Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 221(8)2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38819409

RESUMO

Th17 cell plasticity is crucial for development of autoinflammatory disease pathology. Periodontitis is a prevalent inflammatory disease where Th17 cells mediate key pathological roles, yet whether they exhibit any functional plasticity remains unexplored. We found that during periodontitis, gingival IL-17 fate-mapped T cells still predominantly produce IL-17A, with little diversification of cytokine production. However, plasticity of IL-17 fate-mapped cells did occur during periodontitis, but in the gingiva draining lymph node. Here, some Th17 cells acquired features of Tfh cells, a functional plasticity that was dependent on IL-6. Notably, Th17-to-Tfh diversification was important to limit periodontitis pathology. Preventing Th17-to-Tfh plasticity resulted in elevated periodontal bone loss that was not simply due to increased proportions of conventional Th17 cells. Instead, loss of Th17-to-Tfh cells resulted in reduced IgG levels within the oral cavity and a failure to restrict the biomass of the oral commensal community. Thus, our data identify a novel protective function for a subset of otherwise pathogenic Th17 cells during periodontitis.


Assuntos
Plasticidade Celular , Interleucina-17 , Periodontite , Células Th17 , Células Th17/imunologia , Animais , Periodontite/imunologia , Periodontite/patologia , Plasticidade Celular/imunologia , Interleucina-17/metabolismo , Interleucina-17/imunologia , Camundongos , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Células T Auxiliares Foliculares/imunologia , Gengiva/imunologia , Gengiva/patologia , Imunoglobulina G/imunologia , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/patologia
2.
Nat Commun ; 15(1): 683, 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38267402

RESUMO

Immune cell dysfunction within the tumor microenvironment (TME) undermines the control of cancer progression. Established tumors contain phenotypically distinct, tumor-specific natural killer (NK) cells; however, the temporal dynamics, mechanistic underpinning and functional significance of the NK cell compartment remains incompletely understood. Here, we use photo-labeling, combined with longitudinal transcriptomic and cellular analyses, to interrogate the fate of intratumoral NK cells. We reveal that NK cells rapidly lose effector functions and adopt a distinct phenotypic state with features associated with tissue residency. NK cell depletion from established tumors did not alter tumor growth, indicating that intratumoral NK cells cease to actively contribute to anti-tumor responses. IL-15 administration prevented loss of function and improved tumor control, generating intratumoral NK cells with both tissue-residency characteristics and enhanced effector function. Collectively, our data reveals the fate of NK cells after recruitment into tumors and provides insight into how their function may be revived.


Assuntos
Internato e Residência , Neoplasias , Humanos , Perfilação da Expressão Gênica , Células Matadoras Naturais , Transcriptoma , Microambiente Tumoral
3.
Cell Rep ; 42(11): 113425, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37950867

RESUMO

Innate lymphoid cells (ILCs) are tissue-resident effector cells with roles in tissue homeostasis, protective immunity, and inflammatory disease. Group 3 ILCs (ILC3s) are classically defined by the master transcription factor RORγt. However, ILC3 can be further subdivided into subsets that share type 3 effector modules that exhibit significant ontological, transcriptional, phenotypic, and functional heterogeneity. Notably lymphoid tissue inducer (LTi)-like ILC3s mediate effector functions not typically associated with other RORγt-expressing lymphocytes, suggesting that additional transcription factors contribute to dictate ILC3 subset phenotypes. Here, we identify Bcl6 as a subset-defining transcription factor of LTi-like ILC3s in mice and humans. Deletion of Bcl6 results in dysregulation of the LTi-like ILC3 transcriptional program and markedly enhances expression of interleukin-17A (IL-17A) and IL-17F in LTi-like ILC3s in a manner in part dependent upon the commensal microbiota-and associated with worsened inflammation in a model of colitis. Together, these findings redefine our understanding of ILC3 subset biology.


Assuntos
Linfócitos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Animais , Humanos , Camundongos , Imunidade Inata , Linfócitos/metabolismo , Tecido Linfoide/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Fatores de Transcrição/metabolismo
4.
Mucosal Immunol ; 16(5): 658-670, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37453568

RESUMO

Group 3 innate lymphoid cells (ILC3) are potent effector cells with critical roles in enforcing immunity, barrier integrity and tissue homeostasis along the gastrointestinal tract. ILC3 are considered primarily tissue-resident cells, seeding the gastrointestinal tract during embryonic stages and early life. However, the mechanisms through which mature ILC3 are maintained within adult tissues are poorly understood. Here, we report that lymphoid tissue-inducer-like (LTi-like) ILC3 exhibit minimal turnover in the healthy adult intestinal tract, persist for extended periods of time, and display a quiescent phenotype. Strikingly, during enteric bacterial infection LTi-like ILC3 also exhibit negligible hematopoietic replenishment and remain non-proliferative, despite robustly producing cytokines. Survival of LTi-like ILC3 was found to be dependent upon the balance between the metabolic activity required to drive effector function and anti-apoptotic programs. Notably, the pro-survival protein B-cell lymphoma-2 (Bcl-2) was required for the survival of LTi-like ILC3 ex vivo but was rendered partially dispensable if mitochondrial respiration was inhibited. Together we demonstrate LTi-like ILC3 are a tissue-resident, quiescent population that persist independently of hematopoietic replenishment to survive within the intestinal microenvironment.


Assuntos
Imunidade Inata , Linfócitos , Tecido Linfoide/metabolismo , Citocinas/metabolismo , Fenótipo
5.
Immunity ; 56(5): 1064-1081.e10, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-36948193

RESUMO

The recent revolution in tissue-resident macrophage biology has resulted largely from murine studies performed in C57BL/6 mice. Here, using both C57BL/6 and BALB/c mice, we analyze immune cells in the pleural cavity. Unlike C57BL/6 mice, naive tissue-resident large-cavity macrophages (LCMs) of BALB/c mice failed to fully implement the tissue-residency program. Following infection with a pleural-dwelling nematode, these pre-existing differences were accentuated with LCM expansion occurring in C57BL/6, but not in BALB/c mice. While infection drove monocyte recruitment in both strains, only in C57BL/6 mice were monocytes able to efficiently integrate into the resident pool. Monocyte-to-macrophage conversion required both T cells and interleukin-4 receptor alpha (IL-4Rα) signaling. The transition to tissue residency altered macrophage function, and GATA6+ tissue-resident macrophages were required for host resistance to nematode infection. Therefore, during tissue nematode infection, T helper 2 (Th2) cells control the differentiation pathway of resident macrophages, which determines infection outcome.


Assuntos
Filariose , Filarioidea , Infecções por Nematoides , Camundongos , Animais , Filarioidea/fisiologia , Células Th2 , Monócitos , Cavidade Pleural , Camundongos Endogâmicos C57BL , Macrófagos/fisiologia , Diferenciação Celular , Camundongos Endogâmicos BALB C
6.
J Exp Med ; 219(6)2022 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35472220

RESUMO

Improving the efficacy of immune checkpoint therapies will require a better understanding of how immune cells are recruited and sustained in tumors. Here, we used the photoconversion of the tumor immune cell compartment to identify newly entering lymphocytes, determine how they change over time, and investigate their egress from the tumor. Combining single-cell transcriptomics and flow cytometry, we found that while a diverse mix of CD8 T cell subsets enter the tumor, all CD8 T cells retained within this environment for more than 72 h developed an exhausted phenotype, revealing the rapid establishment of this program. Rather than forming tumor-resident populations, non-effector subsets, which express TCF-1 and include memory and stem-like cells, were continuously recruited into the tumor, but this recruitment was balanced by concurrent egress to the tumor-draining lymph node. Thus, the TCF-1+ CD8 T cell niche in tumors is highly dynamic, with the circulation of cells between the tumor and peripheral lymphoid tissue to bridge systemic and intratumoral responses.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Humanos , Imunoterapia , Tecido Linfoide , Subpopulações de Linfócitos T
7.
Nat Rev Immunol ; 19(10): 599-613, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31350531

RESUMO

Innate lymphoid cells (ILCs) are enriched at barrier surfaces of the mammalian body where they rapidly respond to host, microbial or environmental stimuli to promote immunity or tissue homeostasis. Furthermore, ILCs are dysregulated in multiple human diseases. Over the past decade, substantial advances have been made in identifying the heterogeneity and functional diversity of ILCs, which have revealed striking similarities to T cell subsets. However, emerging evidence indicates that ILCs also have a complex role in directly influencing the adaptive immune response in the context of development, homeostasis, infection or inflammation. In turn, adaptive immunity reciprocally regulates ILCs, which indicates that these interactions are a crucial determinant of immune responses within tissues. Here, we summarize our current understanding of functional interactions between ILCs and the adaptive immune system, discuss limitations and future areas of investigation, and consider the potential for these interactions to be therapeutically harnessed to benefit human health.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Linfócitos/imunologia , Animais , Apresentação de Antígeno , Comunicação Celular , Citocinas/fisiologia , Humanos , Doenças Inflamatórias Intestinais/imunologia , Neoplasias/imunologia
8.
Nat Med ; 22(3): 319-23, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26878233

RESUMO

RAR-related orphan receptor-γt (ROR-γt) directs differentiation of proinflammatory T helper 17 (TH17) cells and is a potential therapeutic target in chronic autoimmune and inflammatory diseases. However, ROR-γt-dependent group 3 innate lymphoid cells ILC3s provide essential immunity and tissue protection in the intestine, suggesting that targeting ROR-γt could also result in impaired host defense after infection or enhanced tissue damage. Here, we demonstrate that transient chemical inhibition of ROR-γt in mice selectively reduces cytokine production from TH17 but not ILCs in the context of intestinal infection with Citrobacter rodentium, resulting in preserved innate immunity. Temporal deletion of Rorc (encoding ROR-γt) in mature ILCs also did not impair cytokine response in the steady state or during infection. Finally, pharmacologic inhibition of ROR-γt provided therapeutic benefit in mouse models of intestinal inflammation and reduced the frequency of TH17 cells but not ILCs isolated from primary intestinal samples of individuals with inflammatory bowel disease (IBD). Collectively, these results reveal differential requirements for ROR-γt in the maintenance of TH17 cell and ILC3 responses and suggest that transient inhibition of ROR-γt is a safe and effective therapeutic approach during intestinal inflammation.


Assuntos
Colite/imunologia , Colo/imunologia , Doença de Crohn/imunologia , Infecções por Enterobacteriaceae/imunologia , Imunidade Inata/imunologia , Linfócitos/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Células Th17/imunologia , Animais , Citrobacter rodentium , Colite/patologia , Colo/citologia , Colo/patologia , Doença de Crohn/patologia , Citometria de Fluxo , Humanos , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética
9.
Int Immunol ; 28(1): 43-52, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26451009

RESUMO

A delicate balance exists between the mammalian immune system and normally beneficial commensal bacteria that colonize the gastrointestinal tract, which is necessary to maintain tissue homeostasis. Dysregulation of these interactions between the host and commensal bacteria is causally associated with chronic inflammation and the development of cancer. In contrast, recent reports have highlighted that commensal bacteria also play an essential role in promoting anti-tumor immune responses in several contexts, highlighting a paradox whereby interactions between the host and commensal bacteria can influence both pro- and anti-tumor immunity. Given the critical roles for group 3 innate lymphoid cells (ILC3s) in regulating inflammation, tissue repair and host-microbe interactions in the intestine, here we discuss new evidence that ILC3s may profoundly influence the development, progression and control of tumors. In this review, we provide an overview of recent advances in understanding the impact of commensal bacteria on tumorigenesis, discuss recent findings identifying ILC3s as critical regulators of host-microbe interactions and highlight the emerging role of this immune cell population in cancer and their potential implication as a therapeutic target.


Assuntos
Imunoterapia , Inflamação/imunologia , Mucosa Intestinal/imunologia , Linfócitos/imunologia , Neoplasias/imunologia , Animais , Carcinogênese , Homeostase , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Mucosa Intestinal/microbiologia , Linfócitos/microbiologia , Simbiose
10.
J Immunol ; 194(4): 1555-64, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25589067

RESUMO

Immunomodulation is a common feature of chronic helminth infections and mainly attributed to the secretion of bioactive molecules, which target and modify host immune cells. In this study, we show that the helminth immunomodulator AvCystatin, a cysteine protease inhibitor, induces a novel regulatory macrophage (Mreg; AvCystatin-Mreg), which is sufficient to mitigate major parameters of allergic airway inflammation and colitis in mice. A single adoptive transfer of AvCystatin-Mreg before allergen challenge suppressed allergen-specific IgE levels, the influx of eosinophils into the airways, local and systemic Th2 cytokine levels, and mucus production in lung bronchioles of mice, whereas increasing local and systemic IL-10 production by CD4(+) T cells. Moreover, a single administration of AvCystatin-Mreg during experimentally induced colitis strikingly reduced intestinal pathology. Phenotyping of AvCystatin-Mreg revealed increased expression of a distinct group of genes including LIGHT, sphingosine kinase 1, CCL1, arginase-1, and costimulatory molecules, CD16/32, ICAM-1, as well as PD-L1 and PD-L2. In cocultures with dendritic cells and CD4(+) T cells, AvCystatin-Mreg strongly induced the production of IL-10 in a cell-contact-independent manner. Collectively, our data identify a specific suppressive macrophage population induced by a single parasite immunomodulator, which protects against mucosal inflammation.


Assuntos
Antígenos de Helmintos/imunologia , Linfócitos T CD4-Positivos/imunologia , Imunidade nas Mucosas/imunologia , Inflamação/prevenção & controle , Macrófagos/imunologia , Acanthocheilonema/imunologia , Transferência Adotiva , Animais , Antígenos de Helmintos/farmacologia , Colite/imunologia , Colite/prevenção & controle , Modelos Animais de Doenças , Feminino , Imunossupressores/imunologia , Imunossupressores/farmacologia , Inflamação/imunologia , Interleucina-10/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pneumonia/imunologia , Pneumonia/prevenção & controle
11.
Science ; 345(6196): 578-82, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25082704

RESUMO

The mammalian intestine is colonized by beneficial commensal bacteria and is a site of infection by pathogens, including helminth parasites. Helminths induce potent immunomodulatory effects, but whether these effects are mediated by direct regulation of host immunity or indirectly through eliciting changes in the microbiota is unknown. We tested this in the context of virus-helminth coinfection. Helminth coinfection resulted in impaired antiviral immunity and was associated with changes in the microbiota and STAT6-dependent helminth-induced alternative activation of macrophages. Notably, helminth-induced impairment of antiviral immunity was evident in germ-free mice, but neutralization of Ym1, a chitinase-like molecule that is associated with alternatively activated macrophages, could partially restore antiviral immunity. These data indicate that helminth-induced immunomodulation occurs independently of changes in the microbiota but is dependent on Ym1.


Assuntos
Infecções por Caliciviridae/imunologia , Coinfecção/imunologia , Gastroenterite/imunologia , Imunomodulação , Lectinas/imunologia , Microbiota/imunologia , Norovirus/imunologia , Trichinella/imunologia , Triquinelose/imunologia , beta-N-Acetil-Hexosaminidases/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Coinfecção/microbiologia , Coinfecção/parasitologia , Gastroenterite/virologia , Vida Livre de Germes , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/virologia , Ativação de Macrófagos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL
12.
Gut Microbes ; 3(5): 476-81, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22892692

RESUMO

Recently, we demonstrated a novel role for gastrointestinal mast cells (MCs) in the early events that lead to the generation of Th2 immunity to helminth infection. Mice lacking MCs (Kit(W) /Kit(W-v) and Kit(W-Sh)) showed a significant inhibition of Th2 cell priming following infection with the parasitic helminth Heligmosomoides polygyrus bakeri (Hp). We showed that MCs degranulate during the early stages of infection when the helminth larvae invade the small intestinal tissue. Furthermore, MC degranulation was required for the enhanced expression and production of the tissue-derived cytokines IL-25, IL-33 and TSLP, which are required for the optimal orchestration and priming of type 2 immunity. In this addendum we aim to address several questions raised by our findings - in particular, the mechanisms through which MCs may recognize helminth exposure in the early stages of infection and by which they may enhance expression of critical tissue cytokines thus, enabling Th2 priming. Furthermore, we will discuss these findings in the context of recently described novel innate immune cells, such as type 2 hematopoietic progenitors and type 2 innate lymphoid cells.


Assuntos
Mastócitos/imunologia , Nematospiroides dubius/imunologia , Células Th2/imunologia , Animais , Degranulação Celular , Citocinas/metabolismo , Camundongos , Modelos Biológicos
13.
PLoS Pathog ; 7(1): e1001248, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-21253577

RESUMO

Parasitic worms alter their host's immune system to diminish the inflammatory responses directed against them, using very efficient immunomodulating molecules. We have previously shown that the helminth immunomodulator cystatin (AvCystatin) profoundly reduces the progression of inflammatory diseases via modulation of macrophages. Here we elucidate the signaling events in macrophages triggered by AvCystatin. Labeled AvCystatin was predominantly taken up by macrophages and subsequently induced the phosphorylation of the mitogen-activated protein kinases (MAPK) ERK1/2 and p38. IL-10 expression induced by AvCystatin in macrophages was tyrosine kinase sensitive and dependent on activation of both MAP kinases, in clear contrast to expression of IL-12/23p40. In addition, phosphorylation of the transcription factors CREB and STAT3 was induced by AvCystatin and regulated by phospho-ERK. Chemical inhibition of phosphoinositide 3-kinase (PI3K) reduced AvCystatin-induced cytokine release; however, AKT, the downstream target of PI3K, was not activated following AvCystatin exposure. To characterize signaling elements involved in alteration of the macrophage phenotype we applied mathematical modeling. Experimental testing of the in silico generated hypotheses identified dual specificity phosphatase (DUSP) 1 and 2, as regulators in AvCystatin triggered macrophages in vitro and in vivo. In particular, DUSP1 was subsequently found to be responsible for regulation of ERK- and p38-phosphorylation and controlled the IL-10 expression in macrophages by AvCystatin. Thus, we show that AvCystatin exploits activation and deactivation pathways of MAP kinases to induce regulatory macrophages. This study provides insights into molecular mechanisms of macrophage manipulation by parasites and highlights the utility of mathematical modeling for the elucidation of regulatory circuits of immune cells.


Assuntos
Inibidores de Cisteína Proteinase/farmacologia , Interações Hospedeiro-Parasita/imunologia , Fatores Imunológicos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Animais , Células Cultivadas , Inibidores de Cisteína Proteinase/imunologia , Citocinas/genética , Citocinas/metabolismo , Regulação para Baixo , Feminino , Expressão Gênica , Inativação Gênica , Interações Hospedeiro-Parasita/genética , Fatores Imunológicos/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , RNA Mensageiro/metabolismo , Transdução de Sinais
14.
Eur J Immunol ; 40(2): 406-16, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19950176

RESUMO

Trichuris muris infection is an ideal model for defining T-cell-driven immunity, and also provides essential insights that may impact on potential helminth therapies currently in development. Conflicting host variables determine the efficiency of such treatments and we have identified host-derived sex steroid hormones as key factors in the development of immunity. The female-associated hormone 17-beta estradiol (E2) significantly enhanced the generation of a Th2 response in vitro; however, this stimulatory effect was found to be dispensable for the generation of immunity to Trichuris in the gender-biased IL-4KO mouse model. In contrast, the male-associated hormone dihydrotestosterone significantly inhibited the T-cell stimulatory capacity of DC and directly suppressed the immune response of male IL-4KO mice, with worm expulsion restored following castration. This finding was associated with dramatically reduced IL-18 mRNA expression suggesting androgens may act via this cytokine to suppress Th2 immunity to Trichuris. This study has critical implications for the development and efficacy of potential helminth therapeutics and identifies host gender - specifically sex hormones - as important factors in the development of Th2 immunity in susceptible and immunocompromised mice.


Assuntos
Hormônios Esteroides Gonadais/metabolismo , Células Th2/imunologia , Tricuríase/imunologia , Trichuris/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Di-Hidrotestosterona/metabolismo , Di-Hidrotestosterona/farmacologia , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Expressão Gênica , Hormônios Esteroides Gonadais/farmacologia , Hormônios Esteroides Gonadais/fisiologia , Interleucina-18/genética , Interleucina-18/fisiologia , Interleucina-4/genética , Interleucina-4/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , Orquiectomia , Ovariectomia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Células Th2/citologia , Células Th2/efeitos dos fármacos , Tricuríase/metabolismo , Tricuríase/parasitologia , Trichuris/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA