Network


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

Hotspot


Dive into the research topics where Sandra White is active.

Publication


Featured researches published by Sandra White.


Journal of Immunology | 2010

Cutting Edge: Caspase-1 Independent IL-1β Production Is Critical for Host Resistance to Mycobacterium tuberculosis and Does Not Require TLR Signaling In Vivo

Katrin D. Mayer-Barber; Daniel L. Barber; Kevin Shenderov; Sandra White; Mark S. Wilson; Allen W. Cheever; David G. Kugler; Sara Hieny; Patricia Caspar; Gabriel Núñez; Dirk Schlueter; Richard A. Flavell; Fayyaz S. Sutterwala; Alan Sher

To investigate the respective contributions of TLR versus IL-1R mediated signals in MyD88 dependent control of Mycobacterium tuberculosis, we compared the outcome of M. tuberculosis infection in MyD88, TRIF/MyD88, IL-1R1, and IL-1β–deficient mice. All four strains displayed acute mortality with highly increased pulmonary bacterial burden suggesting a major role for IL-1β signaling in determining the MyD88 dependent phenotype. Unexpectedly, the infected MyD88 and TRIF/MyD88-deficient mice, rather than being defective in IL-1β expression, displayed increased cytokine levels relative to wild-type animals. Similarly, infected mice deficient in caspase-1 and ASC, which have critical functions in inflammasome-mediated IL-1β maturation, showed unimpaired IL-1β production and importantly, were considerably less susceptible to infection than IL-1β deficient mice. Together our findings reveal a major role for IL-1β in host resistance to M. tuberculosis and indicate that during this infection the cytokine can be generated by a mechanism that does not require TLR signaling or caspase-1.


Science Translational Medicine | 2015

TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma

David F. Choy; Kevin M. Hart; Lee A. Borthwick; Aarti Shikotra; Deepti R. Nagarkar; Salman Siddiqui; Guiquan Jia; Chandra M. Ohri; Emma Doran; Kevin M. Vannella; Claire A. Butler; Beverley Hargadon; Joshua Sciurba; Richard L. Gieseck; Robert W. Thompson; Sandra White; Alexander R. Abbas; Janet Jackman; Lawren C. Wu; Jackson G. Egen; Liam Heaney; Thirumalai R. Ramalingam; Joseph R. Arron; Thomas A. Wynn; Peter Bradding

Concurrent blockade of IL-13 and IL-17A may improve control of asthma. A tale of two asthmas Classifying diseases according to symptoms is rapidly becoming a thing of the past. Targeted therapeutics have shown us that sets of symptoms can be caused by different pathogenic mechanisms. Now, Choy et al. demonstrate that asthma can be divided into three immunological clusters: TH2-high, TH17-high, and TH2/17-low. The TH2-high and TH17-high clusters were inversely correlated in patients. Moreover, neutralizing one signature promoted the other in a mouse model of asthma. These data suggest that combination therapies targeting both pathways may better treat asthmatic individuals. Increasing evidence suggests that asthma is a heterogeneous disorder regulated by distinct molecular mechanisms. In a cross-sectional study of asthmatics of varying severity (n = 51), endobronchial tissue gene expression analysis revealed three major patient clusters: TH2-high, TH17-high, and TH2/17-low. TH2-high and TH17-high patterns were mutually exclusive in individual patient samples, and their gene signatures were inversely correlated and differentially regulated by interleukin-13 (IL-13) and IL-17A. To understand this dichotomous pattern of T helper 2 (TH2) and TH17 signatures, we investigated the potential of type 2 cytokine suppression in promoting TH17 responses in a preclinical model of allergen-induced asthma. Neutralization of IL-4 and/or IL-13 resulted in increased TH17 cells and neutrophilic inflammation in the lung. However, neutralization of IL-13 and IL-17 protected mice from eosinophilia, mucus hyperplasia, and airway hyperreactivity and abolished the neutrophilic inflammation, suggesting that combination therapies targeting both pathways may maximize therapeutic efficacy across a patient population comprising both TH2 and TH17 endotypes.


PLOS Pathogens | 2013

miR-182 and miR-10a are key regulators of Treg specialisation and stability during Schistosome and Leishmania-associated inflammation.

Samir N. Kelada; Praveen Sethupathy; Isobel S. Okoye; Eleni Kistasis; Stephanie Czieso; Sandra White; David Chou; Craig Martens; Stacy Ricklefs; Kimmo Virtaneva; Dan Sturdevant; Stephen F. Porcella; Yasmine Belkaid; Thomas A. Wynn; Mark S. Wilson

A diverse suite of effector immune responses provide protection against various pathogens. However, the array of effector responses must be immunologically regulated to limit pathogen- and immune-associated damage. CD4+Foxp3+ regulatory T cells (Treg) calibrate immune responses; however, how Treg cells adapt to control different effector responses is unclear. To investigate the molecular mechanism of Treg diversity we used whole genome expression profiling and next generation small RNA sequencing of Treg cells isolated from type-1 or type-2 inflamed tissue following Leishmania major or Schistosoma mansoni infection, respectively. In-silico analyses identified two miRNA “regulatory hubs” miR-10a and miR-182 as critical miRNAs in Th1- or Th2-associated Treg cells, respectively. Functionally and mechanistically, in-vitro and in-vivo systems identified that an IL-12/IFNγ axis regulated miR-10a and its putative transcription factor, Creb. Importantly, reduced miR-10a in Th1-associated Treg cells was critical for Treg function and controlled a suite of genes preventing IFNγ production. In contrast, IL-4 regulated miR-182 and cMaf in Th2-associed Treg cells, which mitigated IL-2 secretion, in part through repression of IL2-promoting genes. Together, this study indicates that CD4+Foxp3+ cells can be shaped by local environmental factors, which orchestrate distinct miRNA pathways preserving Treg stability and suppressor function.


PLOS Pathogens | 2014

Incomplete Deletion of IL-4Rα by LysMCre Reveals Distinct Subsets of M2 Macrophages Controlling Inflammation and Fibrosis in Chronic Schistosomiasis

Kevin M. Vannella; Luke Barron; Lee A. Borthwick; Kristen N. Kindrachuk; Prakash Babu Narasimhan; Kevin M. Hart; Robert W. Thompson; Sandra White; Allen W. Cheever; Thirumalai R. Ramalingam; Thomas A. Wynn

Mice expressing a Cre recombinase from the lysozyme M-encoding locus (Lyz2) have been widely used to dissect gene function in macrophages and neutrophils. Here, we show that while naïve resident tissue macrophages from IL-4Rαflox/deltaLysMCre mice almost completely lose IL-4Rα function, a large fraction of macrophages elicited by sterile inflammatory stimuli, Schistosoma mansoni eggs, or S. mansoni infection, fail to excise Il4rα. These F4/80hiCD11bhi macrophages, in contrast to resident tissue macrophages, express lower levels of Lyz2 explaining why this population resists LysMCre-mediated deletion. We show that in response to IL-4 and IL-13, Lyz2loIL-4Rα+ macrophages differentiate into an arginase 1-expressing alternatively-activated macrophage (AAM) population, which slows the development of lethal fibrosis in schistosomiasis. In contrast, we identified Lyz2hiIL-4Rα+ macrophages as the key subset of AAMs mediating the downmodulation of granulomatous inflammation in chronic schistosomiasis. Our observations reveal a limitation on using a LysMCre mouse model to study gene function in inflammatory settings, but we utilize this limitation as a means to demonstrate that distinct populations of alternatively activated macrophages control inflammation and fibrosis in chronic schistosomiasis.


Blood | 2010

Th1-driven immune reconstitution disease in Mycobacterium avium-infected mice.

Daniel L. Barber; Katrin D. Mayer-Barber; Lis Ribeiro do Valle Antonelli; Mark S. Wilson; Sandra White; Patricia Caspar; Sara Hieny; Irini Sereti; Alan Sher

Following antiretroviral therapy, a significant proportion of HIV(+) patients with mycobacterial coinfections develop a paradoxical, poorly understood inflammatory disease termed immune reconstitution inflammatory syndrome (IRIS). Here, we show that Mycobacterium avium-infected T cell-deficient mice injected with CD4 T cells also develop an immune reconstitution disease (IRD) manifesting as weight loss, impaired lung function, and rapid mortality. This form of IRD requires Ag recognition and interferonγ production by the donor CD4 T cells and correlates with marked alterations in blood and tissue CD11b(+) myeloid cells. Interestingly, disease is associated with impaired, rather than augmented, T-cell expansion and function and is not strictly dependent on lymphopenia-induced T-cell proliferation. Instead, our findings suggest that mycobacterial-associated IRIS results from a heightened sensitivity of infected lymphopenic hosts to the detrimental effects of Ag-driven CD4 T-cell responses.


PLOS Pathogens | 2011

IL-10 blocks the development of resistance to re-infection with Schistosoma mansoni.

Mark S. Wilson; Allen W. Cheever; Sandra White; Robert W. Thompson; Thomas A. Wynn

Despite effective chemotherapy to treat schistosome infections, re-infection rates are extremely high. Resistance to reinfection can develop, however it typically takes several years following numerous rounds of treatment and re-infection, and often develops in only a small cohort of individuals. Using a well-established and highly permissive mouse model, we investigated whether immunoregulatory mechanisms influence the development of resistance. Following Praziquantel (PZQ) treatment of S. mansoni infected mice we observed a significant and mixed anti-worm response, characterized by Th1, Th2 and Th17 responses. Despite the elevated anti-worm response in PBMCs, liver, spleen and mesenteric lymph nodes, this did not confer any protection from a secondary challenge infection. Because a significant increase in IL-10-producing CD4+CD44+CD25+GITR+ lymphocytes was observed, we hypothesised that IL-10 was obstructing the development of resistance. Blockade of IL-10 combined with PZQ treatment afforded a greater than 50% reduction in parasite establishment during reinfection, compared to PZQ treatment alone, indicating that IL-10 obstructs the development of acquired resistance. Markedly enhanced Th1, Th2 and Th17 responses, worm-specific IgG1, IgG2b and IgE and circulating eosinophils characterized the protection. This study demonstrates that blocking IL-10 signalling during PZQ treatment can facilitate the development of protective immunity and provide a highly effective strategy to protect against reinfection with S. mansoni.


Science Translational Medicine | 2016

Combinatorial targeting of TSLP, IL-25, and IL-33 in type 2 cytokine-driven inflammation and fibrosis

Kevin M. Vannella; Thirumalai R. Ramalingam; Lee A. Borthwick; Luke Barron; Kevin M. Hart; Robert W. Thompson; Kristen N. Kindrachuk; Allen W. Cheever; Sandra White; Alison L. Budelsky; Michael R. Comeau; Dirk E. Smith; Thomas A. Wynn

Combined blockade of TSLP, IL-25, and IL-33 may be needed to treat some forms of progressive inflammation and fibrosis. Teaming up against inflammation and fibrosis The cult of the individual extends to new therapies—new targets are identified and validated (or not) on a one-on-one basis. However, no protein is an island, and failure with a monotherapy does not invalidate a target. Now, Vannella et al. demonstrate that this is indeed the case for some types of progessive type 2 inflammation and fibrosis. Using a variety of models including helminth infection and allergic lung inflammation, the authors show that individual disruption of the type 2 inflammatory molecules thymic stromal lymphopoietin (TLSP), interleukin-25 (IL-25), and IL-33 had no effect on the progression of type 2–dependent inflammation or fibrosis. However, targeting all three simultaneously blocked disease development and progression. Thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and IL-33 are important initiators of type 2–associated mucosal inflammation and immunity. However, their role in the maintenance of progressive type 2 inflammation and fibrosis is much less clear. Using chronic models of helminth infection and allergic lung inflammation, we show that collective disruption of TSLP, IL-25, and IL-33 signaling suppresses chronic and progressive type 2 cytokine–driven inflammation and fibrosis. In a schistosome lung granuloma model or during chronic Schistosoma mansoni infection in the liver, individual ablation of TSLP, IL-25, or IL-33/ST2 had no impact on the development of IL-4/IL-13–dependent inflammation or fibrosis. However, significant reductions in granuloma-associated eosinophils, hepatic fibrosis, and IL-13–producing type 2 innate lymphoid cells (ILC2s) were observed when signaling of all three mediators was simultaneously disrupted. Combined blockade through monoclonal antibody (mAb) treatment also reduced IL-5 and IL-13 expression during primary and secondary granuloma formation in the lungs. In a model of chronic house dust mite–induced allergic lung inflammation, combined mAb treatment did not decrease established inflammation or fibrosis. TSLP/IL-33 double-knockout mice treated with anti–IL-25 mAb during priming, however, displayed decreased inflammation, mucus production, and lung remodeling in the chronic phase. Together, these studies reveal partially redundant roles for TSLP, IL-25, and IL-33 in the maintenance of type 2 pathology and suggest that in some settings, early combined targeting of these mediators is necessary to ameliorate progressive type 2–driven disease.


Contemporary topics in immunobiology | 1980

Immunologic Reactivity of Lymphoid Cells in Tumors

Ronald B. Herberman; Howard T. Holden; Luigi Varesio; Tadayoshi Taniyama; Paolo Puccetti; Holger Kirchner; James M. Gerson; Sandra White; Yona Keisari; J. Stephen Haskill

There have been many studies of immune responses against tumors and almost all of these have focused on the reactivity in the blood or spleen. From such studies, it has become clear that a wide variety of effector cells and types of immune functions may be involved in antitumor responses. Particular attention has been directed toward T cells that may be directly cytotoxic against tumor cells or may proliferate or produce lymphokines upon stimulation with tumor antigens. However, other effector mechanisms may be involved and need to be considered. These include B cells, which can produce antibodies that affect tumor cells directly or that interact with K cells or macrophages and thereby mediate antibody-dependent cell-mediated cytotoxicity; macrophages and monocytes, which are spontaneously cytotoxic or can be activated to become cytotoxic against tumor cells; and natural killer (NK) cells, a subpopulation of lymphocytes with spontaneous cytotoxic reactivity against tumor cells.


Nature Immunology | 2016

Acidic chitinase primes the protective immune response to gastrointestinal nematodes

Kevin M. Vannella; Thirumalai R. Ramalingam; Kevin M. Hart; Rafael de Queiroz Prado; Joshua Sciurba; Luke Barron; Lee A. Borthwick; Allen Smith; Margaret M. Mentink-Kane; Sandra White; Robert W. Thompson; Allen W. Cheever; Kevin W. Bock; Ian N. Moore; Lori Fitz; Joseph F. Urban; Thomas A. Wynn

Acidic mammalian chitinase (AMCase) is known to be induced by allergens and helminths, yet its role in immunity is unclear. Using AMCase-deficient mice, we show that AMCase deficiency reduced the number of group 2 innate lymphoid cells during allergen challenge but was not required for establishment of type 2 inflammation in the lung in response to allergens or helminths. In contrast, AMCase-deficient mice showed a profound defect in type 2 immunity following infection with the chitin-containing gastrointestinal nematodes Nippostrongylus brasiliensis and Heligmosomoides polygyrus bakeri. The impaired immunity was associated with reduced mucus production and decreased intestinal expression of the signature type 2 response genes Il13, Chil3, Retnlb, and Clca1. CD103+ dendritic cells, which regulate T cell homing, were also reduced in mesenteric lymph nodes of infected AMCase-deficient mice. Thus, AMCase functions as a critical initiator of protective type 2 responses to intestinal nematodes but is largely dispensable for allergic responses in the lung.


Science Translational Medicine | 2017

Type 2 immunity is protective in metabolic disease but exacerbates NAFLD collaboratively with TGF-β

Kevin M. Hart; Thomas Fabre; Joshua Sciurba; Richard L. Gieseck; Lee A. Borthwick; Kevin M. Vannella; Thomas H. Acciani; Rafael de Queiroz Prado; Robert W. Thompson; Sandra White; Geneviève Soucy; Marc Bilodeau; Thirumalai R. Ramalingam; Joseph R. Arron; Naglaa H. Shoukry; Thomas A. Wynn

Development of NASH fibrosis is accompanied by accumulation of hepatic eosinophils and driven by TGF-β and profibrotic type 2 inflammation. Opposing cytokines in obesity and fibrosis Polarized cytokine networks can drive immunopathogenesis forward. Obesity that leads to liver fibrosis involves type 1 cytokines, so Hart et al. expected obese mice prone to type 1 cytokine responses to experience more pronounced fibrosis. Instead, they saw that these mice were resistant to steatohepatitis. Fibrotic livers from mice and human biopsies showed type 2 inflammation and recruitment of eosinophils, unlike the inflammation observed in the adipose tissue during obesity. These findings reveal that cytokine activity that is beneficial for the homeostasis of one tissue can be detrimental to another. Nonalcoholic fatty liver disease (NAFLD) is now the most common progressive liver disease in developed countries and is the second leading indication for liver transplantation due to the extensive fibrosis it causes. NAFLD progression is thought to be tied to chronic low-level type 1 inflammation originating in the adipose tissue during obesity; however, the specific immunological mechanisms regulating the progression of NAFLD-associated fibrosis in the liver are unclear. To investigate the immunopathogenesis of NAFLD more completely, we investigated adipose dysfunction, nonalcoholic steatohepatitis (NASH), and fibrosis in mice that develop polarized type 1 or type 2 immune responses. Unexpectedly, obese interleukin-10 (IL-10)/IL-4–deficient mice (type 1–polarized) were highly resistant to NASH. This protection was associated with an increased hepatic interferon-γ (IFN-γ) signature. Conversely, IFN-γ–deficient mice progressed rapidly to NASH with evidence of fibrosis dependent on transforming growth factor–β (TGF-β) and IL-13 signaling. Unlike increasing type 1 inflammation and the marked loss of eosinophils seen in expanding adipose tissue, progression of NASH was associated with increasing eosinophilic type 2 liver inflammation in mice and human patient biopsies. Finally, simultaneous inhibition of TGF-β and IL-13 signaling attenuated the fibrotic machinery more completely than TGF-β alone in NAFLD-associated fibrosis. Thus, although type 2 immunity maintains healthy metabolic signaling in adipose tissues, it exacerbates the progression of NAFLD collaboratively with TGF-β in the liver.

Collaboration


Dive into the Sandra White's collaboration.

Top Co-Authors

Avatar

Thomas A. Wynn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert W. Thompson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kevin M. Hart

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kevin M. Vannella

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan Sher

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel L. Barber

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joshua Sciurba

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge