Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Semir Beyaz is active.

Publication


Featured researches published by Semir Beyaz.


Nature | 2016

High-fat diet enhances stemness and tumorigenicity of intestinal progenitors

Semir Beyaz; Miyeko D. Mana; Jatin Roper; Dmitriy Kedrin; Assieh Saadatpour; Sue-Jean Hong; Khristian E. Bauer-Rowe; Michael E. Xifaras; Adam Akkad; Erika Arias; Luca Pinello; Yarden Katz; Shweta Shinagare; Monther Abu-Remaileh; Maria M. Mihaylova; Dudley W. Lamming; Rizkullah Dogum; Guoji Guo; George W. Bell; Martin K. Selig; G. Petur Nielsen; Nitin Gupta; Cristina R. Ferrone; Vikram Deshpande; Guo-Cheng Yuan; Stuart H. Orkin; David M. Sabatini; Ömer H. Yilmaz

Little is known about how pro-obesity diets regulate tissue stem and progenitor cell function. Here we show that high-fat diet (HFD)-induced obesity augments the numbers and function of Lgr5+ intestinal stem cells of the mammalian intestine. Mechanistically, a HFD induces a robust peroxisome proliferator-activated receptor delta (PPAR-δ) signature in intestinal stem cells and progenitor cells (non-intestinal stem cells), and pharmacological activation of PPAR-δ recapitulates the effects of a HFD on these cells. Like a HFD, ex vivo treatment of intestinal organoid cultures with fatty acid constituents of the HFD enhances the self-renewal potential of these organoid bodies in a PPAR-δ-dependent manner. Notably, HFD- and agonist-activated PPAR-δ signalling endow organoid-initiating capacity to progenitors, and enforced PPAR-δ signalling permits these progenitors to form in vivo tumours after loss of the tumour suppressor Apc. These findings highlight how diet-modulated PPAR-δ activation alters not only the function of intestinal stem and progenitor cells, but also their capacity to initiate tumours.


Nature | 2017

A single-cell survey of the small intestinal epithelium

Adam L. Haber; Moshe Biton; Noga Rogel; Rebecca H. Herbst; Karthik Shekhar; Christopher Smillie; Grace Burgin; Toni Delorey; Michael R. Howitt; Yarden Katz; Itay Tirosh; Semir Beyaz; Danielle Dionne; Mei Zhang; Raktima Raychowdhury; Wendy S. Garrett; Orit Rozenblatt-Rosen; Hai Ning Shi; Ömer H. Yilmaz; Ramnik J. Xavier; Aviv Regev

Intestinal epithelial cells absorb nutrients, respond to microbes, function as a barrier and help to coordinate immune responses. Here we report profiling of 53,193 individual epithelial cells from the small intestine and organoids of mice, which enabled the identification and characterization of previously unknown subtypes of intestinal epithelial cell and their gene signatures. We found unexpected diversity in hormone-secreting enteroendocrine cells and constructed the taxonomy of newly identified subtypes, and distinguished between two subtypes of tuft cell, one of which expresses the epithelial cytokine Tslp and the pan-immune marker CD45, which was not previously associated with non-haematopoietic cells. We also characterized the ways in which cell-intrinsic states and the proportions of different cell types respond to bacterial and helminth infections: Salmonella infection caused an increase in the abundance of Paneth cells and enterocytes, and broad activation of an antimicrobial program; Heligmosomoides polygyrus caused an increase in the abundance of goblet and tuft cells. Our survey highlights previously unidentified markers and programs, associates sensory molecules with cell types, and uncovers principles of gut homeostasis and response to pathogens.


Molecular Cell | 2014

Distinct and Combinatorial Functions of Jmjd2b/Kdm4b and Jmjd2c/Kdm4c in Mouse Embryonic Stem Cell Identity

Partha P. Das; Zhen Shao; Semir Beyaz; Eftychia Apostolou; Luca Pinello; Alejandro De Los Angeles; Kassandra O’Brien; Jennifer Marino Atsma; Yuko Fujiwara; Minh Nguyen; Damir Ljuboja; Guoji Guo; Andrew J. Woo; Guo-Cheng Yuan; Tamer T. Onder; George Q. Daley; Jonghwan Kim; Stuart H. Orkin

Self-renewal and pluripotency of embryonic stem cells (ESCs) are established by multiple regulatory pathways operating at several levels. The roles of histone demethylases (HDMs) in these programs are incompletely defined. We conducted a functional RNAi screen for HDMs and identified five potential HDMs essential for mouse ESC identity. In-depth analyses demonstrate that the closely related HDMs Jmjd2b and Jmjd2c are necessary for self-renewal of ESCs and induced pluripotent stem cell generation. Genome-wide occupancy studies reveal that Jmjd2b unique, Jmjd2c unique, and Jmjd2b-Jmjd2c common target sites belong to functionally separable Core, Polycomb repressive complex (PRC), and Myc regulatory modules, respectively. Jmjd2b and Nanog act through an interconnected regulatory loop, whereas Jmjd2c assists PRC2 in transcriptional repression. Thus, two HDMs of the same subclass exhibit distinct and combinatorial functions in control of the ESC state. Such complexity of HDM function reveals an aspect of multilayered transcriptional control.


Nature Biotechnology | 2017

In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis

Jatin Roper; Tuomas Tammela; Naniye Malli Cetinbas; Adam Akkad; Ali Roghanian; Steffen Rickelt; Mohammad Almeqdadi; Katherine Wu; Matthias A. Oberli; Francisco J. Sánchez-Rivera; Yoona Park; Xu Liang; George Eng; Martin S. Taylor; Roxana Azimi; Dmitriy Kedrin; Rachit Neupane; Semir Beyaz; Ewa Sicinska; Yvelisse Suarez; James Yoo; Lillian Chen; Lawrence R. Zukerberg; Pekka Katajisto; Vikram Deshpande; Adam J. Bass; Philip N. Tsichlis; Jacqueline A. Lees; Robert Langer; Richard O. Hynes

In vivo interrogation of the function of genes implicated in tumorigenesis is limited by the need to generate and cross germline mutant mice. Here we describe approaches to model colorectal cancer (CRC) and metastasis, which rely on in situ gene editing and orthotopic organoid transplantation in mice without cancer-predisposing mutations. Autochthonous tumor formation is induced by CRISPR-Cas9-based editing of the Apc and Trp53 tumor suppressor genes in colon epithelial cells and by orthotopic transplantation of Apc-edited colon organoids. ApcΔ/Δ;KrasG12D/+;Trp53Δ/Δ (AKP) mouse colon organoids and human CRC organoids engraft in the distal colon and metastasize to the liver. Finally, we apply the orthotopic transplantation model to characterize the clonal dynamics of Lgr5+ stem cells and demonstrate sequential activation of an oncogene in established colon adenomas. These experimental systems enable rapid in vivo characterization of cancer-associated genes and reproduce the entire spectrum of tumor progression and metastasis.


Blood | 2011

LAG-3, TGF-β, and cell-intrinsic PD-1 inhibitory pathways contribute to CD8 but not CD4 T cell tolerance induced by allogeneic BMT with anti-CD40L

Carrie L. Lucas; Creg J. Workman; Semir Beyaz; Samuel LoCascio; Guiling Zhao; Dario A. A. Vignali; Megan Sykes

Administration of a single dose of anti-CD40L mAb at the time of allogeneic BM transplantation tolerizes peripheral alloreactive T cells and permits establishment of mixed hematopoietic chimerism in mice. Once engrafted, mixed chimeras are systemically tolerant to donor Ags through a central deletion mechanism and will accept any donor organ indefinitely. We previously found that the PD-1/PD-L1 pathway is required for CD8 T-cell tolerance in this model. However, the cell population that must express PD-1 and the role of other inhibitory molecules were unknown. Here, we report that LAG-3 is required for long-term peripheral CD8 but not CD4 T-cell tolerance and that this requirement is CD8 cell-extrinsic. In contrast, adoptive transfer studies revealed a CD8 T cell-intrinsic requirement for CTLA4/B7.1/B7.2 and for PD-1 for CD8 T-cell tolerance induction. We also observed that both PD-L1 and PD-L2 are independently required on donor cells to achieve T-cell tolerance. Finally, we uncovered a requirement for TGF-β signaling into T cells to achieve peripheral CD8 but not CD4 T-cell tolerance in this in vivo system.


Nature Immunology | 2016

The histone demethylase UTX regulates the lineage-specific epigenetic program of invariant natural killer T cells

Semir Beyaz; Ji Hyung Kim; Luca Pinello; Michael E. Xifaras; Yu Hu; Jialiang Huang; Marc A. Kerenyi; Partha P. Das; R. Anthony Barnitz; Aurelie Herault; Rizkullah Dogum; W. Nicholas Haining; Ömer H. Yilmaz; Emmanuelle Passegué; Guo-Cheng Yuan; Stuart H. Orkin; Florian Winau

Invariant natural killer T cells (iNKT cells) are innate-like lymphocytes that protect against infection, autoimmune disease and cancer. However, little is known about the epigenetic regulation of iNKT cell development. Here we found that the H3K27me3 histone demethylase UTX was an essential cell-intrinsic factor that controlled an iNKT-cell lineage-specific gene-expression program and epigenetic landscape in a demethylase-activity-dependent manner. UTX-deficient iNKT cells exhibited impaired expression of iNKT cell signature genes due to a decrease in activation-associated H3K4me3 marks and an increase in repressive H3K27me3 marks within the promoters occupied by UTX. We found that JunB regulated iNKT cell development and that the expression of genes that were targets of both JunB and the iNKT cell master transcription factor PLZF was UTX dependent. We identified iNKT cell super-enhancers and demonstrated that UTX-mediated regulation of super-enhancer accessibility was a key mechanism for commitment to the iNKT cell lineage. Our findings reveal how UTX regulates the development of iNKT cells through multiple epigenetic mechanisms.


American Journal of Transplantation | 2014

Allospecific Rejection of MHC Class I‐Deficient Bone Marrow by CD8 T Cells

Fabienne Haspot; Hao Wei Li; Carrie L. Lucas; Thomas Fehr; Semir Beyaz; Megan Sykes

Avoidance of long‐term immunosuppression is a desired goal in organ transplantation. Mixed chimerism offers a promising approach to tolerance induction, and we have aimed to develop low‐toxicity, nonimmunodepleting approaches to achieve this outcome. In a mouse model achieving fully MHC‐mismatched allogeneic bone marrow engraftment with minimal conditioning (3 Gy total body irradiation followed by anti‐CD154 and T cell–depleted allogeneic bone marrow cells), CD4 T cells in the recipient are required to promote tolerance of preexisting alloreactive recipient CD8 T cells and thereby permit chimerism induction. We now demonstrate that mice devoid of CD4 T cells and NK cells reject MHC Class I‐deficient and Class I/Class II‐deficient marrow in a CD8 T cell–dependent manner. This rejection is specific for donor alloantigens, since recipient hematopoiesis is not affected by donor marrow rejection and MHC Class I‐deficient bone marrow that is syngeneic to the recipient is not rejected. Recipient CD8 T cells are activated and develop cytotoxicity against MHC Class I‐deficient donor cells in association with rejection. These data implicate a novel CD8 T cell–dependent bone marrow rejection pathway, wherein recipient CD8 T cells indirectly activated by donor alloantigens promote direct killing, in a T cell receptor–independent manner, of Class I‐deficient donor cells.


Clinical Cancer Research | 2016

Molecular Pathways: Dietary regulation of stemness and tumor initiation by the PPAR-δ pathway.

Semir Beyaz; Ömer H. Yilmaz

Peroxisome proliferator-activated receptor delta (PPAR-δ) is a nuclear receptor transcription factor that regulates gene expression during development and disease states, such as cancer. However, the precise role of PPAR-δ during tumorigenesis is not well understood. Recent data suggest that PPAR-δ may have context-specific oncogenic and tumor-suppressive roles depending on the tissue, cell-type, or diet-induced physiology in question. For example, in the intestine, pro-obesity diets, such as a high-fat diet (HFD), are associated with increased colorectal cancer incidence. Interestingly, many of the effects of an HFD in the stem and progenitor cell compartment are driven by a robust PPAR-δ program and contribute to the early steps of intestinal tumorigenesis. Importantly, the PPAR-δ pathway or its downstream mediators may serve as therapeutic intervention points or biomarkers in colon cancer that arise in patients who are obese. Although potent PPAR-δ agonists and antagonists exist, their clinical utility may be enhanced by uncovering how PPAR-δ mediates tumorigenesis in diverse tissues and cell types as well as in response to diet. Clin Cancer Res; 22(23); 5636–41. ©2016 AACR.


bioRxiv | 2017

T helper cells modulate intestinal stem cell renewal and differentiation

Moshe Biton; Adam L. Haber; Semir Beyaz; Noga Rogel; Christopher Smillie; Karthik Shekhar; Alexandra Schnell; Zuojia Chen; Chuan Wu; Jose Ordovas-Montanes; David Alvarez; Rebecca H. Herbst; Itay Tirosh; Grace Burgin; Danielle Dionne; Michael E. Xifaras; Mei Zhang; Alex K. Shalek; Ulrich H. von Andrian; Daniel B. Graham; Orit Rozenblatt-Rosen; Hai Ning Shi; Vijay K. Kuchroo; Omer Yilmaz; Aviv Regev; Ramnik J. Xavier

In the small intestine, a cellular niche of diverse accessory cell types supports the rapid generation of mature epithelial cell types through self-renewal, proliferation, and differentiation of intestinal stem cells (ISCs). However, not much is known about interactions between immune cells and ISCs, and it is unclear if and how immune cell dynamics affect eventual ISC fate or the balance between self-renewal and differentiation. Here, we used single-cell RNA-seq (scRNA-Seq) of intestinal epithelial cells (IECs) to identify new mechanisms for ISC–immune cell interactions. Surprisingly, MHC class II (MHCII) is enriched in two distinct subsets of Lgr5+ crypt base columnar ISCs, which are also distinguished by higher proliferation rates. Using co-culture of T cells with intestinal organoids, cytokine stimulations, and in vivo mouse models, we confirm that CD4+ T helper (Th) cells communicate with ISCs and affect their differentiation, in a manner specific to the Th subtypes and their signature cytokines and dependent on MHCII expression by ISCs. Specific inducible knockout of MHCII in intestinal epithelial cells in mice in vivo results in expansion of the ISC pool. Mice lacking T cells have expanded ISC pools, whereas specific depletion of Treg cells in vivo results in substantial reduction of ISC numbers. Our findings show that interactions between Th cells and ISCs mediated via MHCII expressed in intestinal epithelial stem cells help orchestrate tissue-wide responses to external signals.


Nature Biotechnology | 2017

Corrigendum: In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis

Jatin Roper; Tuomas Tammela; Naniye Malli Cetinbas; Adam Akkad; Ali Roghanian; Steffen Rickelt; Mohammad Almeqdadi; Katherine Wu; Matthias A. Oberli; Francisco J. Sánchez-Rivera; Yoona Park; Xu Liang; George Eng; Martin S. Taylor; Roxana Azimi; Dmitriy Kedrin; Rachit Neupane; Semir Beyaz; Ewa Sicinska; Yvelisse Suarez; James J. Yoo; Lillian Chen; Lawrence R. Zukerberg; Pekka Katajisto; Vikram Deshpande; Adam J. Bass; Philip N. Tsichlis; Jacqueline A. Lees; Robert Langer; Richard O. Hynes

This corrects the article DOI: 10.1038/nbt.3836

Collaboration


Dive into the Semir Beyaz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ömer H. Yilmaz

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Adam Akkad

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael E. Xifaras

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge