scispace - formally typeset
Search or ask a question
Author

Rachelly Normand

Other affiliations: Rappaport Faculty of Medicine
Bio: Rachelly Normand is an academic researcher from Technion – Israel Institute of Technology. The author has contributed to research in topics: Progenitor cell & Haematopoiesis. The author has an hindex of 4, co-authored 9 publications receiving 146 citations. Previous affiliations of Rachelly Normand include Rappaport Faculty of Medicine.

Papers
More filters
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 Ganna2, Andrea Ganna4, James Gomez1, Johannes C. Melms3, Igor Katsyv3, Erica Normandin2, Erica Normandin1, Pourya Naderi5, Pourya Naderi2, Yury Popov2, Yury Popov5, Siddharth S. Raju1, Siddharth S. Raju2, Sebastian Niezen5, Sebastian Niezen2, Linus T.-Y. Tsai, Katherine J. Siddle2, Katherine J. Siddle1, Malika Sud1, Victoria M. Tran1, Shamsudheen K. Vellarikkal1, Shamsudheen K. Vellarikkal6, Yiping Wang3, Liat Amir-Zilberstein1, Deepak Atri1, Deepak Atri6, Joseph M. Beechem7, Olga R. Brook5, Jonathan H. Chen2, Jonathan H. Chen1, 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. Hecht5, Jonathan L. Hecht2, Tyler Hether7, Judit Jané-Valbuena1, Michael Leney-Greene1, Hui Ma2, Hui Ma1, Cristin McCabe1, Daniel E. McLoughlin2, Eric M. Miller7, Christoph Muus1, Christoph Muus2, Mari Niemi4, Robert F. Padera10, Robert F. Padera2, Robert F. Padera6, 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. Gupta1, Rajat M. Gupta6, Nir Hacohen1, Nir Hacohen2, Hanina Hibshoosh3, Winston Hide, Alkes L. Price2, Jayaraj Rajagopal2, Purushothama Rao Tata11, Stefan Riedel2, Stefan Riedel5, Gyongyi Szabo2, Gyongyi Szabo5, Gyongyi Szabo1, Timothy L. Tickle1, Patrick T. Ellinor1, Deborah T. Hung2, Deborah T. Hung1, Pardis C. Sabeti, Richard M. Novak12, Robert S. Rogers2, Robert S. Rogers5, Donald E. Ingber13, Donald E. Ingber2, Donald E. Ingber12, Z. Gordon Jiang5, Z. Gordon Jiang2, 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-Rosen1, Orit Rozenblatt-Rosen14, 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: Find In Translation (FIT) is presented, a statistical methodology that leverages public gene expression data to extrapolate the results of a new mouse experiment to expression changes in the equivalent human condition and predicted novel disease-associated genes.
Abstract: Cross-species differences form barriers to translational research that ultimately hinder the success of clinical trials, yet knowledge of species differences has yet to be systematically incorporated in the interpretation of animal models. Here we present Found In Translation (FIT; http://www.mouse2man.org ), a statistical methodology that leverages public gene expression data to extrapolate the results of a new mouse experiment to expression changes in the equivalent human condition. We applied FIT to data from mouse models of 28 different human diseases and identified experimental conditions in which FIT predictions outperformed direct cross-species extrapolation from mouse results, increasing the overlap of differentially expressed genes by 20–50%. FIT predicted novel disease-associated genes, an example of which we validated experimentally. FIT highlights signals that may otherwise be missed and reduces false leads, with no experimental cost. The machine learning approach FIT leverages public mouse and human expression data to improve the translation of mouse model results to analogous human disease.

52 citations

Posted ContentDOI
Toni Delorey1, Carly G. K. Ziegler, Graham Heimberg1, Rachelly Normand, Yiming Yang2, Yiming Yang1, 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, Zohar Bloom-Ackermann1, Nick Barkas1, Andrea Ganna3, Andrea Ganna2, James Gomez1, Erica Normandin2, Erica Normandin1, Pourya Naderi2, Pourya Naderi4, Yury Popov2, Yury Popov4, Siddharth S. Raju1, Siddharth S. Raju2, Sebastian Niezen4, Sebastian Niezen2, Linus T.-Y. Tsai, Katherine J. Siddle1, Katherine J. Siddle2, Malika Sud1, Victoria M. Tran1, Shamsudheen K. Vellarikkal1, Shamsudheen K. Vellarikkal5, Liat Amir-Zilberstein1, Joseph M. Beechem6, Olga R. Brook4, Jonathan H. Chen2, Jonathan H. Chen1, Prajan Divakar6, Phylicia Dorceus1, Jesse M. Engreitz7, Jesse M. Engreitz1, Adam Essene4, Donna M. Fitzgerald2, Robin Fropf6, Steven Gazal8, Joshua Gould1, Tyler Harvey1, Jonathan L. Hecht4, Jonathan L. Hecht2, Tyler Hether6, Judit Jané-Valbuena1, Michael Leney-Greene1, Hui Ma1, Hui Ma2, Cristin McCabe1, Daniel E. McLoughlin2, Eric M. Miller6, Christoph Muus2, Christoph Muus1, Mari Niemi3, Robert F. Padera9, Robert F. Padera2, Robert F. Padera5, Liuliu Pan6, Deepti Pant4, Jenna Pfiffner-Borges1, Christopher J. Pinto2, Jason Reeves6, Marty Ross6, Melissa Rudy1, Erroll H. Rueckert6, Michelle Siciliano5, Alexander Sturm1, Ellen Todres1, Avinash Waghray2, Sarah Warren6, Shuting Zhang1, Dan Zollinger6, Lisa A. Cosimi5, Rajat M. Gupta5, Rajat M. Gupta1, Nir Hacohen2, Nir Hacohen1, Winston Hide, Alkes L. Price2, Jayaraj Rajagopal2, Purushothama Rao Tata10, Stefan Riedel2, Stefan Riedel4, Gyongyi Szabo1, Gyongyi Szabo2, Gyongyi Szabo4, Timothy L. Tickle1, Deborah T. Hung1, Deborah T. Hung2, Pardis C. Sabeti, Richard M. Novak11, Robert S. Rogers4, Robert S. Rogers2, Donald E. Ingber11, Donald E. Ingber12, Donald E. Ingber2, Z. Gordon Jiang2, Z. Gordon Jiang4, Dejan Juric2, Mehrtash Babadi1, Samouil L. Farhi1, James R. Stone2, Ioannis S. Vlachos, Isaac H. Solomon5, Orr Ashenberg1, Caroline B. M. Porter1, Bo Li2, Bo Li1, Alex K. Shalek, Alexandra-Chloé Villani, Orit Rozenblatt-Rosen1, Orit Rozenblatt-Rosen13, Aviv Regev 
25 Feb 2021-bioRxiv
TL;DR: In this paper, the authors used single-cell and single-nucleus RNA-Seq of lung, kidney, liver, and heart tissues collected during the clinical autopsy of 17 individuals who succumbed to the SARS-CoV-2 pandemic.
Abstract: The SARS-CoV-2 pandemic has caused over 1 million deaths globally, mostly due to acute lung injury and acute respiratory distress syndrome, or direct complications resulting in multiple-organ failures. Little is known about the host tissue immune and cellular responses associated with COVID-19 infection, symptoms, and lethality. To address this, we collected tissues from 11 organs during the clinical autopsy of 17 individuals who succumbed to COVID-19, resulting in a tissue bank of approximately 420 specimens. We generated comprehensive cellular maps capturing COVID-19 biology related to patients' demise through single-cell and single-nucleus RNA-Seq of lung, kidney, liver and heart tissues, and further contextualized our findings through spatial RNA profiling of distinct lung regions. We developed a computational framework that incorporates removal of ambient RNA and automated cell type annotation to facilitate comparison with other healthy and diseased tissue atlases. In the lung, we uncovered significantly altered transcriptional programs within the epithelial, immune, and stromal compartments and cell intrinsic changes in multiple cell types relative to lung tissue from healthy controls. We observed evidence of: alveolar type 2 (AT2) differentiation replacing depleted alveolar type 1 (AT1) lung epithelial cells, as previously seen in fibrosis; a concomitant increase in myofibroblasts reflective of defective tissue repair; and, putative TP63 + intrapulmonary basal-like progenitor (IPBLP) cells, similar to cells identified in H1N1 influenza, that may serve as an emergency cellular reserve for severely damaged alveoli. Together, these findings suggest the activation and failure of multiple avenues for regeneration of the epithelium in these terminal lungs. SARS-CoV-2 RNA reads were enriched in lung mononuclear phagocytic cells and endothelial cells, and these cells expressed distinct host response transcriptional programs. We corroborated the compositional and transcriptional changes in lung tissue through spatial analysis of RNA profiles in situ and distinguished unique tissue host responses between regions with and without viral RNA, and in COVID-19 donor tissues relative to healthy lung. Finally, we analyzed genetic regions implicated in COVID-19 GWAS with transcriptomic data to implicate specific cell types and genes associated with disease severity. Overall, our COVID-19 cell atlas is a foundational dataset to better understand the biological impact of SARS-CoV-2 infection across the human body and empowers the identification of new therapeutic interventions and prevention strategies.

33 citations

Journal ArticleDOI
TL;DR: In this paper, a single-cell RNA sequencing (scRNA-seq) analysis was performed on HSPC obtained from the bone marrow (BM) compartment of met-low and met-high tumors.
Abstract: Background Metastasis is the major cause of death in patients with cancer. Myeloid skewing of hematopoietic cells is a prominent promoter of metastasis. However, the reservoir of these cells in the bone marrow (BM) compartment and their differentiation pattern from hematopoietic stem and progenitor cells (HSPCs) have not been explored. Methods We used a unique model system consisting of tumor cell clones with low metastatic potential or high metastatic potential (met-low and met-high, respectively) to investigate the fate of HSPC differentiation using murine melanoma and breast carcinoma. Single-cell RNA sequencing (scRNA-seq) analysis was performed on HSPC obtained from the BM of met-low and met-high tumors. A proteomic screen of tumor-conditioned medium integrated with the scRNA-seq data analysis was performed to analyze the potential cross talk between cancer cells and HSPCs. Adoptive transfer of tumor-educated HSPC subsets obtained from green fluorescent protein (GFP)+ tagged mice was then carried out to identify the contribution of committed HSPCs to tumor spread. Peripheral mononuclear cells obtained from patients with breast and lung cancer were analyzed for HSPC subsets. Results Mice bearing met-high tumors exhibited a significant increase in the percentage of HSPCs in the BM in comparison with tumor-free mice or mice bearing met-low tumors. ScRNA-seq analysis of these HSPCs revealed that met-high tumors enriched the monocyte-dendritic progenitors (MDPs) but not granulocyte-monocyte progenitors (GMPs). A proteomic screen of tumor- conditioned medium integrated with the scRNA-seq data analysis revealed that the interleukin 6 (IL-6)–IL-6 receptor axis is highly active in HSPC-derived MDP cells. Consequently, loss of function and gain of function of IL-6 in tumor cells resulted in decreased and increased metastasis and corresponding MDP levels, respectively. Importantly, IL-6-educated MDPs induce metastasis within mice bearing met-low tumors—through further differentiation into immunosuppressive macrophages and not dendritic cells. Consistently, MDP but not GMP levels in peripheral blood of breast and lung cancer patients are correlated with tumor aggressiveness. Conclusions Our study reveals a new role for tumor-derived IL-6 in hijacking the HSPC differentiation program toward prometastatic MDPs that functionally differentiate into immunosuppressive monocytes to support the metastatic switch.

15 citations

Book ChapterDOI
TL;DR: An introduction for the domains of analyses possible using high-throughput sequencing, distinguishing between "counting" and "reading" applications is provided.
Abstract: The dramatic fall in the cost of DNA sequencing has revolutionized the experiments within reach in the life sciences. Here we provide an introduction for the domains of analyses possible using high-throughput sequencing, distinguishing between "counting" and "reading" applications. We discuss the steps in designing a high-throughput sequencing experiment, introduce the most widely used applications, and describe basic sequencing concepts. We review the various software programs available for many of the bioinformatics analysis required to make sense of the sequencing data. We hope that this introduction will be accessible to biologists with no previous background in bioinformatics, yet with a keen interest in applying the power of high-throughput sequencing in their research.

14 citations


Cited by
More filters
01 Jan 2011
TL;DR: The sheer volume and scope of data posed by this flood of data pose a significant challenge to the development of efficient and intuitive visualization tools able to scale to very large data sets and to flexibly integrate multiple data types, including clinical data.
Abstract: Rapid improvements in sequencing and array-based platforms are resulting in a flood of diverse genome-wide data, including data from exome and whole-genome sequencing, epigenetic surveys, expression profiling of coding and noncoding RNAs, single nucleotide polymorphism (SNP) and copy number profiling, and functional assays. Analysis of these large, diverse data sets holds the promise of a more comprehensive understanding of the genome and its relation to human disease. Experienced and knowledgeable human review is an essential component of this process, complementing computational approaches. This calls for efficient and intuitive visualization tools able to scale to very large data sets and to flexibly integrate multiple data types, including clinical data. However, the sheer volume and scope of data pose a significant challenge to the development of such tools.

2,187 citations

Journal ArticleDOI
TL;DR: In this paper , the authors used national healthcare databases from the US Department of Veterans Affairs to build a cohort of 153,760 individuals with COVID-19, as well as two sets of control cohorts with 5,637,647 (contemporary controls) and 5,859,411 (historical controls) individuals, to estimate risks and 1-year burdens of a set of pre-specified incident cardiovascular outcomes.
Abstract: The cardiovascular complications of acute coronavirus disease 2019 (COVID-19) are well described, but the post-acute cardiovascular manifestations of COVID-19 have not yet been comprehensively characterized. Here we used national healthcare databases from the US Department of Veterans Affairs to build a cohort of 153,760 individuals with COVID-19, as well as two sets of control cohorts with 5,637,647 (contemporary controls) and 5,859,411 (historical controls) individuals, to estimate risks and 1-year burdens of a set of pre-specified incident cardiovascular outcomes. We show that, beyond the first 30 d after infection, individuals with COVID-19 are at increased risk of incident cardiovascular disease spanning several categories, including cerebrovascular disorders, dysrhythmias, ischemic and non-ischemic heart disease, pericarditis, myocarditis, heart failure and thromboembolic disease. These risks and burdens were evident even among individuals who were not hospitalized during the acute phase of the infection and increased in a graded fashion according to the care setting during the acute phase (non-hospitalized, hospitalized and admitted to intensive care). Our results provide evidence that the risk and 1-year burden of cardiovascular disease in survivors of acute COVID-19 are substantial. Care pathways of those surviving the acute episode of COVID-19 should include attention to cardiovascular health and disease.

826 citations

Journal ArticleDOI
29 Apr 2021-Nature
TL;DR: In this paper, the authors performed single-nucleus RNA sequencing of about 116,000 nuclei from the lungs of nineteen individuals who died of COVID-19 and underwent rapid autopsy and seven control individuals.
Abstract: Respiratory failure is the leading cause of death in patients with severe SARS-CoV-2 infection1,2, but the host response at the lung tissue level is poorly understood. Here we performed single-nucleus RNA sequencing of about 116,000 nuclei from the lungs of nineteen individuals who died of COVID-19 and underwent rapid autopsy and seven control individuals. Integrated analyses identified substantial alterations in cellular composition, transcriptional cell states, and cell-to-cell interactions, thereby providing insight into the biology of lethal COVID-19. The lungs from individuals with COVID-19 were highly inflamed, with dense infiltration of aberrantly activated monocyte-derived macrophages and alveolar macrophages, but had impaired T cell responses. Monocyte/macrophage-derived interleukin-1β and epithelial cell-derived interleukin-6 were unique features of SARS-CoV-2 infection compared to other viral and bacterial causes of pneumonia. Alveolar type 2 cells adopted an inflammation-associated transient progenitor cell state and failed to undergo full transition into alveolar type 1 cells, resulting in impaired lung regeneration. Furthermore, we identified expansion of recently described CTHRC1+ pathological fibroblasts3 contributing to rapidly ensuing pulmonary fibrosis in COVID-19. Inference of protein activity and ligand–receptor interactions identified putative drug targets to disrupt deleterious circuits. This atlas enables the dissection of lethal COVID-19, may inform our understanding of long-term complications of COVID-19 survivors, and provides an important resource for therapeutic development. Lung samples collected soon after death from COVID-19 are used to provide a single-cell atlas of SARS-CoV-2 infection and the ensuing molecular changes.

286 citations

Journal ArticleDOI
11 Mar 2022-Science
TL;DR: It is hoped that the knowledge gained from this COVID-19 research will be applied in studies of inflammatory processes involved in critical and chronic illnesses, which remain a major unmet need.
Abstract: Considerable research effort has been made worldwide to decipher the immune response triggered upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, identify the drivers of severe and fatal COVID-19, and understand what leads to the prolongation of symptoms after disease resolution. We review the results of almost 2 years of COVID-19 immunology research and discuss definitive findings and remaining questions regarding our understanding of COVID-19 pathophysiology. We discuss emerging understanding of differences in immune responses seen in those with and without Long Covid syndrome, also known as post-acute sequelae of SARS-CoV-2. We hope that the knowledge gained from this COVID-19 research will be applied in studies of inflammatory processes involved in critical and chronic illnesses, which remain a major unmet need. Description

282 citations

Journal ArticleDOI
TL;DR: In this paper , the authors explore recent clinical and experimental advances regarding SARS-CoV-2 pathophysiology and discuss potential mechanisms behind acute respiratory distress syndrome (ARDS), specifically focusing on new insights obtained using novel technologies such as single-cell omics, organoid infection models and CRISPR screens.
Abstract: The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a devastating pandemic. Although most people infected with SARS-CoV-2 develop a mild to moderate disease with virus replication restricted mainly to the upper airways, some progress to having a life-threatening pneumonia. In this Review, we explore recent clinical and experimental advances regarding SARS-CoV-2 pathophysiology and discuss potential mechanisms behind SARS-CoV-2-associated acute respiratory distress syndrome (ARDS), specifically focusing on new insights obtained using novel technologies such as single-cell omics, organoid infection models and CRISPR screens. We describe how SARS-CoV-2 may infect the lower respiratory tract and cause alveolar damage as a result of dysfunctional immune responses. We discuss how this may lead to the induction of a 'leaky state' of both the epithelium and the endothelium, promoting inflammation and coagulation, while an influx of immune cells leads to overexuberant inflammatory responses and immunopathology. Finally, we highlight how these findings may aid the development of new therapeutic interventions against COVID-19.

221 citations