type 2 differentiation and expansion of fibroblasts and putative TP63+ intrapulmonary basal-like progenitor cells. Viral RNAs were enriched in mononuclear phagocytic and endothelial lung cells, which induced specific host programs. Spatial analysis in lung distinguished inflammatory host responses in lung regions with and without viral RNA. Analysis of the other tissue atlases showed transcriptional alterations in multiple cell types in heart tissue from donors with COVID-19, and mapped cell types and genes implicated with disease severity based on COVID-19 genome-wide association studies. Our foundational dataset elucidates the biological effect of severe SARS-CoV-2 infection across the body, a key step towards new treatments. Single-cell analysis of lung, heart, kidney and liver autopsy samples shows the molecular and cellular changes and immune response resulting from severe COVID-19 infection.

380 citations

Journal ArticleDOI
TL;DR: MaAsLin 2 (Microbiome Multivariable Associations with Linear Models) as mentioned in this paper uses generalized linear and mixed models to accommodate a wide variety of modern epidemiological studies, including cross-sectional and longitudinal designs, as well as a variety of data types with or without covariates and repeated measurements.
Abstract: It is challenging to associate features such as human health outcomes, diet, environmental conditions, or other metadata to microbial community measurements, due in part to their quantitative properties. Microbiome multi-omics are typically noisy, sparse (zero-inflated), high-dimensional, extremely non-normal, and often in the form of count or compositional measurements. Here we introduce an optimized combination of novel and established methodology to assess multivariable association of microbial community features with complex metadata in population-scale observational studies. Our approach, MaAsLin 2 (Microbiome Multivariable Associations with Linear Models), uses generalized linear and mixed models to accommodate a wide variety of modern epidemiological studies, including cross-sectional and longitudinal designs, as well as a variety of data types (e.g., counts and relative abundances) with or without covariates and repeated measurements. To construct this method, we conducted a large-scale evaluation of a broad range of scenarios under which straightforward identification of meta-omics associations can be challenging. These simulation studies reveal that MaAsLin 2's linear model preserves statistical power in the presence of repeated measures and multiple covariates, while accounting for the nuances of meta-omics features and controlling false discovery. We also applied MaAsLin 2 to a microbial multi-omics dataset from the Integrative Human Microbiome (HMP2) project which, in addition to reproducing established results, revealed a unique, integrated landscape of inflammatory bowel diseases (IBD) across multiple time points and omics profiles.

369 citations

Journal ArticleDOI
20 Jan 2020
TL;DR: An unexpectedly large number of non-oncology drugs selectively inhibited subsets of cancer cell lines in a manner predictable from the cell lines' molecular features.
Abstract: Anti-cancer uses of non-oncology drugs have occasionally been found, but such discoveries have been serendipitous. We sought to create a public resource containing the growth inhibitory activity of 4,518 drugs tested across 578 human cancer cell lines. We used PRISM, a molecular barcoding method, to screen drugs against cell lines in pools. An unexpectedly large number of non-oncology drugs selectively inhibited subsets of cancer cell lines in a manner predictable from the cell lines' molecular features. Our findings include compounds that killed by inducing PDE3A-SLFN12 complex formation; vanadium-containing compounds whose killing depended on the sulfate transporter SLC26A2; the alcohol dependence drug disulfiram, which killed cells with low expression of metallothioneins; and the anti-inflammatory drug tepoxalin, which killed via the multi-drug resistance protein ABCB1. The PRISM drug repurposing resource (https://depmap.org/repurposing) is a starting point to develop new oncology therapeutics, and more rarely, for potential direct clinical translation.

347 citations

Posted ContentDOI
Pascal Barbry1, Christoph Muus2, Christoph Muus3, Malte D Luecken, Gökcen Eraslan2, Avinash Waghray3, Graham Heimberg2, Lisa Sikkema, Yoshihiko Kobayashi4, Eeshit Dhaval Vaishnav5, Ayshwarya Subramanian2, Christopher Smilie2, Karthik A. Jagadeesh2, Elizabeth Thu Duong6, Evgenij Fiskin2, Elena Torlai Triglia2, Meshal Ansari, Peiwen Cai7, Brian M. Lin3, Justin Buchanan6, Sijia Chen8, Jian Shu2, Jian Shu5, Adam L. Haber3, Adam L. Haber2, Hattie Chung2, Daniel T. Montoro2, Taylor Adams9, Hananeh Aliee, J. Samuel10, Allon Zaneta Andrusivova11, Ilias Angelidis, Orr Ashenberg2, Kevin Bassler12, Christophe Bécavin1, Inbal Benhar3, Joseph Bergenstråhle11, Ludvig Bergenstråhle11, Liam Bolt13, Emelie Braun14, Linh T. Bui15, Mark Chaffin2, Evgeny Chichelnitskiy16, Joshua Chiou6, Thomas M. Conlon, Michael S. Cuoco2, Marie Deprez1, David Fischer, Astrid Gillich, Joshua Gould2, Minzhe Guo17, Austin J. Gutierrez15, Arun C. Habermann18, Tyler Harvey2, Peng He13, Xiaomeng Hou6, Xiaomeng Hou8, Lijuan Hu14, Alok Jaiswal2, Peiyong Jiang19, Theodoros Kapellos12, Christin S. Kuo, Ludvig Larsson11, Michael Leney-Greene2, Kyungtae Lim20, Monika Litviňuková13, Monika Litviňuková21, Ji Lu19, Leif S. Ludwig2, Wendy Luo2, Henrike Maatz21, Elo Madissoon13, Lira Mamanova13, Kasidet Manakongtreecheep3, Kasidet Manakongtreecheep2, Charles-Hugo Marquette1, Ian Mbano, Alexi McAdams22, Ross J. Metzger, Ahmad N. Nabhan, Sarah K. Nyquist10, Lolita Penland, Olivier Poirion6, Sergio Poli9, Cancan Qi23, Rachel Queen24, Daniel Reichart3, Daniel Reichart25, Ivan O. Rosas9, Jonas C. Schupp9, Rahul Sinha, Rene Sit, Kamil Slowikowski3, Kamil Slowikowski2, Michal Slyper2, Neal Smith2, Neal Smith3, Alex Sountoulidis26, Maximilian Strunz, Dawei Sun20, Carlos Talavera-López13, Peng Tan2, Jessica Tantivit3, Jessica Tantivit2, Kyle J. Travaglini, Nathan R. Tucker2, Katherine A. Vernon8, Katherine A. Vernon2, Marc Wadsworth10, Julia Waldman2, Xiuting Wang7, Wenjun Yan3, William Zhao7, Carly Ziegler10 
20 Apr 2020-bioRxiv
TL;DR: Differences in the cell type-specific expression of mediators of SARS-CoV-2 viral entry may be responsible for aspects of COVID-19 epidemiology and clinical course, and point to putative molecular pathways involved in disease susceptibility and pathogenesis.
Abstract: The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, creates an urgent need for identifying molecular mechanisms that mediate viral entry, propagation, and tissue pathology. Cell membrane bound angiotensin-converting enzyme 2 (ACE2) and associated proteases, transmembrane protease serine 2 (TMPRSS2) and Cathepsin L (CTSL), were previously identified as mediators of SARS-CoV2 cellular entry. Here, we assess the cell type-specific RNA expression of ACE2 , TMPRSS2 , and CTSL through an integrated analysis of 107 single-cell and single-nucleus RNA-Seq studies, including 22 lung and airways datasets (16 unpublished), and 85 datasets from other diverse organs. Joint expression of ACE2 and the accessory proteases identifies specific subsets of respiratory epithelial cells as putative targets of viral infection in the nasal passages, airways, and alveoli. Cells that co-express ACE2 and proteases are also identified in cells from other organs, some of which have been associated with COVID-19 transmission or pathology, including gut enterocytes, corneal epithelial cells, cardiomyocytes, heart pericytes, olfactory sustentacular cells, and renal epithelial cells. Performing the first meta-analyses of scRNA-seq studies, we analyzed 1,176,683 cells from 282 nasal, airway, and lung parenchyma samples from 164 donors spanning fetal, childhood, adult, and elderly age groups, associate increased levels of ACE2 , TMPRSS2 , and CTSL in specific cell types with increasing age, male gender, and smoking, all of which are epidemiologically linked to COVID-19 susceptibility and outcomes. Notably, there was a particularly low expression of ACE2 in the few young pediatric samples in the analysis. Further analysis reveals a gene expression program shared by ACE2 + TMPRSS2 + cells in nasal, lung and gut tissues, including genes that may mediate viral entry, subtend key immune functions, and mediate epithelial-macrophage cross-talk. Amongst these are IL6, its receptor and co-receptor, IL1R , TNF response pathways, and complement genes. Cell type specificity in the lung and airways and smoking effects were conserved in mice. Our analyses suggest that differences in the cell type-specific expression of mediators of SARS-CoV-2 viral entry may be responsible for aspects of COVID-19 epidemiology and clinical course, and point to putative molecular pathways involved in disease susceptibility and pathogenesis.

244 citations


Cited by
More filters
Journal ArticleDOI

[...]

08 Dec 2001-BMJ
TL;DR: There is, I think, something ethereal about i —the square root of minus one, which seems an odd beast at that time—an intruder hovering on the edge of reality.
Abstract: There is, I think, something ethereal about i —the square root of minus one. I remember first hearing about it at school. It seemed an odd beast at that time—an intruder hovering on the edge of reality. Usually familiarity dulls this sense of the bizarre, but in the case of i it was the reverse: over the years the sense of its surreal nature intensified. It seemed that it was impossible to write mathematics that described the real world in …

33,785 citations

01 Jan 2020
TL;DR: Prolonged viral shedding provides the rationale for a strategy of isolation of infected patients and optimal antiviral interventions in the future.
Abstract: Summary Background Since December, 2019, Wuhan, China, has experienced an outbreak of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Epidemiological and clinical characteristics of patients with COVID-19 have been reported but risk factors for mortality and a detailed clinical course of illness, including viral shedding, have not been well described. Methods In this retrospective, multicentre cohort study, we included all adult inpatients (≥18 years old) with laboratory-confirmed COVID-19 from Jinyintan Hospital and Wuhan Pulmonary Hospital (Wuhan, China) who had been discharged or had died by Jan 31, 2020. Demographic, clinical, treatment, and laboratory data, including serial samples for viral RNA detection, were extracted from electronic medical records and compared between survivors and non-survivors. We used univariable and multivariable logistic regression methods to explore the risk factors associated with in-hospital death. Findings 191 patients (135 from Jinyintan Hospital and 56 from Wuhan Pulmonary Hospital) were included in this study, of whom 137 were discharged and 54 died in hospital. 91 (48%) patients had a comorbidity, with hypertension being the most common (58 [30%] patients), followed by diabetes (36 [19%] patients) and coronary heart disease (15 [8%] patients). Multivariable regression showed increasing odds of in-hospital death associated with older age (odds ratio 1·10, 95% CI 1·03–1·17, per year increase; p=0·0043), higher Sequential Organ Failure Assessment (SOFA) score (5·65, 2·61–12·23; p Interpretation The potential risk factors of older age, high SOFA score, and d-dimer greater than 1 μg/mL could help clinicians to identify patients with poor prognosis at an early stage. Prolonged viral shedding provides the rationale for a strategy of isolation of infected patients and optimal antiviral interventions in the future. Funding Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences; National Science Grant for Distinguished Young Scholars; National Key Research and Development Program of China; The Beijing Science and Technology Project; and Major Projects of National Science and Technology on New Drug Creation and Development.

4,408 citations

Journal Article
TL;DR: In this paper, the coding exons of the family of 518 protein kinases were sequenced in 210 cancers of diverse histological types to explore the nature of the information that will be derived from cancer genome sequencing.
Abstract: AACR Centennial Conference: Translational Cancer Medicine-- Nov 4-8, 2007; Singapore PL02-05 All cancers are due to abnormalities in DNA. The availability of the human genome sequence has led to the proposal that resequencing of cancer genomes will reveal the full complement of somatic mutations and hence all the cancer genes. To explore the nature of the information that will be derived from cancer genome sequencing we have sequenced the coding exons of the family of 518 protein kinases, ~1.3Mb DNA per cancer sample, in 210 cancers of diverse histological types. Despite the screen being directed toward the coding regions of a gene family that has previously been strongly implicated in oncogenesis, the results indicate that the majority of somatic mutations detected are “passengers”. There is considerable variation in the number and pattern of these mutations between individual cancers, indicating substantial diversity of processes of molecular evolution between cancers. The imprints of exogenous mutagenic exposures, mutagenic treatment regimes and DNA repair defects can all be seen in the distinctive mutational signatures of individual cancers. This systematic mutation screen and others have previously yielded a number of cancer genes that are frequently mutated in one or more cancer types and which are now anticancer drug targets (for example BRAF , PIK3CA , and EGFR ). However, detailed analyses of the data from our screen additionally suggest that there exist a large number of additional “driver” mutations which are distributed across a substantial number of genes. It therefore appears that cells may be able to utilise mutations in a large repertoire of potential cancer genes to acquire the neoplastic phenotype. However, many of these genes are employed only infrequently. These findings may have implications for future anticancer drug development.

2,737 citations

DOI
01 Jan 2020

1,967 citations

Journal ArticleDOI
TL;DR: The hallmarks of cancer conceptualization is a heuristic tool for distilling the vast complexity of cancer phenotypes and genotypes into a provisional set of underlying principles as mentioned in this paper , which are used to understand mechanisms of cancer development and malignant progression, and apply that knowledge to cancer medicine.
Abstract: The hallmarks of cancer conceptualization is a heuristic tool for distilling the vast complexity of cancer phenotypes and genotypes into a provisional set of underlying principles. As knowledge of cancer mechanisms has progressed, other facets of the disease have emerged as potential refinements. Herein, the prospect is raised that phenotypic plasticity and disrupted differentiation is a discrete hallmark capability, and that nonmutational epigenetic reprogramming and polymorphic microbiomes both constitute distinctive enabling characteristics that facilitate the acquisition of hallmark capabilities. Additionally, senescent cells, of varying origins, may be added to the roster of functionally important cell types in the tumor microenvironment. SIGNIFICANCE: Cancer is daunting in the breadth and scope of its diversity, spanning genetics, cell and tissue biology, pathology, and response to therapy. Ever more powerful experimental and computational tools and technologies are providing an avalanche of "big data" about the myriad manifestations of the diseases that cancer encompasses. The integrative concept embodied in the hallmarks of cancer is helping to distill this complexity into an increasingly logical science, and the provisional new dimensions presented in this perspective may add value to that endeavor, to more fully understand mechanisms of cancer development and malignant progression, and apply that knowledge to cancer medicine.

1,838 citations