Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
J Immunol ; 211(8): 1232-1239, 2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37672039

RESUMO

Intestinal inflammatory diseases affect millions of people worldwide, and one class of drugs showing promise toward treatment of several inflammatory diseases is probiotics. Numerous studies have been performed using probiotics to prevent and treat intestinal inflammatory diseases. Most of these studies used intact bacteria, and neither the active molecule nor the molecular mechanisms by which they affect immune responses are known. We have shown that the probiotic Bacillus subtilis is anti-inflammatory and can protect mice from acute colitis induced by the enteric pathogen Citrobacter rodentium. We identified and purified the active molecule, exopolysaccharide (EPS), and showed that it protects mice from C. rodentium-induced colitis by inducing anti-inflammatory M2 macrophages or inhibitory dendritic cells (DCs), both of which inhibit excessive T cell responses. We showed previously that EPS affects macrophages and DCs in a TLR4-dependent manner, and in the current study we asked how EPS induces these anti-inflammatory cells and how they function to inhibit T cells. By investigating the signaling downstream of TLR4 that leads to acquisition of inhibitory properties of macrophages and DCs, we found that EPS induces expression of the inhibitory molecule IDO in bone marrow-derived DCs, and that inhibition of T cell proliferation by IDO-expressing bone marrow-derived DCs utilizes the kynurenine/aryl hydrocarbon receptor circuit. Furthermore, unlike LPS, EPS does not induce inflammatory cytokines upon injection in vivo, directly demonstrating different outcomes induced by two different TLR4 agonists.


Assuntos
Colite , Probióticos , Humanos , Camundongos , Animais , Receptor 4 Toll-Like/metabolismo , Bacillus subtilis , Anti-Inflamatórios/farmacologia , Células Dendríticas
2.
J Immunol ; 206(9): 2101-2108, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33846225

RESUMO

Acute graft-versus-host disease (aGvHD) is a severe, often lethal, complication of hematopoietic stem cell transplantation, and although prophylactic regimens are given as standard pretransplantation therapy, up to 60% of these patients develop aGvHD, and require additional immunosuppressive intervention. We treated mice with a purified probiotic molecule, exopolysaccharide (EPS) from Bacillus subtilis, shortly before and after induction of aGvHD and found that, whereas only 10% of control mice survived to day 80, 70% of EPS-treated mice survived to 80 d. EPS treatment of donor-only mice resulted in ∼60% survival. Using a biosensor mouse model to assess inflammation in live mice during aGvHD, we found that EPS prevented the activation of alloreactive donor T cells. In vitro, EPS did not affect T cells directly but, instead, induced bone marrow-derived dendritic cells (BMDCs) that displayed characteristics of inhibitory dendritic cells (DCs). Development of these BMDCs required TLR4 signaling through both MyD88 and TRIF pathways. Using BMDCs derived from IDO knockout mice, we showed that T cell inhibition by EPS-treated BMDCs was mediated through the suppressive effects of IDO. These studies describe a bacterial molecule that modulates immune responses by inducing inhibitory DCs in a TLR4-dependent manner, and these cells have the capacity to inhibit T cell activation through IDO. We suggest that EPS or EPS-treated DCs can serve as novel agents for preventing aGvHD.


Assuntos
Bacillus subtilis/química , Doença Enxerto-Hospedeiro/imunologia , Polissacarídeos Bacterianos/imunologia , Animais , Bacillus subtilis/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Immunology ; 163(3): 293-309, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33524161

RESUMO

The fetal and neonatal immune systems are uniquely poised to generate tolerance to self, maternal and environmental antigens encountered in the womb and shortly after birth. However, the tolerogenic nature of fetal and neonatal immunity can be detrimental in the context of pathogens, leading to overwhelming bacterial infections or chronic viral infections. A variety of mechanisms contribute to fetal and neonatal tolerance, including a propensity to generate Foxp3+ regulatory T cells (Treg cells). However, the mechanism(s) of fetal Foxp3+ T-cell differentiation, the specific antigen-presenting cells required and factors that inhibit Treg generation after the neonatal period are poorly understood. Here, we demonstrate that a subset of CD14+ monocytes expressing the scavenger molecule, CD36, can generate CD4+ and CD8+ T cells that coexpress Foxp3 and T-bet from both umbilical cord blood. These Foxp3+ T-bet+ T cells potently suppress T-cell proliferation and ameliorate xenogeneic graft-versus-host disease. CD14+  CD36+ monocytes provide known Treg-inducing signals: membrane-bound transforming growth factor-beta and retinoic acid. Unexpectedly, adult peripheral blood monocytes are also capable of inducing Foxp3+ T cells from both cord blood and adult peripheral naïve T cells. The induction of Foxp3+ T cells in umbilical cord blood by monocytes was inhibited by the lymphoid fraction of adult peripheral blood cells. These studies highlight a novel immunoregulatory role of monocytes and suggest that antigen presentation by CD36hi monocytes may contribute to the peripheral development of Foxp3+ T-bet+ T cells with regulatory functions in both neonates and adults.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Monócitos/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Antígenos CD36/metabolismo , Diferenciação Celular , Células Cultivadas , Sangue Fetal/citologia , Feto , Fatores de Transcrição Forkhead/metabolismo , Humanos , Tolerância Imunológica , Terapia de Imunossupressão , Ativação Linfocitária , Proteínas com Domínio T/genética , Transplante Heterólogo
4.
J Immunol ; 203(9): 2497-2507, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31562211

RESUMO

Inflammasomes are multiprotein complexes that coordinate cellular inflammatory responses and mediate host defense. Following recognition of pathogens and danger signals, inflammasomes assemble and recruit and activate caspase-1, the cysteine protease that cleaves numerous downstream targets, including pro-IL-1ß and pro-IL-18 into their biologically active form. In this study, we sought to develop a biosensor that would allow us to monitor the initiation, progression, and resolution of inflammation in living animals. To this end, we inserted a known caspase-1 target sequence into a circularly permuted luciferase construct that becomes bioluminescent upon protease cleavage. This biosensor was activated in response to various inflammatory stimuli in human monocytic cell lines and murine bone marrow-derived macrophages. Next, we generated C57BL/6 transgenic mice constitutively expressing the caspase-1 biosensor. We were able to monitor the spatiotemporal dynamics of caspase-1 activation and onset of inflammation in individual animals in the context of a systemic bacterial infection, colitis, and acute graft-versus-host disease. These data established a model whereby the development and progression of inflammatory responses can be monitored in the context of these and other mouse models of disease.


Assuntos
Técnicas Biossensoriais/métodos , Caspase 1/análise , Inflamação/etiologia , Animais , Apoptose , Colite/enzimologia , Progressão da Doença , Doença Enxerto-Hospedeiro/enzimologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/enzimologia , Células THP-1
5.
Infect Immun ; 88(4)2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-31932326

RESUMO

Staphylococcus aureus is a Gram-positive opportunistic pathogen that causes a variety of diseases. Bloodstream infection is the most severe, with mortality rates reaching 20 to 50%. Exopolysaccharide (EPS) from the probiotic Bacillus subtilis reduces bacterial burden and inflammation during S. aureus bloodstream infection in mice. Protection is due, in part, to hybrid macrophages that restrict S. aureus growth through reactive oxygen species and to limiting superantigen-induced T cell activation and interferon gamma (IFN-γ) production during infection. A decrease in IFN-γ production was observed within 24 h after infection, and here, we investigated how EPS abrogates its production. We discovered that S. aureus uses a rapid, superantigen-independent mechanism to induce host IFN-γ and that this is mediated by interleukin-12 (IL-12) activation of NK cells. Furthermore, we found that EPS limits IFN-γ production by modulating host immunity in a Toll-like receptor 4 (TLR4)-dependent manner, a signaling pathway that is required for EPS-mediated protection from S. aureus infection in vivo We conclude that EPS protects hosts from acute bloodstream S. aureus infection not only by inducing macrophages that restrict S. aureus growth and inhibit superantigen-activated T cells but also by limiting NK cell production of IFN-γ after S. aureus infection in a TLR4-dependent manner.


Assuntos
Bacteriemia/prevenção & controle , Interferon gama/antagonistas & inibidores , Células Matadoras Naturais/imunologia , Polissacarídeos Bacterianos/administração & dosagem , Probióticos/administração & dosagem , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/imunologia , Animais , Bacteriemia/imunologia , Modelos Animais de Doenças , Fatores Imunológicos/administração & dosagem , Interleucina-12/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/imunologia , Análise de Sobrevida , Receptor 4 Toll-Like/metabolismo
6.
Infect Immun ; 87(1)2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30396894

RESUMO

Staphylococcus aureus causes severe systemic infection with high mortality rates. We previously identified exopolysaccharide (EPS) from a probiotic, Bacillus subtilis, that induces anti-inflammatory macrophages with an M2 phenotype and protects mice from Citrobacter rodentium-induced colitis. We tested if EPS could protect from systemic infection induced by S. aureus and found that EPS-treated mice had enhanced survival as well as reduced weight loss, systemic inflammation, and bacterial burden. While macrophages from EPS-treated mice display an M2 phenotype, they also restrict growth of internalized S. aureus through reactive oxygen species (ROS), reminiscent of proinflammatory phagocytes. These EPS-induced macrophages also limit T cell activation by S. aureus superantigens, and EPS abrogates systemic induction of gamma interferon after infection. We conclude that B. subtilis EPS is an immunomodulatory agent that induces hybrid macrophages that bolster antibacterial immunity and simultaneously limit inflammation, reducing disease burden and promoting host survival.


Assuntos
Fatores Imunológicos/administração & dosagem , Inflamação/prevenção & controle , Macrófagos/imunologia , Polissacarídeos Bacterianos/administração & dosagem , Probióticos/administração & dosagem , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/crescimento & desenvolvimento , Animais , Modelos Animais de Doenças , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Staphylococcus aureus/efeitos dos fármacos , Análise de Sobrevida , Resultado do Tratamento
7.
J Immunol ; 198(9): 3471-3479, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28320833

RESUMO

B lymphopoiesis arrests precipitously in rabbits such that by 2-4 mo of age, before sexual maturity, little to no B lymphopoiesis occurs in the bone marrow (BM). Previously, we showed that in mice, adipocytes inhibit B lymphopoiesis in vitro by inducing inflammatory myeloid cells, which produce IL-1ß. In this study, we characterized rabbit BM after the arrest of B lymphopoiesis and found a dramatic increase in fat, increased CD11b+ myeloid cells, and upregulated expression of the inflammatory molecules, IL-1ß and S100A9, by the myeloid cells. We added BM fat, CD11b+ myeloid cells, and recombinant S100A9 to B lymphopoiesis cultures and found that they inhibited B lymphopoiesis and enhanced myelopoiesis. Unlike IL-1ß, which inhibits B lymphopoiesis by acting on early lymphoid progenitors, S100A9 inhibits B lymphopoiesis by acting on myeloid cells and promoting the release of inflammatory molecules, including IL-1ß. Many molecules produced by adipocytes activate the NLRP3 inflammasome, and the NLRP3 inhibitor, glibenclamide, restored B lymphopoiesis and minimized induction of myeloid cells induced by adipocyte-conditioned medium in vitro. We suggest that fat provides an inflammatory microenvironment in the BM and promotes/activates myeloid cells to produce inflammatory molecules such as IL-1ß and S100A9, which negatively regulate B lymphopoiesis.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Linfócitos B/fisiologia , Células da Medula Óssea/fisiologia , Calgranulina B/metabolismo , Microambiente Celular , Tecido Adiposo/patologia , Animais , Antígeno CD11b/metabolismo , Células Cultivadas , Glibureto/farmacologia , Interleucina-1beta/metabolismo , Linfopoese/efeitos dos fármacos , Camundongos , Mielopoese/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Coelhos
8.
J Immunol ; 198(7): 2689-2698, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28202619

RESUMO

Commensal bacteria contribute to immune homeostasis in the gastrointestinal tract; however, the underlying mechanisms for this are not well understood. A single dose of exopolysaccharide (EPS) from the probiotic spore-forming bacterium Bacillus subtilis protects mice from acute colitis induced by the enteric pathogen Citrobacter rodentium Adoptive transfer of macrophage-rich peritoneal cells from EPS-treated mice confers protection from disease to recipient mice. In vivo, EPS induces development of anti-inflammatory M2 macrophages in a TLR4-dependent manner, and these cells inhibit T cell activation in vitro and in C. rodentium-infected mice. In vitro, M2 macrophages inhibit CD4+ and CD8+ T cells. The inhibition of CD4+ T cells is dependent on TGF-ß, whereas inhibition of CD8+ T cells is dependent on TGF-ß and PD-L1. We suggest that administration of B. subtilis EPS can be used to broadly inhibit T cell activation and, thus, control T cell-mediated immune responses in numerous inflammatory diseases.


Assuntos
Colite/imunologia , Microbioma Gastrointestinal/imunologia , Macrófagos/imunologia , Polissacarídeos Bacterianos/imunologia , Linfócitos T/imunologia , Transferência Adotiva , Animais , Bacillus subtilis/imunologia , Citrobacter rodentium , Colite/microbiologia , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/patologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
9.
J Immunol ; 195(6): 2666-74, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26268654

RESUMO

B lymphopoiesis declines with age, and this decline correlates with increased adipose tissue in the bone marrow (BM). Also, adipocyte-derived factors are known to inhibit B lymphopoiesis. Using cocultures of mouse BM cells with OP9 stromal cells, we found that adipocyte-conditioned medium induces the generation of CD11b(+)Gr1(+) myeloid cells, which inhibit B cell development in vitro. Adipocyte-conditioned medium-induced CD11b(+)Gr1(+) cells express Arg1 (arginase) and Nos2 (inducible NO synthase) and suppress CD4(+) T cell proliferation, indicating that these cells are myeloid-derived suppressor cells (MDSCs). Blocking arginase and inducible NO synthase did not restore B lymphopoiesis, indicating that inhibition is not mediated by these molecules. Transwell and conditioned-medium experiments showed that MDSCs inhibit B lymphopoiesis via soluble factors, and by cytokine array we identified IL-1 as an important factor. Addition of anti-IL-1 Abs restored B lymphopoiesis in BM cultures containing MDSCs, showing that MDSC inhibition of B lymphopoiesis is mediated by IL-1. By treating hematopoietic precursors with IL-1, we found that multipotent progenitors are targets of IL-1. This study uncovers a novel function for MDSCs to inhibit B lymphopoiesis through IL-1. We suggest that inflammaging contributes to a decline of B lymphopoiesis in aged individuals, and furthermore, that MDSCs and IL-1 provide therapeutic targets for restoration of B lymphopoiesis in aged and obese individuals.


Assuntos
Adipócitos/citologia , Linfócitos B/citologia , Imunossenescência/imunologia , Interleucina-1/imunologia , Linfopoese/efeitos dos fármacos , Adipócitos/imunologia , Animais , Anticorpos/farmacologia , Arginase/biossíntese , Linfócitos B/imunologia , Células da Medula Óssea/citologia , Antígeno CD11b/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Inflamação/imunologia , Interleucina-1/antagonistas & inibidores , Interleucina-1/biossíntese , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/imunologia , Células Mieloides/imunologia , Óxido Nítrico Sintase Tipo II/biossíntese
10.
J Immunol ; 192(10): 4813-20, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24740503

RESUMO

Host inflammatory responses against pathogenic organisms can be abrogated by commensals; however, the molecular mechanisms by which pathogenesis is prevented are still poorly understood. Previous studies demonstrated that administration of a single dose of Bacillus subtilis prevented disease and inflammation by the enteric mouse pathogen Citrobacter rodentium, which causes disease similar to the human pathogen enteropathogenic Escherichia coli. No protection was observed when an exopolysaccharide (EPS)-deficient mutant of B. subtilis was used, suggesting that EPS are the protective factor. In this study, we isolated and characterized EPS and showed that they also prevent C. rodentium-associated intestinal disease after a single injection. Protection is TLR4 dependent because EPS-treated TLR4 knockout mice developed disease. Furthermore, protection could be conveyed to wild-type mice by adoptive transfer of macrophage-rich peritoneal cells from EPS-treated mice. We found that EPS specifically bind peritoneal macrophages, and because mice lacking MyD88 signaling in myeloid cells were not protected by EPS, we conclude that bacterial EPS prevent colitis in a TLR4-dependent manner that requires myeloid cells. These studies provide a simple means of preventing intestinal inflammation caused by enteric pathogens.


Assuntos
Bactérias/imunologia , Intestinos/imunologia , Macrófagos Peritoneais/imunologia , Polissacarídeos Bacterianos/imunologia , Animais , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Intestinos/microbiologia , Intestinos/patologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Polissacarídeos Bacterianos/farmacologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
11.
J Immunol ; 193(12): 5951-9, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25385821

RESUMO

Microbial and host cell interactions stimulate rabbit B cells to diversify the primary Ab repertoire in GALT. B cells at the base of appendix follicles begin proliferating and diversifying their V-(D)-J genes around 1 wk of age, ∼5 d after B cells first begin entering appendix follicles. To gain insight into the microbial and host cell interactions that stimulate B cells to diversify the primary Ab repertoire, we analyzed B cell trafficking within follicles during the first week of life. We visualized B cells, as well as chemokines that mediate B cell homing in lymphoid tissues, by in situ hybridization, and we examined B cell chemokine receptor expression by flow cytometry. We found that B cells were activated and began downregulating their BCRs well before a detectable B cell proliferative region appeared at the follicle base. The proliferative region was similar to germinal center dark zones, in that it exhibited elevated CXCL12 mRNA expression, and B cells that upregulated CXCR4 mRNA in response to signals acquired from selected intestinal commensals localized in this region. Our results suggest that after entering appendix follicles, B cells home sequentially to the follicle-associated epithelium, the follicular dendritic cell network, the B cell/T cell boundary, and, ultimately, the base of the follicle, where they enter a proliferative program and diversify the primary Ab repertoire.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Movimento Celular , Quimiocinas/metabolismo , Animais , Animais Recém-Nascidos , Apêndice/imunologia , Apêndice/metabolismo , Quimiocina CCL19/genética , Quimiocina CCL19/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/metabolismo , Quimiocina CCL21/genética , Quimiocina CCL21/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Expressão Gênica , Imuno-Histoquímica , Imunofenotipagem , Modelos Biológicos , Fenótipo , RNA Mensageiro/genética , Coelhos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo
12.
Infect Immun ; 83(11): 4383-91, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26324536

RESUMO

Clostridium difficile infection (CDI) is a major cause of health care-associated disease. CDI initiates with ingestion of C. difficile spores, germination in the gastrointestinal (GI) tract, and then colonization of the large intestine. The interactions between C. difficile cells and other bacteria and with host mucosa during CDI remain poorly understood. Here, we addressed the hypothesis that, in a mouse model of CDI, C. difficile resides in multicellular communities (biofilms) in association with host mucosa. To do this, we paraffin embedded and then sectioned the GI tracts of infected mice at various days postinfection (p.i.). We then used fluorescent in situ hybridization (FISH) with 16S rRNA probes targeting most bacteria as well as C. difficile specifically. The results revealed that C. difficile is present as a minority member of communities in the outer (loose) mucus layer, in the cecum and colon, starting at day 1 p.i. To generate FISH probes that identify bacteria within mucus-associated communities harboring C. difficile, we characterized bacterial populations in the infected mouse GI tract using 16S rRNA gene sequence analysis of bacterial DNA prepared from intestinal content. This analysis revealed the presence of genera of several families belonging to Bacteroidetes and Firmicutes. These data suggest that formation of multispecies communities associated with the mucus of the cecum and colon is an important early step in GI tract colonization. They raise the possibility that other bacterial species in these communities modulate the ability of C. difficile to successfully colonize and, thereby, cause disease.


Assuntos
Bactérias/isolamento & purificação , Clostridioides difficile/fisiologia , Infecções por Clostridium/microbiologia , Animais , Bactérias/classificação , Bactérias/genética , Modelos Animais de Doenças , Trato Gastrointestinal/microbiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microbiota
13.
Infect Immun ; 82(2): 522-31, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478068

RESUMO

Clostridium difficile rivals methicillin-resistant Staphylococcus aureus as the primary hospital-acquired infection. C. difficile infection (CDI) caused by toxins A and/or B can manifest as mild diarrhea to life-threatening pseudomembranous colitis. Although most patients recover fully from CDI, ~20% undergo recurrent disease. Several studies have demonstrated a correlation between anti-toxin antibody (Ab) and decreased recurrence; however, the contributions of the systemic and mucosal Ab responses remain unclear. Our goal was to use the CDI mouse model to characterize the protective immune response to C. difficile. C57BL/6 mice infected with epidemic C. difficile strain BI17 developed protective immunity against CDI and did not develop CDI upon rechallenge; they generated systemic IgG and IgA as well as mucosal IgA Ab to toxin. To determine if protective immunity to C. difficile could be generated in immunodeficient individuals, we infected CD4(-/-) mice and found that they generated both mucosal and serum IgA anti-toxin Abs and were protected from CDI upon rechallenge, with protection dependent on major histocompatibility complex class II (MHCII) expression; no IgG anti-toxin Ab was found. We found that protection was likely due to neutralizing mucosal IgA Ab. In contrast, pIgR(-/-) mice, which lack the receptor to transcytose polymeric Ab across the epithelium, were also protected from CDI, suggesting that although mucosal anti-toxin Ab may contribute to protection, it is not required. We conclude that protection from CDI can occur by several mechanisms and that the mechanism of protection is determined by the state of immunocompetence of the host.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Clostridioides difficile/imunologia , Infecções por Clostridium/imunologia , Receptores de Imunoglobulina Polimérica/imunologia , Animais , Anticorpos Antibacterianos/sangue , Infecções por Clostridium/microbiologia , Infecções por Clostridium/prevenção & controle , Modelos Animais de Doenças , Imunoglobulina A/análise , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Imunoglobulina Polimérica/deficiência
14.
Eur J Immunol ; 43(6): 1484-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23468368

RESUMO

In species other than mouse, little is known about the origin and development of marginal zone (MZ) B cells. Using cross-reactive antibodies, we identified and characterized splenic MZ B cells in rabbits as CD27(+) CD23(-). In rabbits in which organized gut-associated lymphoid tissue (GALT) was surgically removed at birth, we found only CD23(+) follicular (FO) B cells and almost no CD27(+) MZ B cells in the spleen, indicating that GALT is required for the development of splenic MZ B cells. These findings lead us to suggest that commensal microbiota contribute to the development of MZ B cells.


Assuntos
Linfócitos B/imunologia , Intestinos/imunologia , Baço/imunologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Tecido Linfoide/cirurgia , Metagenoma/imunologia , Camundongos , Coelhos , Receptores de IgE/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
16.
J Immunol ; 189(9): 4379-86, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23002443

RESUMO

B lymphopoiesis declines with age, and in rabbits this occurs by 8 wk of age. We found that CFU fibroblasts (CFU-Fs) in the bone marrow (BM) decrease 10-fold by a few weeks of age and that the CFU-Fs preferentially differentiate into adipocytes instead of osteoblasts. BM becomes filled with fat spaces during this time, making rabbit a unique model to study the effects of accelerated fat accumulation on B lymphopoiesis. We show that adipocytes of both rabbit and human secrete a soluble factor(s) that inhibits B lymphopoiesis, and we tested if this inhibition was due to effects on the BM stroma or hematopoietic progenitors. Pretreatment of BM mononuclear cells with adipocyte conditioned medium dramatically inhibited their differentiation into proB cells in cocultures with OP9 stromal cells. In contrast, pretreatment of OP9 stromal cells with adipocyte conditioned medium had no effect on B lymphopoiesis. Using human hematopoietic stem cells, we show that inhibition by the adipocyte-derived factor occurred at the common lymphoid progenitor to preproB cell stage. We propose that the age-related decline in B lymphopoiesis is due to a decrease in CFU-Fs, an increase in adipocytes, and an adipocyte-derived factor that blocks B lymphopoiesis at the common lymphoid progenitor to preproB cell stage.


Assuntos
Adipócitos/fisiologia , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/imunologia , Proteínas de Ligação a Ácido Graxo/fisiologia , Inibidores do Crescimento/fisiologia , Linfopoese/imunologia , Animais , Subpopulações de Linfócitos B/metabolismo , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Humanos , Osteoblastos/citologia , Osteoblastos/imunologia , Osteoblastos/metabolismo , Coelhos , Células-Tronco/citologia , Células-Tronco/imunologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
17.
Transplant Cell Ther ; 30(1): 79.e1-79.e10, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37924979

RESUMO

Graft-versus-host disease (GVHD) is a primary and often lethal complication of allogenic hematopoietic stem cell transplantation (HSCT). Prophylactic regimens for GVHD are given as standard pretransplantation therapy; however, up to 50% of these patients develop acute GVHD (aGVHD) and require additional immunosuppressive intervention. Using a mouse GVHD model, we previously showed that injecting mice with exopolysaccharide (EPS) from Bacillus subtilis prior to GVHD induction significantly increased 80-day survival after transplantation of complete allogeneic major histocompatibility complex-mismatched cells. To ask whether EPS might also inhibit GVHD in humans, we used humanized NSG-HLA-A2 mice and induced GVHD by i.v. injection of A2neg human peripheral blood mononuclear cells (PBMCs). Because we could not inject human donors with EPS, we transferred EPS-pretreated dendritic cells (DCs) to inhibit aGVHD. We derived these DCs from CD34+ human cord blood cells, treated them with EPS, and then injected them together with PBMCs into the NSG-HLA-A2 mice. We found that all mice that received untreated DCs were dead by day 35, whereas 25% of mice receiving EPS-treated DCs (EPS-DCs) survived. This DC cell therapy could be readily translatable to humans, because we can generate large numbers of human EPS-DCs and use them as an "off the shelf" treatment for patients undergoing HSCT.


Assuntos
Doença Enxerto-Hospedeiro , Antígeno HLA-A2 , Animais , Humanos , Transplante Homólogo/efeitos adversos , Leucócitos Mononucleares , Doença Enxerto-Hospedeiro/prevenção & controle , Modelos Animais de Doenças , Células Dendríticas
18.
Blood ; 117(14): 3770-9, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21285437

RESUMO

B lymphopoiesis arrests in rabbits by 4 months of age. To identify molecules that contribute to this arrest, cDNA-representational difference analysis on BM stromal cells from young and adult rabbits showed that expression of Postn that encodes for the extracellular matrix protein periostin dramatically reduced with age. Postn-small interfering RNA OP9 cells lost their capacity to support B-cell development from rabbit or murine BM cells, and reexpression of periostin restored this potential, indicating an in vitro requirement for periostin in B lymphopoiesis. In our system, we determined that periostin deficiency leads to increased cell death and decreased proliferation of B-lineage progenitors. Further, RGD peptide inhibition of periostin/α(v)ß(3) interaction resulted in a marked decrease in B lymphopoiesis in vitro. Microarray analysis of the Postn-small interfering RNA OP9 cells showed decreased expression of key B-lymphopoietic factors, including IL-7 and CXCL12. In vivo, unidentified molecule(s) probably compensate periostin loss because Postn(-/-) mice had normal numbers of B-cell progenitors in BM. We conclude that the decline in periostin expression in adult rabbit BM does not solely explain the arrest of B lymphopoiesis. However, the interaction of periostin with α(v)ß(3) on lymphoid progenitors probably provides both proliferative and survival signals for cells in the B-cell development pathway.


Assuntos
Linfócitos B/fisiologia , Moléculas de Adesão Celular/fisiologia , Linfopoese/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Linfopoese/efeitos dos fármacos , Linfopoese/fisiologia , Camundongos , Camundongos Knockout , Células Precursoras de Linfócitos B/efeitos dos fármacos , Células Precursoras de Linfócitos B/metabolismo , Células Precursoras de Linfócitos B/fisiologia , RNA Interferente Pequeno/farmacologia , Coelhos
19.
J Immunol ; 186(11): 6437-44, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21525392

RESUMO

The peripheral B cell compartment in mice and humans is maintained by continuous production of transitional B cells in the bone marrow. In other species, however, including rabbits, B lymphopoiesis in the bone marrow abates early in life, and it is unclear how the peripheral B cell compartment is maintained. We identified transitional B cells in rabbits and classified them into T1 (CD24(high)CD21(low)) and T2 (CD24(high)CD21(+)) B cell subsets. By neutralizing B cell-activating factor in vivo, we found an arrest in peripheral B cell development at the T1 B cell stage. Surprisingly, T1 B cells were present in GALT, blood, and spleen of adult rabbits, long after B lymphopoiesis was arrested. T1 B cells were distinct from their counterparts in other species because they are proliferating and the Ig genes are somatically diversified. We designate these newly described cells as T1d B cells and propose a model in which they develop in GALT, self renew, continuously differentiate into mature B cells, and thereby maintain peripheral B cell homeostasis in adults in the absence of B lymphopoiesis.


Assuntos
Linfócitos B/imunologia , Proliferação de Células , Homeostase/imunologia , Células Precursoras de Linfócitos B/imunologia , Animais , Fator Ativador de Células B/imunologia , Fator Ativador de Células B/metabolismo , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/metabolismo , Sequência de Bases , Antígeno CD24/imunologia , Antígeno CD24/metabolismo , Diferenciação Celular/imunologia , Citometria de Fluxo , Trato Gastrointestinal/citologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Humanos , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Camundongos , Dados de Sequência Molecular , Células Precursoras de Linfócitos B/metabolismo , Coelhos , Receptores de Complemento 3d/imunologia , Receptores de Complemento 3d/metabolismo , Homologia de Sequência do Ácido Nucleico , Fatores de Tempo
20.
J Immunol ; 187(9): 4714-20, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21957145

RESUMO

Pre-BCR signaling is a critical checkpoint in B cell development in which B-lineage cells expressing functional IgH µ-chain are selectively expanded. B cell development is delayed in mutant ali/ali rabbits because the a-allotype encoding V(H)1 gene, which is normally used in VDJ gene rearrangements in wt rabbits, is deleted, and instead, most B-lineage cells use the a-allotype encoding V(H)4 gene [V(H)4(a)], which results in a severe developmental block at the pre-B cell stage. We found that V(H)4(a)-utilizing pre-B cells exhibit reduced pre-BCR signaling and do not undergo normal expansion in vitro. Transduction of murine 38B9 pre-B cells with chimeric rabbit-VDJ mouse-Cµ encoding retroviruses showed V(H)4(a)-encoded µ-chains do not readily form signal-competent pre-BCR, thereby explaining the reduction in pre-BCR signaling and pre-B cell expansion. Development of V(H)4(a)-utilizing B cells can be rescued in vivo by the expression of an Igκ transgene, indicating that V(H)4(a)-µ chains are not defective for conventional BCR formation and signaling. The ali/ali rabbit model system is unique because V(H)4(a)-µ chains have the capacity to pair with a variety of conventional IgL chains and yet lack the capacity to form a signal-competent pre-BCR. This system could allow for identification of critical structural parameters that govern pre-BCR formation/signaling.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/patologia , Cadeias Pesadas de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/metabolismo , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/patologia , Animais , Animais Geneticamente Modificados , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Morte Celular/genética , Morte Celular/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Técnicas de Cocultura , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Camundongos , Receptores de Células Precursoras de Linfócitos B/biossíntese , Receptores de Células Precursoras de Linfócitos B/genética , Receptores de Células Precursoras de Linfócitos B/fisiologia , Coelhos , Transdução de Sinais/genética , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA