Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Infect Dis ; 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687212

ABSTRACT

Proinflammatory cytokine levels and host genetic makeup are key determinants of Clostridioides difficile infection (CDI) outcomes. We previously reported that blocking the inflammatory cytokine macrophage migration inhibitory factor (MIF) ameliorates CDI. Here, we determined kinetics of MIF production and its association with a common genetic variant in leptin receptor (LEPR) using blood from patients with CDI. We found highest plasma MIF early after C difficile exposure and in individuals who express mutant/derived LEPR. Our data suggest that early-phase CDI provides a possible window of opportunity in which MIF targeting, potentially in combination with LEPR genotype, could have therapeutic utility.

2.
Nat Immunol ; 10(11): 1178-84, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19783988

ABSTRACT

Regulatory T cells (T(reg) cells) that express the transcription factor Foxp3 suppress the activity of other cells. Here we show that interleukin 10 (IL-10) produced by CD11b(+) myeloid cells in recombination-activating gene 1-deficient (Rag1(-/-)) recipient mice was needed to prevent the colitis induced by transferred CD4(+)CD45RB(hi) T cells. In Il10(-/-)Rag1(-/-) mice, T(reg) cells failed to maintain Foxp3 expression and regulatory activity. The loss of Foxp3 expression occurred only in recipients with colitis, which indicates that the requirement for IL-10 is manifested in the presence of inflammation. IL-10 receptor-deficient (Il10rb(-/-)) T(reg) cells also failed to maintain Foxp3 expression, which suggested that host IL-10 acted directly on the T(reg) cells. Our data indicate that IL-10 released from myeloid cells acts in a paracrine manner on T(reg) cells to maintain Foxp3 expression.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/immunology , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD11 Antigens/immunology , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Interleukin-10/metabolism , Intestines/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/immunology
3.
Anaerobe ; 53: 56-63, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29944928

ABSTRACT

Clostridium difficile is an important cause of nosocomial diarrhea in the western world. Toxins (A, B, and binary toxins) generated by C. difficile bacteria damage intestinal epithelial cells. Hallmarks of host response to C. difficile infection (CDI) include upregulation of inflammatory mediators and tissue infiltration by immune cells. Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine that is known to enhance the host immune response to infectious pathogens. Additionally, MIF can adversely impact host survival to numerous infections. The role of MIF in the pathogenesis of CDI remains poorly understood. Here, we show that patients with CDI had significantly higher circulating MIF compared to patients who had diarrhea but tested negative for C. difficile (non-CDI controls). Similarly, in a mouse model, C. difficile challenge significantly increased levels of plasma and tissue MIF. Antibody-mediated depletion of MIF decreased C. difficile-induced inflammatory responses, clinical disease, and mortality. Together, these results uncover a potential role for MIF in exacerbating CDI and suggest that use of anti-MIF antibodies may represent a therapeutic strategy to curb host inflammatory responses and improve disease outcomes in CDI.


Subject(s)
Antibodies/administration & dosage , Clostridioides difficile/growth & development , Clostridium Infections/pathology , Clostridium Infections/therapy , Immunologic Factors/administration & dosage , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/blood , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Female , Humans , Male , Mice, Inbred C57BL , Survival Analysis , Treatment Outcome
4.
Anaerobe ; 45: 101-105, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28223256

ABSTRACT

Clostridium difficile is the leading cause of healthcare-associated infections in the United States. Clinically, C. difficile-associated disease can present as asymptomatic colonization, self-limited diarrheal illness or severe colitis (that may result in death). This variability in disease course and outcomes suggests that host factors play an important role as key determinants of disease severity. Currently, there are several scoring indices to estimate severity of C. difficile-associated disease. Leukocytosis and renal failure are considered to be the most important predictors of C. difficile disease severity in hosts with a normal immune system. The degree of leukocytosis which is considered significant for severe disease and how it is scored vary amongst scoring indices. None of the scores have been prospectively validated, and while total WBC count is useful to estimate the magnitude of the host response in most patient populations, in immune-compromised patients like those receiving chemotherapy, solid organ transplant patients or hematopoietic stem cell transplants the WBC response can be variable or even absent making this marker of severity difficult to interpret. Other cellular subsets like neutrophils, eosinophils and lymphocytes provide important information about the host immune status and play an important role in the immune response against C. difficile infection. However, under the current scoring systems the role of these cellular subsets have been underestimated and only total white blood cell counts are taken into account. In this review we highlight the role of host leukocyte response to C. difficile challenge in the normal and immunocompromised host, and propose possible ways that would allow for a better representation of the different immune cell subsets (neutrophils, lymphocytes and eosinophils) in the current scoring indices.


Subject(s)
Clostridium Infections/immunology , Clostridium Infections/pathology , Immunocompromised Host , Leukocytes/immunology , Leukocytosis/pathology , Clostridium Infections/complications , Humans , Prospective Studies
5.
Anaerobe ; 41: 85-90, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27063896

ABSTRACT

Clostridium difficile is the most important cause of nosocomial infectious diarrhea in the western world. C. difficile infections are a major healthcare burden with approximately 500,000 new cases every year and an estimated annual cost of nearly $1 billion in the U.S. Furthermore, the infections are no longer restricted to health care facilities, and recent studies indicate spread of C. difficile infection to the community as well. The clinical spectrum of C. difficile infection ranges from asymptomatic colonization to severe diarrhea, fulminant colitis and death. This spectrum results from a complex interplay between bacterial virulence factors, the colonic microbiome and the host inflammatory response. The overall vigor of host inflammatory response is believed to be an important determinant of C. difficile disease severity, and a more robust immune response is associated with worse outcomes. Neutrophils are the primary cells that respond to C. difficile invasion and neutrophilic inflammation is the hallmark of C. difficile-associated disease. In this review, we will focus on the role of neutrophils (infiltration to infected tissue, pathogen clearance and resolution of inflammation) in the immuno-pathogenesis of C. difficile-associated disease (CDAD).


Subject(s)
Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/immunology , Neutrophils/immunology , Animals , Colitis/immunology , Colitis/microbiology , Enterocolitis, Pseudomembranous/microbiology , Humans , Immunity, Innate , Neutrophil Infiltration
6.
Nature ; 457(7229): 585-8, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-19060881

ABSTRACT

Aeroallergy results from maladaptive immune responses to ubiquitous, otherwise innocuous environmental proteins. Although the proteins targeted by aeroallergic responses represent a tiny fraction of the airborne proteins humans are exposed to, allergenicity is a quite public phenomenon-the same proteins typically behave as aeroallergens across the human population. Why particular proteins tend to act as allergens in susceptible hosts is a fundamental mechanistic question that remains largely unanswered. The main house-dust-mite allergen, Der p 2, has structural homology with MD-2 (also known as LY96), the lipopolysaccharide (LPS)-binding component of the Toll-like receptor (TLR) 4 signalling complex. Here we show that Der p 2 also has functional homology, facilitating signalling through direct interactions with the TLR4 complex, and reconstituting LPS-driven TLR4 signalling in the absence of MD-2. Mirroring this, airway sensitization and challenge with Der p 2 led to experimental allergic asthma in wild type and MD-2-deficient, but not TLR4-deficient, mice. Our results indicate that Der p 2 tends to be targeted by adaptive immune responses because of its auto-adjuvant properties. The fact that other members of the MD-2-like lipid-binding family are allergens, and that most defined major allergens are thought to be lipid-binding proteins, suggests that intrinsic adjuvant activity by such proteins and their accompanying lipid cargo may have some generality as a mechanism underlying the phenomenon of allergenicity.


Subject(s)
Allergens/immunology , Allergens/metabolism , Antigens, Dermatophagoides/immunology , Antigens, Dermatophagoides/metabolism , Molecular Mimicry/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Air , Allergens/chemistry , Allergens/genetics , Animals , Antigens, Dermatophagoides/chemistry , Antigens, Dermatophagoides/genetics , Arthropod Proteins , Asthma/genetics , Asthma/immunology , Cell Line , Disease Models, Animal , Female , Humans , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/deficiency , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/immunology , Lymphocyte Antigen 96/metabolism , Mice , Protein Binding , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
7.
Anaerobe ; 34: 182-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25638400

ABSTRACT

Obesity is among the most pressing health concerns in the world since it is increasingly common even in the developing world, and is clearly associated with increased risk for chronic debilitating diseases and death. Furthermore, obesity can influence the pathogenesis of infectious diseases by affecting the balance of pathogen clearance and pathological inflammation. The mechanisms that result in enhanced inflammation in obese individuals are poorly understood. Clostridium difficile is a major cause of nosocomial infections worldwide. Recent studies have shown that obesity is associated with increased risk of C. difficile infections. In this review, we will discuss our current knowledge of the role of obesity in determining risk of C. difficile infections, and focus on the role of the adipose tissue-derived cytokine leptin in C. difficile infections.


Subject(s)
Adipose Tissue/metabolism , Clostridioides difficile/physiology , Clostridium Infections/epidemiology , Host-Pathogen Interactions , Leptin/metabolism , Obesity/complications , Clostridium Infections/microbiology , Humans
8.
Infect Immun ; 82(1): 341-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166957

ABSTRACT

The role of leptin in the mucosal immune response to Clostridium difficile colitis, a leading cause of nosocomial infection, was studied in humans and in a murine model. Previously, a mutation in the receptor for leptin (LEPR) was shown to be associated with susceptibility to infectious colitis and liver abscess due to Entamoeba histolytica as well as to bacterial peritonitis. Here we discovered that European Americans homozygous for the same LEPR Q223R mutation (rs1137101), known to result in decreased STAT3 signaling, were at increased risk of C. difficile infection (odds ratio, 3.03; P = 0.015). The mechanism of increased susceptibility was studied in a murine model. Mice lacking a functional leptin receptor (db/db) had decreased clearance of C. difficile from the gut lumen and diminished inflammation. Mutation of tyrosine 1138 in the intracellular domain of LepRb that mediates signaling through the STAT3/SOCS3 pathway also resulted in decreased mucosal chemokine and cell recruitment. Collectively, these data support a protective mucosal immune function for leptin in C. difficile colitis partially mediated by a leptin-STAT3 inflammatory pathway that is defective in the LEPR Q223R mutation. Identification of the role of leptin in protection from C. difficile offers the potential for host-directed therapy and demonstrates a connection between metabolism and immunity.


Subject(s)
Clostridioides difficile , Clostridium Infections , Colitis/microbiology , Leptin/physiology , Receptors, Leptin/genetics , Animals , Chemokines/metabolism , Clostridioides difficile/genetics , Clostridioides difficile/immunology , Clostridium Infections/genetics , Clostridium Infections/immunology , Colitis/genetics , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry , Genetic Predisposition to Disease , Intestinal Mucosa/immunology , Intestines/microbiology , Leptin/immunology , Logistic Models , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Odds Ratio , Polymorphism, Genetic , Receptors, Leptin/deficiency , STAT3 Transcription Factor , Signal Transduction/physiology , Tyrosine/genetics
9.
J Infect Dis ; 208(6): 917-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23776194

ABSTRACT

Clostridium difficile is currently the leading cause of hospital-acquired infections in the United States. Here, we observed increased interleukin 23 (IL-23) protein levels in human colon biopsy specimens positive for C. difficile toxins, compared with levels in negative controls (P = .008) We also investigated the role of IL-23 during C. difficile infection, using 2 distinct murine models. Mice lacking IL-23 signaling had a significant increase in survival (100% [12 mice]), compared with control mice (16.7%-50% [12 mice]). These data suggest a new potential drug target for human C. difficile treatment and indicate the first link between IL-23 and disease severity during murine infection.


Subject(s)
Clostridioides difficile , Clostridium Infections/immunology , Colitis/immunology , Interleukin-23/immunology , Signal Transduction , Animals , Biopsy , Colitis/microbiology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Specimen Handling
10.
Toxins (Basel) ; 16(6)2024 May 24.
Article in English | MEDLINE | ID: mdl-38922136

ABSTRACT

Clostridioides difficile, a Gram-positive anaerobic bacterium, is the leading cause of hospital-acquired antibiotic-associated diarrhea worldwide. The severity of C. difficile infection (CDI) varies, ranging from mild diarrhea to life-threatening conditions such as pseudomembranous colitis and toxic megacolon. Central to the pathogenesis of the infection are toxins produced by C. difficile, with toxin A (TcdA) and toxin B (TcdB) as the main virulence factors. Additionally, some strains produce a third toxin known as C. difficile transferase (CDT). Toxins damage the colonic epithelium, initiating a cascade of cellular events that lead to inflammation, fluid secretion, and further tissue damage within the colon. Mechanistically, the toxins bind to cell surface receptors, internalize, and then inactivate GTPase proteins, disrupting the organization of the cytoskeleton and affecting various Rho-dependent cellular processes. This results in a loss of epithelial barrier functions and the induction of cell death. The third toxin, CDT, however, functions as a binary actin-ADP-ribosylating toxin, causing actin depolymerization and inducing the formation of microtubule-based protrusions. In this review, we summarize our current understanding of the interaction between C. difficile toxins and host cells, elucidating the functional consequences of their actions. Furthermore, we will outline how this knowledge forms the basis for developing innovative, toxin-based strategies for treating and preventing CDI.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Host Microbial Interactions , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Clostridium Infections/pathology , Gene Order , Inflammation/pathology , Humans , Animals
11.
Pathogens ; 13(2)2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38392856

ABSTRACT

Clostridioides difficile is the most important cause of healthcare-associated diarrhea in the United States. The high incidence and recurrence rates of C. difficile infection (CDI), associated with high morbidity and mortality, pose a public health challenge. Although antibiotics targeting C. difficile bacteria are the first treatment choice, antibiotics also disrupt the indigenous gut flora and, therefore, create an environment that is favorable for recurrent CDI. The challenge of treating CDI is further exacerbated by the rise of antibiotic-resistant strains of C. difficile, placing it among the top five most urgent antibiotic resistance threats in the USA. The evolution of antibiotic resistance in C. difficile involves the acquisition of new resistance mechanisms, which can be shared among various bacterial species and different C. difficile strains within clinical and community settings. This review provides a summary of commonly used diagnostic tests and antibiotic treatment strategies for CDI. In addition, it discusses antibiotic treatment and its resistance mechanisms. This review aims to enhance our current understanding and pinpoint knowledge gaps in antimicrobial resistance mechanisms in C. difficile, with an emphasis on CDI therapies.

12.
J Immunol ; 187(2): 733-47, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21666057

ABSTRACT

Although several subsets of intestinal APCs have been described, there has been no systematic evaluation of their phenotypes, functions, and regional localization to date. In this article, we used 10-color flow cytometry to define the major APC subsets in the small and large intestine lamina propria. Lamina propria APCs could be subdivided into CD11c(+)CD11b(-), CD11c(+)CD11b(+), and CD11c(dull)CD11b(+) subsets. CD11c(+)CD11b(-) cells were largely CD103(+)F4/80(-) dendritic cells (DCs), whereas the CD11c(+)CD11b(+) subset comprised CD11c(+)CD11b(+)CD103(+)F4/80(-) DCs and CD11c(+)CD11b(+)CD103(-)F4/80(+) macrophage-like cells. The majority of CD11c(dull)CD11b(+) cells were CD103(-)F4/80(+) macrophages. Although macrophages were more efficient at inducing Foxp3(+) regulatory T (T(reg)) cells than DCs, at higher T cell/APC ratios, all of the DC subsets efficiently induced Foxp3(+) T(reg) cells. In contrast, only CD11c(+)CD11b(+)CD103(+) DCs efficiently induced Th17 cells. Consistent with this, the regional distribution of CD11c(+)CD11b(+)CD103(+) DCs correlated with that of Th17 cells, with duodenum > jejunum > ileum > colon. Conversely, CD11c(+)CD11b(-)CD103(+) DCs, macrophages, and Foxp3(+) T(reg) cells were most abundant in the colon and scarce in the duodenum. Importantly, however, the ability of DC and macrophage subsets to induce Foxp3(+) T(reg) cells versus Th17 cells was strikingly dependent on the source of the mouse strain. Thus, DCs from C57BL/6 mice from Charles River Laboratories (that have segmented filamentous bacteria, which induce robust levels of Th17 cells in situ) were more efficient at inducing Th17 cells and less efficient at inducing Foxp3(+) T(reg) cells than DCs from B6 mice from The Jackson Laboratory. Thus, the functional specializations of APC subsets in the intestine are dependent on the T cell/APC ratio, regional localization, and source of the mouse strain.


Subject(s)
Antigen-Presenting Cells/cytology , Dendritic Cells/immunology , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Amino Acid Sequence , Animals , Antigen-Presenting Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Chronic Disease , Coculture Techniques , Colitis/genetics , Colitis/immunology , Colitis/pathology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Intestinal Mucosa/metabolism , Lymphocyte Count , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Organ Specificity/genetics , Organ Specificity/immunology , Species Specificity , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/cytology , Th17 Cells/metabolism
13.
J Immunol ; 183(2): 797-801, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19570826

ABSTRACT

IL-27 has recently been identified as a differentiation factor for the generation of IL-10-producing regulatory type 1 (Tr1) T cells. However, how IL-27 induces the expansion of Tr1 cells has not been elucidated. In this study we demonstrate that IL-27 drives the expansion and differentiation of IL-10-producing murine Tr1 cells by inducing three key elements: the transcription factor c-Maf, the cytokine IL-21, and the costimulatory receptor ICOS. IL-27-driven c-Maf expression transactivates IL-21 production, which acts as an autocrine growth factor for the expansion and/or maintenance of IL-27-induced Tr1 cells. ICOS further promotes IL-27-driven Tr1 cells. Each of those elements is essential, because loss of c-Maf, IL-21-signaling, or ICOS decreases the frequency of IL-27-induced differentiation of IL-10-producing Tr1 cells.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Cell Differentiation , Interleukins/immunology , Interleukins/physiology , Proto-Oncogene Proteins c-maf/physiology , T-Lymphocytes, Regulatory/cytology , Transcriptional Activation , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Inducible T-Cell Co-Stimulator Protein , Interleukin-10/biosynthesis , Interleukins/genetics , Mice , Proto-Oncogene Proteins c-maf/genetics , T-Lymphocytes, Regulatory/metabolism
14.
J Immunol ; 183(4): 2312-20, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19620304

ABSTRACT

IL-10 plays a central role in restraining the vigor of inflammatory responses, but the critical cellular sources of this counter-regulatory cytokine remain speculative in many disease models. Using a novel IL-10 transcriptional reporter mouse, we found an unexpected predominance of B cells (including plasma cells) among IL-10-expressing cells in peripheral lymphoid tissues at baseline and during diverse models of in vivo immunological challenge. Use of a novel B cell-specific IL-10 knockout mouse revealed that B cell-derived IL-10 nonredundantly decreases virus-specific CD8(+) T cell responses and plasma cell expansion during murine cytomegalovirus infection and modestly restrains immune activation after challenge with foreign Abs to IgD. In contrast, no role for B cell-derived IL-10 was evident during endotoxemia; however, although B cells dominated lymphoid tissue IL-10 production in this model, myeloid cells were dominant in blood and liver. These data suggest that B cells are an underappreciated source of counter-regulatory IL-10 production in lymphoid tissues, provide a clear rationale for testing the biological role of B cell-derived IL-10 in infectious and inflammatory disease, and underscore the utility of cell type-specific knockouts for mechanistic limning of immune counter-regulation.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Interleukin-10/physiology , Animals , B-Lymphocyte Subsets/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Disease Models, Animal , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Inflammation Mediators/physiology , Interleukin-10/biosynthesis , Interleukin-10/deficiency , Interleukin-10/genetics , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muromegalovirus/immunology , NIH 3T3 Cells
15.
Front Cell Infect Microbiol ; 11: 619192, 2021.
Article in English | MEDLINE | ID: mdl-33718269

ABSTRACT

Neutrophils are key first-responders in the innate immune response to C. difficile infection (CDI) and play a central role in disease pathogenesis. Studies have clearly shown that tissue neutrophil numbers need to be tightly regulated for optimal CDI outcomes: while excessive colonic neutrophilia is associated with severe CDI, neutrophil depletion also results in worse outcomes. However, the biological mechanisms that control CDI-induced neutrophilia remain poorly defined. C-X-C chemokine receptor 2 (CXCR2) is a chemotactic receptor that is critical in neutrophil mobilization from bone marrow to blood and tissue sites. We have previously reported that a single nucleotide polymorphism (SNP) in leptin receptor (LEPR), present in up to 50% of people, influenced CDI-induced neutrophil CXCR2 expression and tissue neutrophilia. Homozygosity for mutant LEPR (i.e. RR genotype) was associated with higher CXCR2 expression and more tissue neutrophils. Here, we investigated the biological mechanisms that regulate neutrophil CXCR2 expression after CDI, and the influence of host genetics on this process. Our data reveal that: a) CXCR2 plays a key role in CDI-induced neutrophil extravasation from blood to colonic tissue; b) plasma from C. difficile-infected mice upregulated CXCR2 on bone marrow neutrophils; c) plasma from C. difficile-infected RR mice induced a higher magnitude of CXCR2 upregulation and had more IL-1ß; and d) IL-1ß neutralization reduced CXCR2 expression on bone marrow and blood neutrophils and their subsequent accrual to colonic tissue. In sum, our data indicate that IL-1ß is a key molecular mediator that communicates between gastro-intestinal tract (i.e. site of CDI) and bone marrow (i.e. primary neutrophil reservoir) and regulates the intensity of CDI-induced tissue neutrophilia by modulating CXCR2 expression. Further, our studies highlight the importance of host genetics in affecting these innate immune responses and provide novel insights into the mechanisms by which a common SNP influences CDI-induced neutrophilia.


Subject(s)
Clostridioides difficile , Neutrophils , Animals , Clostridioides , Clostridium Infections , Interleukin-1beta/genetics , Mice , Polymorphism, Single Nucleotide , Receptors, Interleukin-8B , Receptors, Leptin
16.
Mucosal Immunol ; 14(6): 1224-1234, 2021 11.
Article in English | MEDLINE | ID: mdl-33958704

ABSTRACT

Epidemiological evidence establishes obesity as an independent risk factor for increased susceptibility and severity to viral respiratory pneumonias associated with H1N1 influenza and SARS-CoV-2 pandemics. Given the global obesity prevalence, a better understanding of the mechanisms behind obese susceptibility to infection is imperative. Altered immune cell metabolism and function are often perceived as a key causative factor of dysregulated inflammation. However, the contribution of adipocytes, the dominantly altered cell type in obesity with broad inflammatory properties, to infectious disease pathogenesis remains largely ignored. Thus, skewing of adipocyte-intrinsic cellular metabolism may lead to the development of pathogenic inflammatory adipocytes, which shape the overall immune responses by contributing to either premature immunosenescence, delayed hyperinflammation, or cytokine storm in infections. In this review, we discuss the underappreciated contribution of adipocyte cellular metabolism and adipocyte-produced mediators on immune system modulation and how such interplay may modify disease susceptibility and pathogenesis of influenza and SARS-CoV-2 infections in obese individuals.


Subject(s)
Adipocytes/metabolism , COVID-19/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Influenza, Human/metabolism , SARS-CoV-2/metabolism , Adipocytes/pathology , Adipocytes/virology , COVID-19/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Influenza, Human/pathology
17.
Mucosal Immunol ; 14(2): 500-510, 2021 03.
Article in English | MEDLINE | ID: mdl-32811993

ABSTRACT

Severe Clostridiodes difficile infection (CDI) is life-threatening and responds poorly to treatment. Obesity is associated with development of severe CDI. Therefore, to define the mechanisms that exacerbate disease severity, we examined CDI pathogenesis in high-fat diet (HFD)-fed obese mice. Compared to control mice, HFD-fed mice failed to clear C. difficile bacteria which resulted in protracted diarrhea, weight loss and colonic damage. After infection, HFD-induced obese mice had an intestinal bile acid (BA) pool that was dominated by primary BAs which are known promoters of C. difficile spore germination, and lacked secondary BAs that inhibit C. difficile growth. Concurrently, synthesis of primary BAs from liver was significantly increased in C. difficile-infected HFD-fed mice. A key pathway that regulates hepatic BA synthesis is via feedback inhibition from intestinal Farnesoid X receptors (FXRs). Our data reveal that the proportion of FXR agonist BAs to FXR antagonist BAs in the intestinal lumen was significantly reduced in HFD-fed mice after CDI. Treatment of HFD-fed mice with an FXR agonist Obeticholic acid, resulted in decreased primary BA synthesis, fewer C. difficile bacteria and better CDI outcomes. Thus, OCA treatment holds promise as a therapy for severe CDI.


Subject(s)
Anticholesteremic Agents/therapeutic use , Chenodeoxycholic Acid/analogs & derivatives , Clostridioides difficile/physiology , Clostridium Infections/drug therapy , Obesity/drug therapy , Animals , Chenodeoxycholic Acid/therapeutic use , Diet, High-Fat , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Obese , RNA-Binding Proteins/metabolism
18.
Mol Pharmacol ; 73(6): 1844-56, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18372398

ABSTRACT

Crossing the Cyp1a1/1a2(-/-) double-knockout mouse with the Cyp1b1(-/-) single-knockout mouse, we generated the Cyp1a1/1a2/1b1(-/-) triple-knockout mouse. In this triple-knockout mouse, statistically significant phenotypes (with incomplete penetrance) included slower weight gain and greater risk of embryolethality before gestational day 11, hydrocephalus, hermaphroditism, and cystic ovaries. Oral benzo[a]pyrene (BaP) daily for 18 days in the Cyp1a1/1a2(-/-) produced the same degree of marked immunosuppression as seen in the Cyp1a1(-/-) mouse; we believe this reflects the absence of intestinal CYP1A1. Oral BaP-treated Cyp1a1/1a2/1b1(-/-) mice showed the same "rescued" response as that seen in the Cyp1a1/1b1(-/-) mouse; we believe this reflects the absence of CYP1B1 in immune tissues. Urinary metabolite profiles were dramatically different between untreated triple-knockout and wild-type; principal components analysis showed that the shifts in urinary metabolite patterns in oral BaP-treated triple-knockout and wild-type mice were also strikingly different. Liver microarray cDNA differential expression (comparing triple-knockout with wild-type) revealed at least 89 genes up- and 62 genes down-regulated (P-value < or = 0.00086). Gene Ontology "classes of genes" most perturbed in the untreated triple-knockout (compared with wild-type) include lipid, steroid, and cholesterol biosynthesis and metabolism; nucleosome and chromatin assembly; carboxylic and organic acid metabolism; metal-ion binding; and ion homeostasis. In the triple-knockout compared with the wild-type mice, response to zymosan-induced peritonitis was strikingly exaggerated, which may well reflect down-regulation of Socs2 expression. If a single common molecular pathway is responsible for all of these phenotypes, we suggest that functional effects of the loss of all three Cyp1 genes could be explained by perturbations in CYP1-mediated eicosanoid production, catabolism and activities.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2/genetics , Phenotype , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 CYP1B1 , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
J Leukoc Biol ; 82(2): 265-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17470533

ABSTRACT

As all immune responses have potential for damaging the host, tight regulation of such responses--in amplitude, space, time and character--is essential for maintaining health and homeostasis. It was thus inevitable that the initial wave of papers on the role of Toll-like receptors (TLRs), NOD-like receptors (NLRs) and RIG-I-like receptors (RLRs) in activating innate and adaptive immune responses would be followed by a second wave of reports focusing on the mechanisms responsible for restraining and modulating signaling by these receptors. This overview outlines current knowledge and controversies about the immunobiology of the RP105/MD-1 complex, a modulator of the most robustly signaling TLR, TLR4.


Subject(s)
Antigens, CD/physiology , Signal Transduction , Toll-Like Receptor 4/physiology , Animals , Antigens, CD/immunology , Gene Expression Regulation , Humans , Toll-Like Receptor 4/genetics
20.
J Immunol Methods ; 311(1-2): 19-30, 2006 Apr 20.
Article in English | MEDLINE | ID: mdl-16563424

ABSTRACT

Functional analysis of the DNA regulatory regions that control gene expression has largely been performed through transient transfection of promoter-reporter constructs into transformed cells. However, transformed cells are often poor models of primary cells. To directly analyze DNA regulatory regions in primary cells, we generated a novel adenoviral luciferase reporter vector, pShuttle-luciferase-GFP (pSLUG) that contains a promoterless luciferase cassette (with an upstream cloning site) for probing promoter activity, and a GFP expression cassette that allows for the identification of transduced cells. Recombinant adenoviruses generated from this vector can transduce a wide range of primary immune cells with high efficiency, including human macrophages, dendritic cells and T cells; and mouse T cells transgenic for the coxsackie and adenoviral receptor (CAR). In primary T cells, we show inducible nuclear factor of activated T cells (NF-AT) activity using a recombinant pSLUG adenovirus containing a consensus NF-AT promoter. We further show inducible IL-12/23 p40 promoter activity in primary macrophages and dendritic cells using a recombinant pSLUG adenovirus containing the proximal human IL-12/23 p40 promoter. The pSLUG system promises to be a powerful tool for the analysis of DNA regulatory regions in diverse types of primary immune cells.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Promoter Regions, Genetic/physiology , Regulatory Sequences, Nucleic Acid/immunology , T-Lymphocytes/physiology , Transduction, Genetic/methods , Animals , Dendritic Cells/immunology , Dendritic Cells/physiology , Flow Cytometry , Gene Expression Regulation/genetics , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-2/immunology , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/genetics , Interleukins/immunology , Jurkat Cells , Luciferases/genetics , Macrophages/immunology , Macrophages/physiology , Mice , Mice, Transgenic , NFATC Transcription Factors/biosynthesis , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Promoter Regions, Genetic/immunology , Regulatory Sequences, Nucleic Acid/genetics , T-Lymphocytes/immunology , T-Lymphocytes/virology
SELECTION OF CITATIONS
SEARCH DETAIL