Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Cell ; 186(9): 1846-1862.e26, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37028428

RESUMO

The use of probiotics by cancer patients is increasing, including among those undergoing immune checkpoint inhibitor (ICI) treatment. Here, we elucidate a critical microbial-host crosstalk between probiotic-released aryl hydrocarbon receptor (AhR) agonist indole-3-aldehyde (I3A) and CD8 T cells within the tumor microenvironment that potently enhances antitumor immunity and facilitates ICI in preclinical melanoma. Our study reveals that probiotic Lactobacillus reuteri (Lr) translocates to, colonizes, and persists within melanoma, where via its released dietary tryptophan catabolite I3A, it locally promotes interferon-γ-producing CD8 T cells, thereby bolstering ICI. Moreover, Lr-secreted I3A was both necessary and sufficient to drive antitumor immunity, and loss of AhR signaling within CD8 T cells abrogated Lr's antitumor effects. Further, a tryptophan-enriched diet potentiated both Lr- and ICI-induced antitumor immunity, dependent on CD8 T cell AhR signaling. Finally, we provide evidence for a potential role of I3A in promoting ICI efficacy and survival in advanced melanoma patients.


Assuntos
Limosilactobacillus reuteri , Melanoma , Microambiente Tumoral , Humanos , Dieta , Inibidores de Checkpoint Imunológico , Limosilactobacillus reuteri/metabolismo , Melanoma/terapia , Triptofano/metabolismo , Linfócitos T CD8-Positivos/imunologia , Receptores de Hidrocarboneto Arílico/agonistas
2.
Nat Immunol ; 18(9): 995-1003, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28759002

RESUMO

Among the features that distinguish type 1 innate lymphoid cells (ILC1s) from natural killer (NK) cells is a gene signature indicative of 'imprinting' by cytokines of the TGF-ß family. We studied mice in which ILC1s and NK cells lacked SMAD4, a signal transducer that facilitates the canonical signaling pathway common to all cytokines of the TGF-ß family. While SMAD4 deficiency did not affect ILC1 differentiation, NK cells unexpectedly acquired an ILC1-like gene signature and were unable to control tumor metastasis or viral infection. Mechanistically, SMAD4 restrained non-canonical TGF-ß signaling mediated by the cytokine receptor TGFßR1 in NK cells. NK cells from a SMAD4-deficient person affected by polyposis were also hyper-responsive to TGF-ß. These results identify SMAD4 as a previously unknown regulator that restricts non-canonical TGF-ß signaling in NK cells.


Assuntos
Células Matadoras Naturais/citologia , Linfopoese/genética , Proteína Smad4/genética , Fator de Crescimento Transformador beta/imunologia , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/imunologia , Animais , Estudos de Casos e Controles , Diferenciação Celular , Perfilação da Expressão Gênica , Humanos , Imunidade Inata/imunologia , Immunoblotting , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Linfócitos/citologia , Melanoma Experimental/imunologia , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Proteína Smad4/imunologia
3.
J Immunol ; 212(3): 369-374, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38117750

RESUMO

NKp44 is a human receptor originally found on activated NK cells, group 1 and group 3 innate lymphoid cells that binds dimers of platelet-derived growth factor D (PDGF-DD). NKp44 is also expressed on tissue plasmacytoid dendritic cells (PDCs), but NKp44-PDGF-DD interaction on PDCs remains unstudied. Engagement of NKp44 with PDGF-DD in vitro enhanced PDC secretion of IFN-α, TNF, and IL-6 in response to the TLR9 ligand CpG-ODN, but not TLR7/8 ligands. In tissues, PDCs were found in close contact with PDGF-DD-expressing cells in the high endothelial venules and epithelium of tonsils, melanomas, and skin lesions infected with Molluscum contagiosum. Recombinant PDGF-DD enhanced the serum IFN-α response to systemic HSV-1 infection in a humanized mouse model. We conclude that NKp44 integrates with TLR9 signaling to enhance PDC cytokine production. These findings may have bearings for immune responses to TLR9-based adjuvants, therapy for tumors expressing PDGF-DD, and infections with DNA viruses that induce PDGF-DD expression to enhance viral spread.


Assuntos
Imunidade Inata , Receptor Toll-Like 9 , Animais , Camundongos , Humanos , Receptor Toll-Like 9/metabolismo , Interferon-alfa/metabolismo , Células Dendríticas , Células Matadoras Naturais
4.
Trends Immunol ; 43(11): 858-860, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36243620

RESUMO

CD4+ intraepithelial lymphocytes (CD4IEL) are tissue-resident T cells with cytotoxic and regulatory properties; together with peripheral regulatory T cells, they control intestinal inflammation. Recently, Bousbaine and colleagues identified a microbiota-derived conserved antigen that induces CD4IEL differentiation and promotes their regulatory function, attenuating the severity of murine colitis.


Assuntos
Linfócitos Intraepiteliais , Microbiota , Humanos , Camundongos , Animais , Linfócitos T CD4-Positivos , Linfócitos T Reguladores , Antígenos , Mucosa Intestinal
5.
Immunity ; 44(5): 1127-39, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27156386

RESUMO

The signals guiding differentiation of innate lymphoid cells (ILCs) within tissues are not well understood. Salivary gland (SG) ILCs as well as liver and intestinal intraepithelial ILC1 have markers that denote tissue residency and transforming growth factor-ß (TGF-ß) imprinting. We deleted Tgfbr2 in cells expressing the ILC and NK marker NKp46 and found that SG ILCs were reduced in number. They lost distinct tissue markers, such as CD49a, and the effector molecules TRAIL and CD73. Expression of the transcription factor Eomes, which promotes NK cell differentiation, was elevated. Conversely, Eomes deletion in NKp46(+) cells enhanced TGF-ß-imprinting of SG ILCs. Thus, TGF-ß induces SG ILC differentiation by suppressing Eomes. TGF-ß acted through a JNK-dependent, Smad4-independent pathway. Transcriptome analysis demonstrated that SG ILCs had characteristic of both NK cells and ILC1. Finally, TGF-ß imprinting of SG ILCs was synchronized with SG development, highlighting the impact of tissue microenvironment on ILC development.


Assuntos
Diferenciação Celular , Células Matadoras Naturais/fisiologia , Linfócitos/fisiologia , Glândulas Salivares/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos Ly/metabolismo , Microambiente Celular , Perfilação da Expressão Gênica , Imunidade Inata , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Knockout , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad4/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34462359

RESUMO

Oral infection with Toxoplasma gondii results in dysbiosis and enteritis, both of which revert to normal during chronic infection. However, whether infection leaves a lasting impact on mucosal responses remains uncertain. Here we examined the effect of the chemical irritant dextran sodium sulfate (DSS) on intestinal damage and wound healing in chronically infected mice. Our findings indicate that prior infection with T. gondii exacerbates damage to the colon caused by DSS and impairs wound healing by suppressing stem cell regeneration of the epithelium. Enhanced tissue damage was attributable to inflammatory monocytes that emerge preactivated from bone marrow, migrate to the intestine, and release inflammatory mediators, including nitric oxide. Tissue damage was reversed by neutralization of inflammatory monocytes or nitric oxide, revealing a causal mechanism for tissue damage. Our findings suggest that chronic infection with T. gondii enhances monocyte activation to increase inflammation associated with a secondary environmental insult.


Assuntos
Colite/complicações , Toxoplasmose/complicações , Animais , Doença Crônica , Suscetibilidade a Doenças , Microbioma Gastrointestinal , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/patologia , Regeneração , Células-Tronco/patologia
7.
Nat Immunol ; 12(2): 137-43, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21217758

RESUMO

The 5' cap structures of higher eukaryote mRNAs have ribose 2'-O-methylation. Likewise, many viruses that replicate in the cytoplasm of eukaryotes have evolved 2'-O-methyltransferases to autonomously modify their mRNAs. However, a defined biological role for 2'-O-methylation of mRNA remains elusive. Here we show that 2'-O-methylation of viral mRNA was critically involved in subverting the induction of type I interferon. We demonstrate that human and mouse coronavirus mutants lacking 2'-O-methyltransferase activity induced higher expression of type I interferon and were highly sensitive to type I interferon. Notably, the induction of type I interferon by viruses deficient in 2'-O-methyltransferase was dependent on the cytoplasmic RNA sensor Mda5. This link between Mda5-mediated sensing of viral RNA and 2'-O-methylation of mRNA suggests that RNA modifications such as 2'-O-methylation provide a molecular signature for the discrimination of self and non-self mRNA.


Assuntos
Infecções por Coronavirus/metabolismo , Coronavirus/fisiologia , RNA Helicases DEAD-box/metabolismo , Metiltransferases/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Coronavirus/patogenicidade , Infecções por Coronavirus/genética , Infecções por Coronavirus/imunologia , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , Humanos , Imunidade Inata/genética , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon , Metilação , Metiltransferases/genética , Metiltransferases/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Viral/metabolismo , Receptor de Interferon alfa e beta/genética , Receptores de Reconhecimento de Padrão/genética , Ribose/metabolismo , Proteínas Virais/genética , Proteínas Virais/imunologia , Virulência/genética , Replicação Viral/genética
8.
J Immunol ; 204(8): 2257-2268, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32169845

RESUMO

Plasmacytoid dendritic cells (pDCs) produce abundant type I IFNs (IFN-I) in response to viral nucleic acids. Generation of pDCs from bone marrow dendritic cell (DC) progenitors and their maintenance is driven by the transcription factor E2-2 and inhibited by its repressor Id2. In this study, we find that mouse pDCs selectively express the receptor for LIF that signals through STAT3. Stimulation of pDCs with LIF inhibited IFN-I, TNF, and IL-6 responses to CpG and induced expression of the STAT3 targets SOCS3 and Bcl3, which inhibit IFN-I and NF-κB signaling. Moreover, although STAT3 has been also reported to induce E2-2, LIF paradoxically induced its repressor Id2. A late-stage bone marrow DC progenitor expressed low amounts of LIFR and developed into pDCs less efficiently after being exposed to LIF, consistent with the induction of Id2. Conversely, pDC development and serum IFN-I responses to lymphocytic choriomeningitis virus infection were augmented in newly generated mice lacking LIFR in either CD11c+ or hematopoietic cells. Thus, an LIF-driven STAT3 pathway induces SOCS3, Bcl3, and Id2, which render pDCs and late DC progenitors refractory to physiological stimuli controlling pDC functions and development. This pathway can be potentially exploited to prevent inappropriate secretion of IFN-I in autoimmune diseases or promote IFN-I secretion during viral infections.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Fator Inibidor de Leucemia/metabolismo , Animais , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Transcrição STAT3/imunologia , Transdução de Sinais/imunologia
9.
PLoS Pathog ; 13(2): e1006195, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28158275

RESUMO

Coronaviruses are of veterinary and medical importance and include highly pathogenic zoonotic viruses, such as SARS-CoV and MERS-CoV. They are known to efficiently evade early innate immune responses, manifesting in almost negligible expression of type-I interferons (IFN-I). This evasion strategy suggests an evolutionary conserved viral function that has evolved to prevent RNA-based sensing of infection in vertebrate hosts. Here we show that the coronavirus endonuclease (EndoU) activity is key to prevent early induction of double-stranded RNA (dsRNA) host cell responses. Replication of EndoU-deficient coronaviruses is greatly attenuated in vivo and severely restricted in primary cells even during the early phase of the infection. In macrophages we found immediate induction of IFN-I expression and RNase L-mediated breakdown of ribosomal RNA. Accordingly, EndoU-deficient viruses can retain replication only in cells that are deficient in IFN-I expression or sensing, and in cells lacking both RNase L and PKR. Collectively our results demonstrate that the coronavirus EndoU efficiently prevents simultaneous activation of host cell dsRNA sensors, such as Mda5, OAS and PKR. The localization of the EndoU activity at the site of viral RNA synthesis-within the replicase complex-suggests that coronaviruses have evolved a viral RNA decay pathway to evade early innate and intrinsic antiviral host cell responses.


Assuntos
Coronaviridae/enzimologia , Infecções por Coronavirus/imunologia , Endonucleases/imunologia , Evasão da Resposta Imune/fisiologia , Proteínas Virais/imunologia , Animais , Coronaviridae/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
11.
Immunity ; 29(6): 986-97, 2008 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-19062318

RESUMO

Dendritic cells are critically involved in the promotion and regulation of T cell responses. Here, we report a mouse strain that lacks conventional CD11c(hi) dendritic cells (cDCs) because of constitutive cell-type specific expression of a suicide gene. As expected, cDC-less mice failed to mount effective T cell responses resulting in impaired viral clearance. In contrast, neither thymic negative selection nor T regulatory cell generation or T cell homeostasis were markedly affected. Unexpectedly, cDC-less mice developed a progressive myeloproliferative disorder characterized by prominent extramedullary hematopoiesis and increased serum amounts of the cytokine Flt3 ligand. Our data identify a critical role of cDCs in the control of steady-state hematopoiesis, revealing a feedback loop that links peripheral cDCs to myelogenesis through soluble growth factors, such as Flt3 ligand.


Assuntos
Células Dendríticas/imunologia , Proteínas de Membrana/imunologia , Transtornos Mieloproliferativos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proliferação de Células , Células Dendríticas/metabolismo , Homeostase/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos Mieloproliferativos/metabolismo , Síndrome , Linfócitos T Reguladores/metabolismo
12.
J Immunol ; 192(11): 5192-200, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24778443

RESUMO

Abs play a significant role in protection against the intracellular bacterium Salmonella Typhi. In this article, we investigated how long-term protective IgM responses can be elicited by a S. Typhi outer-membrane protein C- and F-based subunit vaccine (porins). We found that repeated Ag exposure promoted a CD4(+) T cell-dependent germinal center reaction that generated mutated IgM-producing B cells and was accompanied by a strong expansion of IFN-γ-secreting T follicular helper cells. Genetic ablation of individual cytokine receptors revealed that both IFN-γ and IL-17 are required for optimal germinal center reactions and production of porin-specific memory IgM(+) B cells. However, more profound reduction of porin-specific IgM B cell responses in the absence of IFN-γR signaling indicated that this cytokine plays a dominant role. Importantly, mutated IgM mAbs against porins exhibited bactericidal capacity and efficiently augmented S. Typhi clearance. In conclusion, repeated vaccination with S. Typhi porins programs type I T follicular helper cell responses that contribute to the diversification of B cell memory and promote the generation of protective IgM Abs.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Centro Germinativo/imunologia , Imunoglobulina M/imunologia , Memória Imunológica , Interferon gama/imunologia , Salmonella typhi/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Feminino , Centro Germinativo/patologia , Humanos , Interferon gama/genética , Masculino , Camundongos , Camundongos Knockout , Vacinas contra Salmonella/genética , Vacinas contra Salmonella/imunologia , Febre Tifoide/genética , Febre Tifoide/imunologia , Febre Tifoide/patologia , Febre Tifoide/prevenção & controle
14.
Proc Natl Acad Sci U S A ; 109(8): 3012-7, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22315415

RESUMO

Infections with persistent viruses are a frequent cause of immunosuppression, autoimmune sequelae, and/or neoplastic disease. Plasmacytoid dendritic cells (pDCs) are innate immune cells that produce type I interferon (IFN-I) and other cytokines in response to virus-derived nucleic acids. Persistent viruses often cause depletion or functional impairment of pDCs, but the role of pDCs in the control of these viruses remains unclear. We used conditional targeting of pDC-specific transcription factor E2-2 to generate mice that constitutively lack pDCs in peripheral lymphoid organs and tissues. The profound impact of pDC deficiency on innate antiviral responses was revealed by the failure to control acute infection with the cytopathic mouse hepatitis virus. Furthermore, pDC-deficient animals failed to clear lymphocytic choriomeningitis virus (LCMV) from hematopoietic organs during persistent LCMV infection. This failure was associated with reduced numbers and functionality of LCMV-specific CD4(+) helper T cells and impaired antiviral CD8(+) T-cell responses. Adoptive transfer of LCMV-specific T cells revealed that both CD4(+) and CD8(+) T cells required IFN-I for expansion, but only CD4(+) T cells required the presence of pDCs. In contrast, mice with pDC-specific loss of MHC class II expression supported normal CD4(+) T-cell response to LCMV. These data suggest that pDCs facilitate CD4(+) helper T-cell responses to persistent viruses independently of direct antigen presentation. Thus pDCs provide an essential link between innate and adaptive immunity to chronic viral infection, likely through the secretion of IFN-I and other cytokines.


Assuntos
Células Dendríticas/imunologia , Linfócitos T/imunologia , Viroses/imunologia , Viroses/virologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Doença Crônica , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Marcação de Genes , Coriomeningite Linfocítica/sangue , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/prevenção & controle , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/imunologia , Fator de Transcrição 4
15.
Proc Natl Acad Sci U S A ; 109(4): 1233-8, 2012 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-22232667

RESUMO

Generation of antiviral IgM is usually considered as a marker of a short-lived initial antibody response that is replaced by hypermutated and more-efficient IgG. However, once viruses have established a particular niche for their persistence (e.g., within the CNS), the immune system has to specifically mobilize a broad range of antimicrobial effectors to contain the pathogen in the long term. Infection of the CNS with the mouse hepatitis virus (MHV) provides a unique model situation in which the extent of inflammatory CNS disease is determined by the balance between antiviral immune control, viral replication, and immune-mediated damage. We show here that whereas antibody- or B cell-deficient mice failed to contain MHV CNS infection and developed progressive demyelinating disease, germline IgM produced in activation-induced cytidine deaminase-deficient mice (aicda(-/-)) provided long-term protection against the chronic multiple sclerosis-like disease. Furthermore, we found that appropriate B-cell activation within the CNS-draining lymph node and subsequent CXCR3-mediated migration of antiviral IgM-secreting cells to the infected CNS was dependent on CD40-mediated interaction of B cells with T helper cells. These data indicate that the CD40-mediated collaboration of T and B cells is critical to secure neuroprotective IgM responses during viral CNS infection.


Assuntos
Antígenos CD40/imunologia , Sistema Nervoso Central/virologia , Doenças Desmielinizantes/virologia , Imunoglobulina M/imunologia , Vírus da Hepatite Murina/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Análise de Variância , Animais , Linfócitos B/imunologia , Sistema Nervoso Central/imunologia , Citidina Desaminase/deficiência , Doenças Desmielinizantes/imunologia , ELISPOT , Citometria de Fluxo , Imunofluorescência , Imunoglobulina M/genética , Camundongos , Camundongos Knockout
16.
J Immunol ; 188(8): 3678-85, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22407917

RESUMO

Regulatory T cells (Tregs) are important for the attenuation of immune reactions. During viral CNS infections, however, an indiscriminate maintenance of CNS immune privilege through Treg-mediated negative regulation could prevent autoimmune sequelae but impair the control of viral replication. We analyzed in this study the impact of Tregs on the development of acute viral encephalomyelitis, T cell-mediated antiviral protection, and prevention of CNS autoimmunity following intranasal infection with the gliatropic mouse hepatitis virus strain A59. To assess the contribution of Tregs in vivo, we specifically depleted CD4(+)Foxp3(+) T cells in a diphtheria toxin-dependent manner. We found that depletion of Tregs had no impact on viral distribution and clearance and did not significantly alter virus-specific CD4(+) and CD8(+) T cell responses. However, Treg depletion led to a more severe CNS inflammation associated with neuronal damage. Dissection of the underlying immunopathological mechanisms revealed the elaborate Treg-dependent regulation of self-reactive CD4(+) T cell proliferation within the CNS-draining lymph node and downtuning of CXCR3 expression on T cells. Taken together, these results suggest that Tregs preserve CNS immune privilege through selective control of CNS-specific Th cells while keeping protective antiviral immunity fully operative.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Infecções do Sistema Nervoso Central/imunologia , Infecções por Coronavirus/imunologia , Encefalomielite/imunologia , Vírus da Hepatite Murina/imunologia , Linfócitos T Reguladores/imunologia , Doença Aguda , Administração Intranasal , Animais , Autoantígenos/imunologia , Autoimunidade , Antígenos CD4/imunologia , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Infecções do Sistema Nervoso Central/virologia , Infecções por Coronavirus/virologia , Encefalomielite/virologia , Fatores de Transcrição Forkhead/imunologia , Humanos , Imunidade Celular , Linfonodos/imunologia , Linfonodos/patologia , Ativação Linfocitária , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Receptores CXCR3/imunologia , Linfócitos T Reguladores/patologia
17.
Eur J Immunol ; 42(12): 3116-25, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23255008

RESUMO

The immune system exhibits an enormous complexity. High throughput methods such as the "-omic'' technologies generate vast amounts of data that facilitate dissection of immunological processes at ever finer resolution. Using high-resolution data-driven systems analysis, causal relationships between complex molecular processes and particular immunological phenotypes can be constructed. However, processes in tissues, organs, and the organism itself (so-called higher level processes) also control and regulate the molecular (lower level) processes. Reverse systems engineering approaches, which focus on the examination of the structure, dynamics and control of the immune system, can help to understand the construction principles of the immune system. Such integrative mechanistic models can properly describe, explain, and predict the behavior of the immune system in health and disease by combining both higher and lower level processes. Moving from molecular and cellular levels to a multiscale systems understanding requires the development of methodologies that integrate data from different biological levels into multiscale mechanistic models. In particular, 3D imaging techniques and 4D modeling of the spatiotemporal dynamics of immune processes within lymphoid tissues are central for such integrative approaches. Both dynamic and global organ imaging technologies will be instrumental in facilitating comprehensive multiscale systems immunology analyses as discussed in this review.


Assuntos
Imageamento Tridimensional , Sistema Imunitário/fisiologia , Imunidade/fisiologia , Modelos Imunológicos , Biologia de Sistemas/métodos , Alergia e Imunologia , Animais , Humanos
18.
Cell Rep ; 42(10): 113140, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37768824

RESUMO

Dietary fiber strongly impacts the microbiota. Here, we show that a low-fiber diet changes the small intestinal (SI) microbiota and impairs SI Th17, TCRαß+CD8αß+ and TCRαß+CD8αα+ intraepithelial T cell development. We restore T cell development with dietary fiber supplementation, but this defect becomes persistent over generations with constant low-fiber diets. Offspring of low-fiber diet-fed mice have reduced SI T cells even after receiving a fiber-rich diet due to loss of bacteria important for T cell development. In these mice, only a microbiota transplant from a fiber-rich diet-fed mouse and a fiber-rich diet can restore T cell development. Low-fiber diets reduce segmented filamentous bacteria (SFB) abundance, impairing its vertical transmission. SFB colonization and a fiber-rich diet partially restore T cell development. Finally, we observe that low-fiber diet-induced T cell defects render mice more susceptible to Citrobacter rodentium infection. Together, these results demonstrate the importance of fiber to microbiota vertical transmission and host immune system development.


Assuntos
Microbioma Gastrointestinal , Linfócitos Intraepiteliais , Microbiota , Camundongos , Animais , Intestino Delgado/microbiologia , Receptores de Antígenos de Linfócitos T alfa-beta , Mucosa Intestinal/microbiologia , Fibras na Dieta , Camundongos Endogâmicos C57BL
19.
PLoS Pathog ; 6(7): e1001017, 2010 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-20661432

RESUMO

Plasmacytoid dendritic cell (pDC)-mediated protection against cytopathic virus infection involves various molecular, cellular, tissue-scale, and organism-scale events. In order to better understand such multiscale interactions, we have implemented a systems immunology approach focusing on the analysis of the structure, dynamics and operating principles of virus-host interactions which constrain the initial spread of the pathogen. Using high-resolution experimental data sets coming from the well-described mouse hepatitis virus (MHV) model, we first calibrated basic modules including MHV infection of its primary target cells, i.e. pDCs and macrophages (Mphis). These basic building blocks were used to generate and validate an integrative mathematical model for in vivo infection dynamics. Parameter estimation for the system indicated that on a per capita basis, one infected pDC secretes sufficient type I IFN to protect 10(3) to 10(4) Mphis from cytopathic viral infection. This extremely high protective capacity of pDCs secures the spleen's capability to function as a 'sink' for the virus produced in peripheral organs such as the liver. Furthermore, our results suggest that the pDC population in spleen ensures a robust protection against virus variants which substantially down-modulate IFN secretion. However, the ability of pDCs to protect against severe disease caused by virus variants exhibiting an enhanced liver tropism and higher replication rates appears to be rather limited. Taken together, this systems immunology analysis suggests that antiviral therapy against cytopathic viruses should primarily limit viral replication within peripheral target organs.


Assuntos
Alergia e Imunologia , Células Dendríticas/imunologia , Biologia de Sistemas/métodos , Viroses/imunologia , Antivirais , Sistemas de Liberação de Medicamentos , Interações Hospedeiro-Patógeno/imunologia , Fígado/virologia , Macrófagos/imunologia , Macrófagos/virologia , Modelos Biológicos , Modelos Teóricos , Viroses/tratamento farmacológico
20.
Cell Host Microbe ; 30(1): 10-12, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-35026131

RESUMO

In this issue of Cell Host & Microbe, Alexander et al. show that the enzyme cardiac glycoside reductase 2 (cgr2), which is produced by Eggerthella lenta, metabolizes RORγT inhibitors, resulting in an increased Th17 response and more severe inflammation in colitis models. The effect of cgr2 can be neutralized by a diet rich in arginine.


Assuntos
Colite , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Humanos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Oxirredutases , Células Th17
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA