Network


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

Hotspot


Dive into the research topics where Lauren F. Young is active.

Publication


Featured researches published by Lauren F. Young.


Journal of Experimental Medicine | 2012

Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation

Robert R. Jenq; Carles Ubeda; Ying Taur; Clarissa C. Menezes; Raya Khanin; Jarrod A. Dudakov; Chen Liu; Mallory L. West; Natalie V. Singer; Michele Equinda; Asia Gobourne; Lauren Lipuma; Lauren F. Young; Odette M. Smith; Arnab Ghosh; Alan M. Hanash; Jenna D. Goldberg; Kazutoshi Aoyama; Bruce R. Blazar; Eric G. Pamer; Marcel R.M. van den Brink

GVHD is associated with significant shifts in the composition of the intestinal microbiota in human and mouse models; manipulating the microbiota can alter the severity of GVHD in mice.


Immunity | 2012

Interleukin-22 Protects Intestinal Stem Cells from Immune-Mediated Tissue Damage and Regulates Sensitivity to Graft versus Host Disease

Alan M. Hanash; Jarrod A. Dudakov; Guoqiang Hua; Margaret H. O’Connor; Lauren F. Young; Natalie V. Singer; Mallory L. West; Robert R. Jenq; Amanda M. Holland; Lucy W. Kappel; Arnab Ghosh; Jennifer J. Tsai; Uttam K. Rao; Nury Yim; Odette M. Smith; Enrico Velardi; Elena B. Hawryluk; George F. Murphy; Chen Liu; Lynette A. Fouser; Richard Kolesnick; Bruce R. Blazar; Marcel R.M. van den Brink

Little is known about the maintenance of intestinal stem cells (ISCs) and progenitors during immune-mediated tissue damage or about the susceptibility of transplant recipients to tissue damage mediated by the donor immune system during graft versus host disease (GVHD). We demonstrate here that deficiency of recipient-derived IL-22 increased acute GVHD tissue damage and mortality, that ISCs were eliminated during GVHD, and that ISCs as well as their downstream progenitors expressed the IL-22 receptor. Intestinal IL-22 was produced after bone marrow transplant by IL-23-responsive innate lymphoid cells (ILCs) from the transplant recipients, and intestinal IL-22 increased in response to pretransplant conditioning. However, ILC frequency and IL-22 amounts were decreased by GVHD. Recipient IL-22 deficiency led to increased crypt apoptosis, depletion of ISCs, and loss of epithelial integrity. Our findings reveal IL-22 as a critical regulator of tissue sensitivity to GVHD and a protective factor for ISCs during inflammatory intestinal damage.


Nature | 2015

Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration

Caroline A. Lindemans; Marco Calafiore; Anna Mertelsmann; Margaret H. O’Connor; Jarrod A. Dudakov; Robert R. Jenq; Enrico Velardi; Lauren F. Young; Odette M. Smith; Gillian Lawrence; Juliet Ivanov; Ya-Yuan Fu; Shuichiro Takashima; Guoqiang Hua; Maria Laura Martin; Kevin P. O’Rourke; Yuan-Hung Lo; Michal Mokry; Monica Romera-Hernandez; Lukas E. Dow; Edward E. S. Nieuwenhuis; Noah F. Shroyer; Chen Liu; Richard Kolesnick; Marcel R.M. van den Brink; Alan M. Hanash

Epithelial regeneration is critical for barrier maintenance and organ function after intestinal injury. The intestinal stem cell (ISC) niche provides Wnt, Notch and epidermal growth factor (EGF) signals supporting Lgr5+ crypt base columnar ISCs for normal epithelial maintenance. However, little is known about the regulation of the ISC compartment after tissue damage. Using ex vivo organoid cultures, here we show that innate lymphoid cells (ILCs), potent producers of interleukin-22 (IL-22) after intestinal injury, increase the growth of mouse small intestine organoids in an IL-22-dependent fashion. Recombinant IL-22 directly targeted ISCs, augmenting the growth of both mouse and human intestinal organoids, increasing proliferation and promoting ISC expansion. IL-22 induced STAT3 phosphorylation in Lgr5+ ISCs, and STAT3 was crucial for both organoid formation and IL-22-mediated regeneration. Treatment with IL-22 in vivo after mouse allogeneic bone marrow transplantation enhanced the recovery of ISCs, increased epithelial regeneration and reduced intestinal pathology and mortality from graft-versus-host disease. ATOH1-deficient organoid culture demonstrated that IL-22 induced epithelial regeneration independently of the Paneth cell niche. Our findings reveal a fundamental mechanism by which the immune system is able to support the intestinal epithelium, activating ISCs to promote regeneration.


Science | 2012

Interleukin-22 Drives Endogenous Thymic Regeneration in Mice

Jarrod A. Dudakov; Alan M. Hanash; Robert R. Jenq; Lauren F. Young; Arnab Ghosh; Natalie V. Singer; Mallory L. West; Odette M. Smith; Amanda M. Holland; Jennifer J. Tsai; Richard L. Boyd; Marcel R.M. van den Brink

IL-22 Protects the Thymus One of the side effects associated with radiation treatment and some types of chemotherapy is damage to the thymus. Immunological T cells develop in the thymus, and so damage to this organ results in immunodeficiency and increased susceptibility to infectious disease. Although the organ eventually recovers, therapies that speed this recovery process are of interest. Dudakov et al. (p. 91, published online 1 March; see the Perspective by Bhandoola and Artis) now show in mice that interleukin-22 (IL-22) production in the thymus is increased in response to radiation damage and that this cytokine promotes thymic repair. After radiation treatment, IL-23 production by thymic dendritic cells induced IL-22 secretion by a population of radio-resistant innate lymphoid cells. IL-22 appeared to mediate its effects by promoting the survival and proliferation of thymic epithelial cells. Damage to the thymus caused by infection or radiation is reversed by a cytokine. Endogenous thymic regeneration is a crucial function that allows for renewal of immune competence after stress, infection, or immunodepletion. However, the mechanisms governing this regeneration remain poorly understood. We detail such a mechanism, centered on interleukin-22 (IL-22) and triggered by the depletion of CD4+CD8+ double-positive thymocytes. Intrathymic levels of IL-22 were increased after thymic insult, and thymic recovery was impaired in IL-22–deficient mice. IL-22, which signaled through thymic epithelial cells and promoted their proliferation and survival, was up-regulated by radio-resistant RORγ(t)+CCR6+NKp46– lymphoid tissue inducer cells after thymic injury in an IL-23–dependent manner. Administration of IL-22 enhanced thymic recovery after total body irradiation. These studies reveal mechanisms of endogenous thymic repair and offer innovative regenerative strategies for improving immune competence.


Nature Cell Biology | 2013

Nrf2 regulates haematopoietic stem cell function

Jennifer J. Tsai; Jarrod A. Dudakov; Koichi Takahashi; Jae Hung Shieh; Enrico Velardi; Amanda M. Holland; Natalie V. Singer; Mallory L. West; Odette M. Smith; Lauren F. Young; Yusuke Shono; Arnab Ghosh; Alan M. Hanash; Hien Tran; Malcolm A. S. Moore; Marcel R.M. van den Brink

Coordinating the balance between haematopoietic stem cell (HSC) quiescence and self-renewal is crucial for maintaining haematopoiesis lifelong. Equally important for haematopoietic function is modulating HSC localization within the bone marrow niches, as maintenance of HSC function is tightly controlled by a complex network of intrinsic molecular mechanisms and extrinsic signalling interactions with their surrounding microenvironment. In this study we demonstrate that nuclear factor erythroid 2-related factor 2 (Nfe2l2, or Nrf2), well established as a global regulator of the oxidative stress response, plays a regulatory role in several aspects of HSC homeostasis. Nrf2 deficiency results in an expansion of the haematopoietic stem and progenitor cell compartment due to cell-intrinsic hyperproliferation, which was accomplished at the expense of HSC quiescence and self-renewal. We further show that Nrf2 modulates both migration and retention of HSCs in their niche. Moreover, we identify a previously unrecognized link between Nrf2 and CXCR4, contributing, at least partially, to the maintenance of HSC function.


Science Translational Medicine | 2016

Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice

Yusuke Shono; Melissa D. Docampo; Jonathan U. Peled; Suelen M. Perobelli; Enrico Velardi; Jennifer J. Tsai; Ann E. Slingerland; Odette M. Smith; Lauren F. Young; Jyotsna Gupta; Sophia R. Lieberman; Hillary Jay; Katya F. Ahr; Kori A. Porosnicu Rodriguez; Ke Xu; Marco Calarfiore; Hendrik Poeck; Silvia Caballero; Sean M. Devlin; Franck Rapaport; Jarrod A. Dudakov; Alan M. Hanash; Boglarka Gyurkocza; George F. Murphy; Camilla Borges Ferreira Gomes; Chen Liu; Eli L. Moss; Shannon B. Falconer; Ami S. Bhatt; Ying Taur

Treating neutropenic fever with broad-spectrum antibiotics after allogeneic hematopoietic stem cell transplant is associated with an increase in graft-versus-host disease in mice and humans. Antibiotics for allogeneic transplant—A double-edged sword Patients undergoing allogeneic hematopoietic stem cell transplantation often receive antibiotics for infections, which can also unfortunately kill intestinal bacteria. These symbiotic bacteria in the gut generally do not cause disease and are thought to suppress inflammation. In a new study, Shono et al. examined the records of 857 transplant patients and found that certain antibiotics were linked with development of graft-versus-host disease (GVHD), which can cause severe intestinal inflammation. Using a mouse model, the authors showed that these antibiotics may select for bacteria that consume intestinal mucus and lead to loss of this important layer of protection for the gut, thus exacerbating GVHD in the intestine. This study suggests that not all antibiotic regimens are appropriate for treating transplant patients. Intestinal bacteria may modulate the risk of infection and graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Allo-HSCT recipients often develop neutropenic fever, which is treated with antibiotics that may target anaerobic bacteria in the gut. We retrospectively examined 857 allo-HSCT recipients and found that treatment of neutropenic fever with imipenem-cilastatin and piperacillin-tazobactam antibiotics was associated with increased GVHD-related mortality at 5 years (21.5% for imipenem-cilastatin–treated patients versus 13.1% for untreated patients, P = 0.025; 19.8% for piperacillin-tazobactam–treated patients versus 11.9% for untreated patients, P = 0.007). However, two other antibiotics also used to treat neutropenic fever, aztreonam and cefepime, were not associated with GVHD-related mortality (P = 0.78 and P = 0.98, respectively). Analysis of stool specimens from allo-HSCT recipients showed that piperacillin-tazobactam administration was associated with perturbation of gut microbial composition. Studies in mice demonstrated aggravated GVHD mortality with imipenem-cilastatin or piperacillin-tazobactam compared to aztreonam (P < 0.01 and P < 0.05, respectively). We found pathological evidence for increased GVHD in the colon of imipenem-cilastatin–treated mice (P < 0.05), but no difference in the concentration of short-chain fatty acids or numbers of regulatory T cells. Notably, imipenem-cilastatin treatment of mice with GVHD led to loss of the protective mucus lining of the colon (P < 0.01) and the compromising of intestinal barrier function (P < 0.05). Sequencing of mouse stool specimens showed an increase in Akkermansia muciniphila (P < 0.001), a commensal bacterium with mucus-degrading capabilities, raising the possibility that mucus degradation may contribute to murine GVHD. We demonstrate an underappreciated risk for the treatment of allo-HSCT recipients with antibiotics that may exacerbate GVHD in the colon.


Nature Medicine | 2017

Donor CD19 CAR T cells exert potent graft-versus-lymphoma activity with diminished graft-versus-host activity

Arnab Ghosh; Melody Smith; Scott James; Marco L. Davila; Enrico Velardi; Kimon V. Argyropoulos; Gertrude Gunset; Fabiana Perna; Fabiana M Kreines; Emily R Levy; Sophie Lieberman; Hillary Jay; Andrea Z. Tuckett; Johannes L. Zakrzewski; Lisa Tan; Lauren F. Young; Kate Takvorian; Jarrod A. Dudakov; Robert R. Jenq; Alan M. Hanash; Ana Carolina Fragoso Motta; George F. Murphy; Chen Liu; Andrea Schietinger; Michel Sadelain; Marcel R.M. van den Brink

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for hematological malignancies. However, graft-versus-host disease (GVHD) and relapse after allo-HSCT remain major impediments to the success of allo-HSCT. Chimeric antigen receptors (CARs) direct tumor cell recognition of adoptively transferred T cells. CD19 is an attractive CAR target, which is expressed in most B cell malignancies, as well as in healthy B cells. Clinical trials using autologous CD19-targeted T cells have shown remarkable promise in various B cell malignancies. However, the use of allogeneic CAR T cells poses a concern in that it may increase risk of the occurrence of GVHD, although this has not been reported in selected patients infused with donor-derived CD19 CAR T cells after allo-HSCT. To understand the mechanism whereby allogeneic CD19 CAR T cells may mediate anti-lymphoma activity without causing a significant increase in the incidence of GVHD, we studied donor-derived CD19 CAR T cells in allo-HSCT and lymphoma models in mice. We demonstrate that alloreactive T cells expressing CD28-costimulated CD19 CARs experience enhanced stimulation, resulting in the progressive loss of both their effector function and proliferative potential, clonal deletion, and significantly decreased occurrence of GVHD. Concurrently, the other CAR T cells that were present in bulk donor T cell populations retained their anti-lymphoma activity in accordance with the requirement that both the T cell receptor (TCR) and CAR be engaged to accelerate T cell exhaustion. In contrast, first-generation and 4-1BB-costimulated CAR T cells increased the occurrence of GVHD. These findings could explain the reduced risk of GVHD occurring with cumulative TCR and CAR signaling.


Journal of Experimental Medicine | 2014

Sex steroid blockade enhances thymopoiesis by modulating Notch signaling

Enrico Velardi; Jennifer J. Tsai; Amanda M. Holland; Tobias Wertheimer; Vionnie W.C. Yu; Johannes L. Zakrzewski; Andrea Z. Tuckett; Natalie V. Singer; Mallory L. West; Odette M. Smith; Lauren F. Young; Fabiana M Kreines; Emily R Levy; Richard L. Boyd; David T. Scadden; Jarrod A. Dudakov; Marcel R.M. van den Brink

Velardi et al. show that sex steroids regulate thymopoiesis by directly modulating Notch signaling, and provide a novel clinical strategy to boost immune regeneration.


Journal of Clinical Investigation | 2013

Adoptively transferred TRAIL + T cells suppress GVHD and augment antitumor activity

Arnab Ghosh; Yildirim Dogan; Maxim Moroz; Amanda M. Holland; Nury Yim; Uttam K. Rao; Lauren F. Young; Daniel Tannenbaum; Durva Masih; Enrico Velardi; Jennifer J. Tsai; Robert R. Jenq; Olaf Penack; Alan M. Hanash; Odette M. Smith; Kelly Piersanti; Cecilia Lezcano; George F. Murphy; Chen Liu; M. Lia Palomba; Martin G. Sauer; Michel Sadelain; Vladimir Ponomarev; Marcel R.M. van den Brink

Current strategies to suppress graft-versus-host disease (GVHD) also compromise graft-versus-tumor (GVT) responses. Furthermore, most experimental strategies to separate GVHD and GVT responses merely spare GVT function without actually enhancing it. We have previously shown that endogenously expressed TNF-related apoptosis-inducing ligand (TRAIL) is required for optimal GVT activity against certain malignancies in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT). In order to model a donor-derived cellular therapy, we genetically engineered T cells to overexpress TRAIL and adoptively transferred donor-type unsorted TRAIL+ T cells into mouse models of allo-HSCT. We found that murine TRAIL+ T cells induced apoptosis of alloreactive T cells, thereby reducing GVHD in a DR5-dependent manner. Furthermore, murine TRAIL+ T cells mediated enhanced in vitro and in vivo antilymphoma GVT response. Moreover, human TRAIL+ T cells mediated enhanced in vitro cytotoxicity against both human leukemia cell lines and against freshly isolated chronic lymphocytic leukemia (CLL) cells. Finally, as a model of off-the-shelf, donor-unrestricted antitumor cellular therapy, in vitro-generated TRAIL+ precursor T cells from third-party donors also mediated enhanced GVT response in the absence of GVHD. These data indicate that TRAIL-overexpressing donor T cells could potentially enhance the curative potential of allo-HSCT by increasing GVT response and suppressing GVHD.


Journal of Immunology | 2013

Age-Associated Changes in the Differentiation Potentials of Human Circulating Hematopoietic Progenitors to T- or NK-Lineage Cells

Seishi Kyoizumi; Yoshiko Kubo; Junko Kajimura; Kengo Yoshida; Kazue Imai; Tomonori Hayashi; Kei Nakachi; Lauren F. Young; Malcolm A. S. Moore; Marcel R.M. van den Brink; Yoichiro Kusunoki

Age-associated changes of T and NK cell (T/NK) potential of human hematopoietic stem cells are unknown. In this study, we enumerate and characterize T/NK precursors among CD34+Lin− cell populations circulating in normal human adult peripheral blood (PB) by a limiting-dilution assay using coculture with OP9-DL1 stroma cells expressing Notch 1 ligand, Delta–like 1. The frequency of T cell precursors in CD34+Lin− cells was found to decrease with donor age, whereas the ratio of NK to T cell precursor frequency (NK/T ratio) increased with age, suggesting that lymphoid differentiation potential of PB progenitors shifts from T to NK cell lineage with aging. Clonal analyses of CD34+Lin− cells showed that differences in the NK/T ratio were attributable to different distributions of single- and dual-lineage T/NK precursor clones. Because nearly all of the clones retained monocyte and/or granulocyte differentiation potentials in coculture with OP9-DL1 cells, T/NK precursors in PB are considered to be contained in the pool of T/NK/myeloid multipotent progenitors. The age-associated increase in NK over T cell commitment might occur in precursor cells with T/NK/myeloid potential.

Collaboration


Dive into the Lauren F. Young's collaboration.

Top Co-Authors

Avatar

Marcel R.M. van den Brink

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Alan M. Hanash

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Odette M. Smith

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jarrod A. Dudakov

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Robert R. Jenq

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Enrico Velardi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jennifer J. Tsai

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amanda M. Holland

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mallory L. West

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Natalie V. Singer

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge