Network


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

Hotspot


Dive into the research topics where Carey Ann Gallini is active.

Publication


Featured researches published by Carey Ann Gallini.


Science | 2013

The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis

Patrick M. Smith; Michael R. Howitt; Nicolai S. Panikov; Monia Michaud; Carey Ann Gallini; Mohammad Bohlooly-Y; Jonathan N. Glickman; Wendy S. Garrett

Protecting the Guts Regulatory T cells (Tregs) in the gut are important sentinels in maintaining the peace between our gut and its trillions of resident bacteria and have been shown to be regulated by specific strains of bacteria in mouse models. Smith et al. (p. 569, published online 4 July; see the Perspective by Bollrath and Powrie) asked whether metabolite(s) generated by resident bacterial species may regulate Tregs in the gut. Indeed, short-chain fatty acids (SCFAs), bacterial fermentation products of dietary fibers produced by a range of bacteria, restored colonic Treg numbers in mice devoid of a gut microbiota and increased Treg numbers in colonized mice. The effects of SCFAs on Tregs were mediated through GPCR43, a receptor for SCFAs, which is expressed on colonic Tregs. Mice fed SCFAs were protected against experimentally induced colitis in a manner that was dependent on GPR43-expressing Tregs. Bacterial fermentation products regulate the number and function of regulatory T cells in the mouse colon. [Also see Perspective by Bollrath and Powrie] Regulatory T cells (Tregs) that express the transcription factor Foxp3 are critical for regulating intestinal inflammation. Candidate microbe approaches have identified bacterial species and strain-specific molecules that can affect intestinal immune responses, including species that modulate Treg responses. Because neither all humans nor mice harbor the same bacterial strains, we posited that more prevalent factors exist that regulate the number and function of colonic Tregs. We determined that short-chain fatty acids, gut microbiota–derived bacterial fermentation products, regulate the size and function of the colonic Treg pool and protect against colitis in a Ffar2-dependent manner in mice. Our study reveals that a class of abundant microbial metabolites underlies adaptive immune microbiota coadaptation and promotes colonic homeostasis and health.


Cell Host & Microbe | 2010

Enterobacteriaceae Act in Concert with the Gut Microbiota to Induce Spontaneous and Maternally Transmitted Colitis

Wendy S. Garrett; Carey Ann Gallini; Tanya Yatsunenko; Monia Michaud; Andrea M. DuBois; Mary L. Delaney; Shivesh Punit; Maria Karlsson; Lynn Bry; Jonathan N. Glickman; Jeffrey I. Gordon; Andrew B. Onderdonk; Laurie H. Glimcher

Disruption of homeostasis between the host immune system and the intestinal microbiota leads to inflammatory bowel disease (IBD). Whether IBD is instigated by individual species or disruptions of entire microbial communities remains controversial. We characterized the fecal microbial communities in the recently described T-bet(-/-) ×Rag2(-/-) ulcerative colitis (TRUC) model driven by T-bet deficiency in the innate immune system. 16S rRNA-based analysis of TRUC and Rag2(-/-) mice revealed distinctive communities that correlate with host genotype. The presence of Klebsiella pneumoniae and Proteus mirabilis correlates with colitis in TRUC animals, and these TRUC-derived strains can elicit colitis in Rag2(-/-) and WT adults but require a maternally transmitted endogenous microbial community for maximal intestinal inflammation. Cross-fostering experiments indicated a role for these organisms in maternal transmission of disease. Our findings illustrate how gut microbial communities work in concert with specific culturable colitogenic agents to cause IBD.


Science | 2016

Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut

Michael R. Howitt; Sydney Lavoie; Monia Michaud; Arthur M. Blum; Sara V. Tran; Joel V. Weinstock; Carey Ann Gallini; Kevin Redding; Robert F. Margolskee; Lisa C. Osborne; David Artis; Wendy S. Garrett

Tuft cells help contain parasites Trillions of microbes inhabit our guts, including worms and other parasites. Epithelial cells that line the gut orchestrate parasite-targeted immune responses. Howitt et al. now identify a key cellular player in immunity to parasites: tuft cells (see the Perspective by Harris). Tuft cells make up a small fraction of gut epithelial cells but expand when parasites colonize or infect the gut. Parasites cause tuft cells to secrete large amounts of interleukin-25, a key cytokine for parasite clearance that also indirectly feeds back on tuft cells to expand their numbers. Tuft cells express chemosensory signaling machinery: disrupting this blocked parasite-triggered tuft cell expansion and weakened the ability of mice to control a parasitic infection. Science, this issue p. 1329; see also p. 1264 Gut epithelial tuft cells are key players in mucosal immune responses against parasites. [Also see Perspective by Harris] The intestinal epithelium forms an essential barrier between a host and its microbiota. Protozoa and helminths are members of the gut microbiota of mammals, including humans, yet the many ways that gut epithelial cells orchestrate responses to these eukaryotes remain unclear. Here we show that tuft cells, which are taste-chemosensory epithelial cells, accumulate during parasite colonization and infection. Disruption of chemosensory signaling through the loss of TRMP5 abrogates the expansion of tuft cells, goblet cells, eosinophils, and type 2 innate lymphoid cells during parasite colonization. Tuft cells are the primary source of the parasite-induced cytokine interleukin-25, which indirectly induces tuft cell expansion by promoting interleukin-13 production by innate lymphoid cells. Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Bifidobacterium animalis subsp. lactis fermented milk product reduces inflammation by altering a niche for colitogenic microbes

Patrick Veiga; Carey Ann Gallini; Chloé Beal; Monia Michaud; Mary L. Delaney; Andrea M. DuBois; Artem Khlebnikov; Johan van Hylckama Vlieg; Shivesh Punit; Jonathan N. Glickman; Andrew B. Onderdonk; Laurie H. Glimcher; Wendy S. Garrett

Intestinal health requires the coexistence of eukaryotic self with the gut microbiota and dysregulated host-microbial interactions can result in intestinal inflammation. Here, we show that colitis improved in T-bet−/−Rag2−/− mice that consumed a fermented milk product containing Bifidobacterium animalis subsp. lactis DN-173 010 strain. A decrease in cecal pH and alterations in short chain fatty acid profiles occurred with consumption, and there were concomitant increases in the abundance of select lactate-consuming and butyrate-producing bacteria. These metabolic shifts created a nonpermissive environment for the Enterobacteriaceae recently identified as colitogenic in a T-bet−/−Rag2−/− ulcerative colitis mouse model. In addition, 16S rRNA-based analysis of the T-bet−/−Rag2−/−fecal microbiota suggest that the structure of the endogenous gut microbiota played a key role in shaping the host response to the bacterial strains studied herein. We have identified features of the gut microbiota, at the membership and functional level, associated with response to this B. lactis-containing fermented milk product, and therefore this model provides a framework for evaluating and optimizing probiotic-based functional foods.


The ISME Journal | 2014

Gut microbiome composition and function in experimental colitis during active disease and treatment-induced remission

Michelle G. Rooks; Patrick Veiga; Leslie Wardwell-Scott; Timothy L. Tickle; Nicola Segata; Monia Michaud; Carey Ann Gallini; Chloé Beal; Johan Et van Hylckama-Vlieg; Sonia Arora Ballal; Xochitl C. Morgan; Jonathan N. Glickman; Dirk Gevers; Curtis Huttenhower; Wendy S. Garrett

Dysregulated immune responses to gut microbes are central to inflammatory bowel disease (IBD), and gut microbial activity can fuel chronic inflammation. Examining how IBD-directed therapies influence gut microbiomes may identify microbial community features integral to mitigating disease and maintaining health. However, IBD patients often receive multiple treatments during disease flares, confounding such analyses. Preclinical models of IBD with well-defined disease courses and opportunities for controlled treatment exposures provide a valuable solution. Here, we surveyed the gut microbiome of the T-bet−/− Rag2−/− mouse model of colitis during active disease and treatment-induced remission. Microbial features modified among these conditions included altered potential for carbohydrate and energy metabolism and bacterial pathogenesis, specifically cell motility and signal transduction pathways. We also observed an increased capacity for xenobiotics metabolism, including benzoate degradation, a pathway linking host adrenergic stress with enhanced bacterial virulence, and found decreased levels of fecal dopamine in active colitis. When transferred to gnotobiotic mice, gut microbiomes from mice with active disease versus treatment-induced remission elicited varying degrees of colitis. Thus, our study provides insight into specific microbial clades and pathways associated with health, active disease and treatment interventions in a mouse model of colitis.


Cancer Cell | 2009

Colitis-associated colorectal cancer driven by T-bet deficiency in dendritic cells.

Wendy S. Garrett; Shivesh Punit; Carey Ann Gallini; Monia Michaud; Dorothy Zhang; Kirsten Sigrist; Graham M. Lord; Jonathan N. Glickman; Laurie H. Glimcher

We previously described a mouse model of ulcerative colitis linked to T-bet deficiency in the innate immune system. Here, we report that the majority of T-bet(-/-)RAG2(-/-) ulcerative colitis (TRUC) mice spontaneously progress to colonic dysplasia and rectal adenocarcinoma solely as a consequence of MyD88-independent intestinal inflammation. Dendritic cells (DCs) are necessary cellular effectors for a proinflammatory program that is carcinogenic. Whereas these malignancies arise in the setting of a complex inflammatory environment, restoration of T-bet selectively in DCs was sufficient to reduce colonic inflammation and prevent the development of neoplasia. TRUC colitis-associated colorectal cancer resembles the human disease and provides ample opportunity to probe how inflammation drives colorectal cancer development and to test preventative and therapeutic strategies preclinically.


Cell Reports | 2015

CCL2 Promotes Colorectal Carcinogenesis by Enhancing Polymorphonuclear Myeloid-Derived Suppressor Cell Population and Function.

Eunyoung Chun; Sydney Lavoie; Monia Michaud; Carey Ann Gallini; Jason Kim; Genevieve Soucy; Robert D. Odze; Jonathan N. Glickman; Wendy S. Garrett

Summary Our study reveals a non-canonical role for CCL2 in modulating non-macrophage, myeloid-derived suppressor cells (MDSCs) and shaping a tumor-permissive microenvironment during colon cancer development. We found that intratumoral CCL2 levels increased in patients with colitis-associated colorectal cancer (CRC), adenocarcinomas, and adenomas. Deletion of CCL2 blocked progression from dysplasia to adenocarcinoma and reduced the number of colonic MDSCs in a spontaneous mouse model of colitis-associated CRC. In a transplantable mouse model of adenocarcinoma and an APC-driven adenoma model, CCL2 fostered MDSC accumulation in evolving colonic tumors and enhanced polymorphonuclear (PMN)-MDSC immunosuppressive features. Mechanistically, CCL2 regulated T cell suppression of PMN-MDSCs in a STAT3-mediated manner. Furthermore, CCL2 neutralization decreased tumor numbers and MDSC accumulation and function. Collectively, our experiments support that perturbing CCL2 and targeting MDSCs may afford therapeutic opportunities for colon cancer interception and prevention.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Host lysozyme-mediated lysis of Lactococcus lactis facilitates delivery of colitis-attenuating superoxide dismutase to inflamed colons

Sonia Arora Ballal; Patrick Veiga; Kathrin Fenn; Monia Michaud; Jason Kim; Carey Ann Gallini; Jonathan N. Glickman; Gaelle Quere; Peggy Garault; Chloé Beal; Muriel Derrien; Pascal Courtin; Saulius Kulakauskas; Marie-Pierre Chapot-Chartier; Johan van Hylckama Vlieg; Wendy S. Garrett

Significance Microbes hold promise as an inflammatory bowel disease (IBD) therapy. Lactococcus lactis, which has not been appreciated as a beneficial microbe, attenuated colitis in three preclinical mouse IBD models. Neither colonization nor an intact bacterium throughout the colon per se was required. Rather, host lysozyme-mediated lysis in an inflamed colon led to L. lactis’s release of its superoxide dismutase, which was necessary for its colitis-attenuating and oxidative stress-reducing activity. Overall, these findings unveil a mechanism by which a bacterium offers benefits to the host but requires the host for targeted release of this beneficial activity. Furthermore, because L. lactis is generally regarded as safe, it represents an opportunity for rapid bench-to-bedside testing in IBD. Beneficial microbes that target molecules and pathways, such as oxidative stress, which can negatively affect both host and microbiota, may hold promise as an inflammatory bowel disease therapy. Prior work showed that a five-strain fermented milk product (FMP) improved colitis in T-bet−/− Rag2−/− mice. By varying the number of strains used in the FMP, we found that Lactococcus lactis I-1631 was sufficient to ameliorate colitis. Using comparative genomic analyses, we identified genes unique to L. lactis I-1631 involved in oxygen respiration. Respiration of oxygen results in reactive oxygen species (ROS) generation. Also, ROS are produced at high levels during intestinal inflammation and cause tissue damage. L. lactis I-1631 possesses genes encoding enzymes that detoxify ROS, such as superoxide dismutase (SodA). Thus, we hypothesized that lactococcal SodA played a role in attenuating colitis. Inactivation of the sodA gene abolished L. lactis I-1631’s beneficial effect in the T-bet−/− Rag2−/− model. Similar effects were obtained in two additional colonic inflammation models, Il10−/− mice and dextran sulfate sodium-treated mice. Efforts to understand how a lipophobic superoxide anion (O2−) can be detoxified by cytoplasmic lactoccocal SodA led to the finding that host antimicrobial-mediated lysis is a prerequisite for SodA release and SodA’s extracytoplasmic O2− scavenging. L. lactis I-1631 may represent a promising vehicle to deliver antioxidant, colitis-attenuating SodA to the inflamed intestinal mucosa, and host antimicrobials may play a critical role in mediating SodA’s bioaccessibility.


Cellular Microbiology | 2011

Host and gut microbiota symbiotic factors: lessons from inflammatory bowel disease and successful symbionts.

Sonia Arora Ballal; Carey Ann Gallini; Nicola Segata; Curtis Huttenhower; Wendy S. Garrett

Humans are colonized by a diverse collection of microbes, the largest numbers of which reside in the distal gut. The vast majority of humans coexist in a beneficial equilibrium with these microbes. However, disruption of this mutualistic relationship can manifest itself in human diseases such as inflammatory bowel disease. Thus the study of inflammatory bowel disease and its genetics can provide insight into host pathways that mediate host–microbiota symbiosis. Bacteria of the human intestinal ecosystem face numerous challenges imposed by human dietary intake, the mucosal immune system, competition from fellow members of the gut microbiota, transient ingested microbes and invading pathogens. Considering features of human resident gut bacteria provides the opportunity to understand how microbes have achieved their symbiont status. While model symbionts have provided perspective into host–microbial homeostasis, high‐throughput approaches are becoming increasingly practical for functionally characterizing the gut microbiota as a community.


PLOS ONE | 2012

Tumor Necrosis Factor α Inhibits Expression of the Iron Regulating Hormone Hepcidin in Murine Models of Innate Colitis

Nanda Kumar N. Shanmugam; Shiri Ellenbogen; Estela Trebicka; Lijian Wang; Subhankar Mukhopadhyay; Adam Lacy-Hulbert; Carey Ann Gallini; Wendy S. Garrett; Bobby J. Cherayil

Background Abnormal expression of the liver peptide hormone hepcidin, a key regulator of iron homeostasis, contributes to the pathogenesis of anemia in conditions such as inflammatory bowel disease (IBD). Since little is known about the mechanisms that control hepcidin expression during states of intestinal inflammation, we sought to shed light on this issue using mouse models. Methodology/Principal Findings Hepcidin expression was evaluated in two types of intestinal inflammation caused by innate immune activation—dextran sulfate sodium (DSS)-induced colitis in wild-type mice and the spontaneous colitis occurring in T-bet/Rag2-deficient (TRUC) mice. The role of tumor necrosis factor (TNF) α was investigated by in vivo neutralization, and by treatment of a hepatocyte cell line, as well as mice, with the recombinant cytokine. Expression and activation of Smad1, a positive regulator of hepcidin transcription, were assessed during colitis and following administration or neutralization of TNFα. Hepcidin expression progressively decreased with time during DSS colitis, correlating with changes in systemic iron distribution. TNFα inhibited hepcidin expression in cultured hepatocytes and non-colitic mice, while TNFα neutralization during DSS colitis increased it. Similar results were obtained in TRUC mice. These effects involved a TNFα-dependent decrease in Smad1 protein but not mRNA. Conclusions/Significance TNFα inhibits hepcidin expression in two distinct types of innate colitis, with down-regulation of Smad1 protein playing an important role in this process. This inhibitory effect of TNFα may be superseded by other factors in the context of T cell-mediated colitis given that in the latter form of intestinal inflammation hepcidin is usually up-regulated.

Collaboration


Dive into the Carey Ann Gallini's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea M. DuBois

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Andrew B. Onderdonk

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge