Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Immunol ; 25(5): 820-833, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38600356

RESUMO

Human bone marrow permanently harbors high numbers of neutrophils, and a tumor-supportive bias of these cells could significantly impact bone marrow-confined malignancies. In individuals with multiple myeloma, the bone marrow is characterized by inflammatory stromal cells with the potential to influence neutrophils. We investigated myeloma-associated alterations in human marrow neutrophils and the impact of stromal inflammation on neutrophil function. Mature neutrophils in myeloma marrow are activated and tumor supportive and transcribe increased levels of IL1B and myeloma cell survival factor TNFSF13B (BAFF). Interactions with inflammatory stromal cells induce neutrophil activation, including BAFF secretion, in a STAT3-dependent manner, and once activated, neutrophils gain the ability to reciprocally induce stromal activation. After first-line myeloid-depleting antimyeloma treatment, human bone marrow retains residual stromal inflammation, and newly formed neutrophils are reactivated. Combined, we identify a neutrophil-stromal cell feed-forward loop driving tumor-supportive inflammation that persists after treatment and warrants novel strategies to target both stromal and immune microenvironments in multiple myeloma.


Assuntos
Fator Ativador de Células B , Interleucina-1beta , Mieloma Múltiplo , Neutrófilos , Células Estromais , Microambiente Tumoral , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Humanos , Microambiente Tumoral/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Células Estromais/metabolismo , Células Estromais/imunologia , Fator Ativador de Células B/metabolismo , Interleucina-1beta/metabolismo , Ativação de Neutrófilo , Fator de Transcrição STAT3/metabolismo , Medula Óssea/imunologia , Medula Óssea/patologia
2.
Nat Immunol ; 22(6): 769-780, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34017122

RESUMO

Progression and persistence of malignancies are influenced by the local tumor microenvironment, and future eradication of currently incurable tumors will, in part, hinge on our understanding of malignant cell biology in the context of their nourishing surroundings. Here, we generated paired single-cell transcriptomic datasets of tumor cells and the bone marrow immune and stromal microenvironment in multiple myeloma. These analyses identified myeloma-specific inflammatory mesenchymal stromal cells, which spatially colocalized with tumor cells and immune cells and transcribed genes involved in tumor survival and immune modulation. Inflammatory stromal cell signatures were driven by stimulation with proinflammatory cytokines, and analyses of immune cell subsets suggested interferon-responsive effector T cell and CD8+ stem cell memory T cell populations as potential sources of stromal cell-activating cytokines. Tracking stromal inflammation in individuals over time revealed that successful antitumor induction therapy is unable to revert bone marrow inflammation, predicting a role for mesenchymal stromal cells in disease persistence.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Células-Tronco Mesenquimais/imunologia , Mieloma Múltiplo/imunologia , Recidiva Local de Neoplasia/imunologia , Microambiente Tumoral/imunologia , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Medula Óssea/patologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Pessoa de Meia-Idade , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/prevenção & controle , Cultura Primária de Células , Estudos Prospectivos , RNA-Seq , Análise de Célula Única , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
3.
Mucosal Immunol ; 14(3): 717-727, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33414524

RESUMO

Innate lymphoid cells (ILCs) are enriched in mucosae and have been described as tissue-resident. Interestingly, ILCs are also present within lymph nodes (LNs), in the interfollicular regions, the destination for lymph-migratory cells. We have previously shown that LN ILCs are supplemented by peripheral tissue-derived ILCs. Using thoracic duct cannulations, we here enumerate the intestinal lymph ILCs that traffic from the intestine to the mesenteric LNs (MLNs). We provide, for the first time, a detailed characterisation of these lymph-migratory ILCs. We show that all ILC subsets migrate in lymph, and while global transcriptional analysis reveals a shared signature with tissue-resident ILCs, lymph ILCs express migration-associated genes including S1PRs, SELL (CD62L) and CCR7. Interestingly, we discovered that while Salmonella Typhimurium infections do not increase the numbers of migrating ILCs, infection changes their composition and cytokine profile. Infection increases the proportions of RORyt+ T-bet+ ILCs, levels of IFNγ, and IFNγ/GM-CSF co-expression. Infection-induced changes in migratory ILCs are reflected in colon-draining MLN ILCs, where RORyt+ T-bet+ ILCs accumulate and display corresponding increased cytokine expression. Thus, we reveal that ILCs respond rapidly to intestinal infection and can migrate to the MLN where they produce cytokines.


Assuntos
Mucosa Intestinal/imunologia , Linfonodos/imunologia , Linfa/imunologia , Linfócitos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/fisiologia , Animais , Movimento Celular , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Imunidade Inata , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética
4.
Eur J Immunol ; 51(1): 76-90, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32700362

RESUMO

Upon viral infection, stressed or damaged cells can release alarmins like IL-33 that act as endogenous danger signals alerting innate and adaptive immune cells. IL-33 coming from nonhematopoietic cells has been identified as important factor triggering the expansion of antiviral CD8+ T cells. In LN the critical cellular source of IL-33 is unknown, as is its potential cell-intrinsic function as a chromatin-associated factor. Using IL-33-GFP reporter mice, we identify fibroblastic reticular cells (FRC) and lymphatic endothelial cells (LEC) as the main IL-33 source. In homeostasis, IL-33 is dispensable as a transcriptional regulator in FRC, indicating it functions mainly as released cytokine. Early during infection with lymphocytic choriomeningitis virus (LCMV) clone 13, both FRC and LEC lose IL-33 protein expression suggesting cytokine release, correlating timewise with IL-33 receptor expression by reactive CD8+ T cells and their greatly augmented expansion in WT versus ll33-/- mice. Using mice lacking IL-33 selectively in FRC versus LEC, we identify FRC as key IL-33 source driving acute and chronic antiviral T-cell responses. Collectively, these findings show that LN T-zone FRC not only regulate the homeostasis of naïve T cells but also their expansion and differentiation several days into an antiviral response.


Assuntos
Interleucina-33/metabolismo , Coriomeningite Linfocítica/imunologia , Doença Aguda , Imunidade Adaptativa , Animais , Linfócitos T CD8-Positivos/imunologia , Doença Crônica , Células Endoteliais/imunologia , Fibroblastos/imunologia , Homeostase , Humanos , Imunidade Inata , Interleucina-33/deficiência , Interleucina-33/genética , Linfonodos/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Imunológicos
5.
Cell Rep ; 30(1): 37-45.e3, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31914395

RESUMO

Tissue repair requires temporal control of progenitor cell proliferation and differentiation to replenish damaged cells. In response to acute insult, group 3 innate lymphoid cells (ILC3s) regulate intestinal stem cell maintenance and subsequent tissue repair. ILC3-derived IL-22 is important for stem cell protection, but the mechanisms of ILC3-driven tissue regeneration remain incompletely defined. Here we report that ILC3-driven epithelial proliferation and tissue regeneration are independent of IL-22. In contrast, ILC3s amplify the magnitude of Hippo-Yap1 signaling in intestinal crypt cells, ensuring adequate initiation of tissue repair and preventing excessive pathology. Mechanistically, ILC3-driven tissue repair is Stat3 independent, but it involves activation of Src family kinases. Our findings reveal that ILC3-driven intestinal repair entails distinct transcriptional networks to control stem cell maintenance and epithelial regeneration, which implies that tissue repair and crypt proliferation can be influenced by targeting innate immune cells independent of the well-established effects of IL-22.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Imunidade Inata , Intestinos/imunologia , Intestinos/patologia , Linfócitos/patologia , Cicatrização , Animais , Diferenciação Celular , Proliferação de Células , Receptor gp130 de Citocina/metabolismo , Ativação Enzimática , Interleucinas/metabolismo , Camundongos Endogâmicos C57BL , Multimerização Proteica , Regeneração , Transdução de Sinais , Células-Tronco/patologia , Proteínas de Sinalização YAP , Quinases da Família src/metabolismo , Interleucina 22
6.
Eur J Immunol ; 49(2): 290-301, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30537036

RESUMO

Under homeostatic conditions, dendritic cells (DCs) continuously patrol the intestinal lamina propria. Upon antigen encounter, DCs initiate C-C motif chemokine receptor 7 (CCR7) expression and migrate into lymph nodes to direct T cell activation and differentiation. The mechanistic underpinnings of DC migration from the tissues to lymph nodes have been largely elucidated, contributing greatly to our understanding of DC functionality and intestinal immunity. In contrast, the molecular mechanisms allowing DCs to efficiently migrate through the complex extracellular matrix of the intestinal lamina propria prior to antigen encounter are still incompletely understood. Here we show that small intestinal murine CD11b+ CD103+ DCs express Placenta-expressed transcript 1 (Plet1), a glycophoshatidylinositol (GPI)-anchored surface protein involved in migration of keratinocytes during wound healing. In the absence of Plet1, CD11b+ CD103+ DCs display aberrant migratory behavior, and accumulate in the small intestine, independent of CCR7 responsiveness. RNA-sequencing indicated involvement of Plet1 in extracellular matrix-interactiveness, and subsequent in-vitro migration assays revealed that Plet1 augments the ability of DCs to migrate through extracellular matrix containing environments. In conclusion, our findings reveal that expression of Plet1 facilitates homeostatic interstitial migration of small intestinal DCs.


Assuntos
Movimento Celular/imunologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/imunologia , Intestino Delgado/imunologia , Proteínas da Gravidez/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Movimento Celular/genética , Camundongos , Camundongos Knockout , Proteínas da Gravidez/genética
7.
J Exp Med ; 215(4): 1069-1077, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29472496

RESUMO

IL-7 is essential for the development and homeostasis of T and B lymphocytes and is critical for neonatal lymph node organogenesis because Il7-/- mice lack normal lymph nodes. Whether IL-7 is a continued requirement for normal lymph node structure and function is unknown. To address this, we ablated IL-7 function in normal adult hosts. Either inducible Il7 gene deletion or IL-7R blockade in adults resulted in a rapid loss of lymph node cellularity and a corresponding defect in lymphocyte entry into lymph nodes. Although stromal and dendritic cell components of lymph nodes were present in normal numbers and representation, innate lymphoid cell (ILC) subpopulations were substantially decreased after IL-7 ablation. Testing lymphocyte homing in bone marrow chimeras reconstituted with Rorc-/- bone marrow confirmed that ILC3s in lymph nodes are required for normal lymphocyte homing. Collectively, our data suggest that maintenance of intact lymph nodes relies on IL-7-dependent maintenance of ILC3 cells.


Assuntos
Linfócitos B/citologia , Imunidade Inata , Interleucina-7/metabolismo , Linfonodos/citologia , Linfócitos/citologia , Linfócitos/metabolismo , Linfócitos T/citologia , Animais , Movimento Celular , Células Dendríticas/metabolismo , Deleção de Genes , Loci Gênicos , Contagem de Linfócitos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Receptores de Interleucina-7/metabolismo , Células Estromais/metabolismo
8.
Cell Rep ; 21(3): 823-833, 2017 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045847

RESUMO

A substantial number of human and mouse group 3 innate lymphoid cells (ILC3s) reside in secondary lymphoid organs, yet the phenotype and function of these ILC3s is incompletely understood. Here, we employed an unbiased cross-tissue transcriptomic approach to compare human ILC3s from non-inflamed lymph nodes and spleen to their phenotypic counterparts in inflamed tonsils and from circulation. These analyses revealed that, in the absence of inflammation, lymphoid organ-residing ILC3s lack transcription of cytokines associated with classical ILC3 functions. This was independent of expression of the natural cytotoxicity receptor NKp44. However, and in contrast to ILC3s from peripheral blood, lymphoid organ-residing ILC3s express activating cytokine receptors and have acquired the ability to be recruited into immune responses by inflammatory cytokines. This comprehensive cross-tissue dataset will allow for identification of functional changes in human lymphoid organ ILC3s associated with human disease.


Assuntos
Ciclo Celular/genética , Perfilação da Expressão Gênica/métodos , Imunidade Inata , Inflamação/imunologia , Sistema Linfático/metabolismo , Linfócitos/metabolismo , Linfócitos/patologia , Animais , Comunicação Celular/genética , Citocinas/genética , Citocinas/metabolismo , Humanos , Camundongos , Receptor 2 Desencadeador da Citotoxicidade Natural/metabolismo , Transcrição Gênica , Transcriptoma/genética
9.
Eur J Immunol ; 46(6): 1404-14, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27067635

RESUMO

Interleukin 22 (IL-22) expression is associated with increased joint destruction and disease progression in rheumatoid arthritis (RA). Although IL-22 is considered a pro-inflammatory cytokine, its mechanism of action in RA remains incompletely understood. Here, we used the collagen-induced arthritis model in IL-22 deficient (IL-22(-/-) ) mice to study the role of IL-22 in RA. In spite of normal disease incidence, disease severity is significantly diminished in IL-22(-/-) mice. Moreover, pathogenicity of Th17 cells and development and function of B cells are unaffected. In contrast, splenic plasma cells, as well as serum autoantibody titers, are reduced in the absence of IL-22. At the peak of disease, germinal centers (GCs) are severely reduced in the spleens of IL-22(-/-) mice, correlating with a decline in GC B-cell numbers. Within the GC, we identified IL-22R1 expressing follicular dendritic cell-like stromal cells. Human lymphoid stromal cells respond to IL-22 ex vivo by inducing transcription of CXCL12 and CXCL13. We therefore postulate IL-22 as an important enhancer of the GC reaction, maintaining chemokine levels for the persistence of GC reactions, essential for the production of autoantibody-secreting plasma cells. Blocking IL-22 might therefore prevent immune-complex deposition and destruction of joints in RA patients.


Assuntos
Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Artrite Experimental/etiologia , Autoanticorpos/imunologia , Interleucinas/deficiência , Animais , Especificidade de Anticorpos/imunologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Técnicas de Cocultura , Modelos Animais de Doenças , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Knockout , Plasmócitos/imunologia , Plasmócitos/metabolismo , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Índice de Gravidade de Doença , Células Estromais/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Interleucina 22
10.
J Exp Med ; 212(11): 1783-91, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26392223

RESUMO

Disruption of the intestinal epithelial barrier allows bacterial translocation and predisposes to destructive inflammation. To ensure proper barrier composition, crypt-residing stem cells continuously proliferate and replenish all intestinal epithelial cells within days. As a consequence of this high mitotic activity, mucosal surfaces are frequently targeted by anticancer therapies, leading to dose-limiting side effects. The cellular mechanisms that control tissue protection and mucosal healing in response to intestinal damage remain poorly understood. Type 3 innate lymphoid cells (ILC3s) are regulators of homeostasis and tissue responses to infection at mucosal surfaces. We now demonstrate that ILC3s are required for epithelial activation and proliferation in response to small intestinal tissue damage induced by the chemotherapeutic agent methotrexate. Multiple subsets of ILC3s are activated after intestinal tissue damage, and in the absence of ILC3s, epithelial activation is lost, correlating with increased pathology and severe damage to the intestinal crypts. Using ILC3-deficient Lgr5 reporter mice, we show that maintenance of intestinal stem cells after damage is severely impaired in the absence of ILC3s or the ILC3 signature cytokine IL-22. These data unveil a novel function of ILC3s in limiting tissue damage by preserving tissue-specific stem cells.


Assuntos
Imunidade Inata , Mucosa Intestinal/imunologia , Linfócitos/fisiologia , Células-Tronco/fisiologia , Animais , Interleucinas/fisiologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Metotrexato/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Interleucina 22
11.
J Immunol ; 195(9): 4257-4263, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26378073

RESUMO

Adaptive immunity critically depends on the functional compartmentalization of secondary lymphoid organs. Mesenchymal stromal cells create and maintain specialized niches that support survival, activation, and expansion of T and B cells, and integrated analysis of lymphocytes and their niche has been instrumental in understanding adaptive immunity. Lymphoid organs are also home to type 3 innate lymphoid cells (ILC3), innate effector cells essential for barrier immunity. However, a specialized stromal niche for ILC3 has not been identified. A novel lineage-tracing approach now identifies a subset of murine fetal lymphoid tissue organizer cells that gives rise exclusively to adult marginal reticular cells. Moreover, both cell types are conserved from mice to humans and colocalize with ILC3 in secondary lymphoid tissues throughout life. In sum, we provide evidence that fetal stromal organizers give rise to adult marginal reticular cells and form a dedicated stromal niche for innate ILC3 in adaptive lymphoid organs.


Assuntos
Imunidade Inata/imunologia , Linfócitos/imunologia , Tecido Linfoide/imunologia , Nicho de Células-Tronco/imunologia , Células Estromais/imunologia , Animais , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Quimiocinas/imunologia , Quimiocinas/metabolismo , Feminino , Feto/citologia , Citometria de Fluxo , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/metabolismo , Linfócitos/metabolismo , Tecido Linfoide/citologia , Tecido Linfoide/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Camundongos Transgênicos , Microscopia Confocal , Ligante RANK/genética , Ligante RANK/imunologia , Ligante RANK/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nicho de Células-Tronco/genética , Células Estromais/citologia , Células Estromais/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
12.
Am J Pathol ; 185(7): 1935-43, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25963989

RESUMO

Tertiary lymphoid structures (TLSs) in chronic inflammation, including rheumatoid arthritis (RA) synovial tissue (ST), often contain high endothelial venules and follicular dendritic cells (FDCs). Endothelial cell (EC)-specific lymphotoxin ß (LTß) receptor signaling is critical for the formation of lymph nodes and high endothelial venules. FDCs arise from perivascular platelet-derived growth factor receptor ß(+) precursor cells (preFDCs) that require specific group 3 innate lymphoid cells (ILC3s) and LTß for their expansion. Previously, we showed that RA ST contains ECs that express NF-κB-inducing kinase (NIK), which is pivotal in LTß-induced noncanonical NF-κB signaling. We studied the relation between NIK(+) ECs, (pre)FDCs, and ILC3s with respect to TLSs in RA ST. TLS(+) tissues exhibited a significantly increased expression of genes involved in noncanonical NF-κB signaling, including NIK, and immunohistochemical analysis revealed that NIK was almost exclusively expressed by ECs. ILC3s were present in human RA ST in very low numbers, but not differentially in TLS(+) tissues. In contrast, TLS(+) tissues contained significantly more NIK(+) ECs and perivascular platelet-derived growth factor receptor ß(+) preFDCs, which correlated significantly with the quantity of FDCs. We established a strong link between NIK(+) ECs, (pre)FDCs, and the presence of TLSs, indicating that NIK(+) ECs may not only be important orchestrators of lymph node development but also contribute to the formation of TLSs in chronic inflammation.


Assuntos
Artrite Reumatoide/patologia , Células Endoteliais/enzimologia , Tecido Linfoide/patologia , Transdução de Sinais , Adulto , Idoso , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Células Endoteliais/metabolismo , Feminino , Humanos , Inflamação , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Receptor beta de Linfotoxina/metabolismo , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Membrana Sinovial/imunologia , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Quinase Induzida por NF-kappaB
13.
Blood ; 125(3): 465-9, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25301706

RESUMO

Clusters of cells attached to the endothelium of the main embryonic arteries were first observed a century ago. Present in most vertebrate species, such clusters, or intraaortic hematopoietic clusters (IAHCs), derive from specialized hemogenic endothelial cells and contain the first few hematopoietic stem cells (HSCs) generated during embryonic development. However, some discrepancies remained concerning the spatio-temporal appearance and the numbers of IAHCs and HSCs. Therefore, the exact cell composition and function of IAHCs remain unclear to date. We show here that IAHCs contain pre-HSCs (or HSC precursors) that can mature into HSCs in vivo (as shown by the successful long-term multilineage reconstitution of primary neonates and secondary adult recipients). Such IAHC pre-HSCs could contribute to the HSC pool increase observed at midgestation. The novel insights in pre-HSC to HSC transition represent an important step toward generating transplantable HSCs in vitro that are needed for autologous HSC transplantation therapies.


Assuntos
Aorta/embriologia , Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Animais , Feminino , Camundongos , Técnicas de Cultura de Órgãos
14.
Front Immunol ; 3: 72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22566953

RESUMO

Human RORC(+) lymphoid tissue inducer cells are part of a rapidly expanding family of innate lymphoid cells (ILC) that participate in innate and adaptive immune responses as well as in lymphoid tissue (re) modeling. The assessment of a potential role for innate lymphocyte-derived cytokines in human homeostasis and disease is hampered by a poor characterization of RORC(+) innate cell subsets and a lack of knowledge on the distribution of these cells in adults. Here we show that functionally distinct subsets of human RORC(+) innate lymphoid cells are enriched for secretion of IL-17a or IL-22. Both subsets have an activated phenotype and can be distinguished based on the presence or absence of the natural cytotoxicity receptor NKp44. NKp44(+) IL-22 producing cells are present in tonsils while NKp44(-) IL-17a producing cells are present in fetal developing lymph nodes. Development of human intestinal NKp44(+) ILC is a programmed event that is independent of bacterial colonization and these cells colonize the fetal intestine during the first trimester. In the adult intestine, NKp44(+) ILC are the main ILC subset producing IL-22. NKp44(-) ILC remain present throughout adulthood in peripheral non-inflamed lymph nodes as resting, non-cytokine producing cells. However, upon stimulation lymph node ILC can swiftly initiate cytokine transcription suggesting that secondary human lymphoid organs may function as a reservoir for innate lymphoid cells capable of participating in inflammatory responses.

15.
Eur J Immunol ; 41(11): 3340-50, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21830211

RESUMO

Hepatic NK cells constitute ≈ 40% of hepatic lymphocytes and are phenotypically and functionally distinct from blood NK cells. Whether hepatic NK cells derive from precursors in the BM or develop locally from hepatic progenitors is still unknown. Here, we identify all five known sequential stages of NK-cell development in the adult human liver and demonstrate that CD34(+) hepatic progenitors can generate functional NK cells. While early NK-cell precursors (NKPs) were similar in liver and blood, hepatic stage 3 NKPs displayed immunophenotypical differences, suggesting the onset of a liver-specific NK-cell development. Hepatic stage 3 NKPs were RORC(neg) and did not produce IL-17 or IL-22, excluding them from the lymphoid tissue-inducer (LTi) subset. In vitro culture of hepatic NKPs gave rise to functional NK cells exhibiting strong cytotoxicity against K562 targets. To determine whether hepatic NKPs are stably residing in the liver, we analyzed donor and recipient-derived cells in transplanted livers. Shortly after liver transplantation all donor NKPs in liver grafts were replaced by recipient-derived ones, indicating that hepatic NKPs are recruited from the bloodstream. Together, our results show that NKPs are continuously recruited from peripheral blood into the liver and can potentially differentiate into liver-specific NK cells.


Assuntos
Diferenciação Celular/imunologia , Células-Tronco Hematopoéticas/citologia , Células Matadoras Naturais/citologia , Fígado/citologia , Fígado/imunologia , Adulto , Separação Celular , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Humanos , Células Matadoras Naturais/imunologia , Reação em Cadeia da Polimerase
16.
Cell Stem Cell ; 5(4): 385-95, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19796619

RESUMO

Hematopoietic stem cells (HSCs) are responsible for the life-long production of the blood system and are pivotal cells in hematologic transplantation therapies. During mouse and human development, the first HSCs are produced in the aorta-gonad-mesonephros region. Subsequent to this emergence, HSCs are found in other anatomical sites of the mouse conceptus. While the mouse placenta contains abundant HSCs at midgestation, little is known concerning whether HSCs or hematopoietic progenitors are present and supported in the human placenta during development. In this study we show, over a range of developmental times including term, that the human placenta contains hematopoietic progenitors and HSCs. Moreover, stromal cell lines generated from human placenta at several developmental time points are pericyte-like cells and support human hematopoiesis. Immunostaining of placenta sections during development localizes hematopoietic cells in close contact with pericytes/perivascular cells. Thus, the human placenta is a potent hematopoietic niche throughout development.


Assuntos
Células-Tronco Hematopoéticas/citologia , Sistema Hematopoético/citologia , Placenta/citologia , Animais , Transplante de Células , Feminino , Citometria de Fluxo , Idade Gestacional , Humanos , Imuno-Histoquímica , Camundongos , Reação em Cadeia da Polimerase , Gravidez
17.
Nat Immunol ; 10(1): 66-74, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19029905

RESUMO

The human body contains over 500 individual lymph nodes, yet the biology of their formation is poorly understood. Here we identify human lymphoid tissue-inducer cells (LTi cells) as lineage-negative RORC+ CD127+ cells with the functional ability to interact with mesenchymal cells through lymphotoxin and tumor necrosis factor. Human LTi cells were committed natural killer (NK) cell precursors that produced interleukin 17 (IL-17) and IL-22. In vitro, LTi cells gave rise to RORC+ CD127+ NK cells that retained the ability to produce IL-17 and IL-22. Postnatally, similar populations of LTi cell-like cells and RORC+ CD127+ NK cells were present in tonsils, and both secreted IL-17 and IL-22 but no interferon-gamma. Our data indicate that lymph node organogenesis is controlled by an NK cell precursor population with adaptive immune features and demonstrate a previously unappreciated link between the innate and adaptive immune systems.


Assuntos
Interleucina-17/biossíntese , Linfonodos/embriologia , Linfonodos/imunologia , Células T Matadoras Naturais/imunologia , Organogênese , Células Precursoras de Linfócitos T/imunologia , Animais , Antígeno CD56/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Imunidade Celular , Imunidade Inata , Interferon gama/biossíntese , Subunidade alfa de Receptor de Interleucina-7/imunologia , Interleucinas/biossíntese , Linfonodos/citologia , Tecido Linfoide/embriologia , Tecido Linfoide/imunologia , Linfotoxina-alfa/imunologia , Mesentério/embriologia , Mesentério/imunologia , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Tonsila Palatina/citologia , Tonsila Palatina/imunologia , Receptores do Ácido Retinoico/imunologia , Receptores dos Hormônios Tireóideos/imunologia , Baço/embriologia , Baço/imunologia , Interleucina 22
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...