Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Nature ; 595(7868): 560-564, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34262176

RESUMO

The balance between bacterial colonization and its containment in the intestine is indispensable for the symbiotic relationship between humans and their bacteria. One component to maintain homeostasis at the mucosal surfaces is immunoglobulin A (IgA), the most abundant immunoglobulin in mammals1,2. Several studies have revealed important characteristics of poly-reactive IgA3,4, which is produced naturally without commensal bacteria. Considering the dynamic changes within the gut environment, however, it remains uncertain how the commensal-reactive IgA pool is shaped and how such IgA affects the microbial community. Here we show that acetate-one of the major gut microbial metabolites-not only increases the production of IgA in the colon, but also alters the capacity of the IgA pool to bind to specific microorganisms including Enterobacterales. Induction of commensal-reactive IgA and changes in the IgA repertoire by acetate were observed in mice monocolonized with Escherichia coli, which belongs to Enterobacterales, but not with the major commensal Bacteroides thetaiotaomicron, which suggests that acetate directs selective IgA binding to certain microorganisms. Mechanistically, acetate orchestrated the interactions between epithelial and immune cells, induced microbially stimulated CD4 T cells to support T-cell-dependent IgA production and, as a consequence, altered the localization of these bacteria within the colon. Collectively, we identified a role for gut microbial metabolites in the regulation of differential IgA production to maintain mucosal homeostasis.


Assuntos
Acetatos/farmacologia , Bactérias/imunologia , Microbioma Gastrointestinal/imunologia , Imunoglobulina A/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Colo/imunologia , Dieta , Ácidos Graxos Voláteis/metabolismo , Homeostase/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Simbiose
2.
Nat Immunol ; 13(8): 729-36, 2012 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-22706340

RESUMO

Intestinal microfold cells (M cells) are an enigmatic lineage of intestinal epithelial cells that initiate mucosal immune responses through the uptake and transcytosis of luminal antigens. The mechanisms of M-cell differentiation are poorly understood, as the rarity of these cells has hampered analysis. Exogenous administration of the cytokine RANKL can synchronously activate M-cell differentiation in mice. Here we show the Ets transcription factor Spi-B was induced early during M-cell differentiation. Absence of Spi-B silenced the expression of various M-cell markers and prevented the differentiation of M cells in mice. The activation of T cells via an oral route was substantially impaired in the intestine of Spi-B-deficient (Spib(-/-)) mice. Our study demonstrates that commitment to the intestinal M-cell lineage requires Spi-B as a candidate master regulator.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Mucosa Intestinal/citologia , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Animais , Linhagem da Célula , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Humanos , Imunidade nas Mucosas/genética , Mucosa Intestinal/embriologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Ligante RANK/farmacologia , Linfócitos T/imunologia
3.
Traffic ; 21(1): 34-44, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31647148

RESUMO

Mucosal immune responses in the inductive lymphoid tissues of the intestine begin with uptake of particulate antigens, including components of the gut microbiota by specialized antigen sampling M cells. M cells represent a distinct lineage of enterocytes that arise from crypt stem cells in response to the cytokine receptor of NF-κB ligand (RANKL). Full differentiation of M cells requires the transcription factor Spi-B to yield mature M cells that express multiple receptors for bacteria including glycoprotein 2. M cell differentiation can be recapitulated in vitro using three-dimensional enteroid cultures of primary intestinal stem cells supplemented with RANKL. This article summarizes the current knowledge about the genesis of intestinal M cells and highlights some of the remaining unanswered questions about this enigmatic cell type.


Assuntos
Microbiota , Ligante RANK , Diferenciação Celular , Humanos , Imunidade nas Mucosas , Mucosa Intestinal
4.
Proc Natl Acad Sci U S A ; 114(8): 1976-1981, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28159893

RESUMO

A significant challenge to HIV eradication is the elimination of viral reservoirs in germinal center (GC) T follicular helper (Tfh) cells. However, GCs are considered to be immune privileged for antiviral CD8 T cells. Here, we show a population of simian immunodeficiency virus (SIV)-specific CD8 T cells express CXCR5 (C-X-C chemokine receptor type 5, a chemokine receptor required for homing to GCs) and expand in lymph nodes (LNs) following pathogenic SIV infection in a cohort of vaccinated macaques. This expansion was greater in animals that exhibited superior control of SIV. The CXCR5+ SIV-specific CD8 T cells demonstrated enhanced polyfunctionality, restricted expansion of antigen-pulsed Tfh cells in vitro, and possessed a unique gene expression pattern related to Tfh and Th2 cells. The increase in CXCR5+ CD8 T cells was associated with the presence of higher frequencies of SIV-specific CD8 T cells in the GC. Following TCR-driven stimulation in vitro, CXCR5+ but not CXCR5- CD8 T cells generated both CXCR5+ as well as CXCR5- cells. However, the addition of TGF-ß to CXCR5- CD8 T cells induced a population of CXCR5+ CD8 T cells, suggesting that this cytokine may be important in modulating these CXCR5+ CD8 T cells in vivo. Thus, CXCR5+ CD8 T cells represent a unique subset of antiviral CD8 T cells that expand in LNs during chronic SIV infection and may play a significant role in the control of pathogenic SIV infection.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Centro Germinativo/citologia , Receptores CXCR5/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Doença Crônica , Macaca mulatta , Masculino
5.
PLoS Pathog ; 12(12): e1006075, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27973593

RESUMO

Many natural prion diseases of humans and animals are considered to be acquired through oral consumption of contaminated food or pasture. Determining the route by which prions establish host infection will identify the important factors that influence oral prion disease susceptibility and to which intervention strategies can be developed. After exposure, the early accumulation and replication of prions within small intestinal Peyer's patches is essential for the efficient spread of disease to the brain. To replicate within Peyer's patches, the prions must first cross the gut epithelium. M cells are specialised epithelial cells within the epithelia covering Peyer's patches that transcytose particulate antigens and microorganisms. M cell-development is dependent upon RANKL-RANK-signalling, and mice in which RANK is deleted only in the gut epithelium completely lack M cells. In the specific absence of M cells in these mice, the accumulation of prions within Peyer's patches and the spread of disease to the brain was blocked, demonstrating a critical role for M cells in the initial transfer of prions across the gut epithelium in order to establish host infection. Since pathogens, inflammatory stimuli and aging can modify M cell-density in the gut, these factors may also influence oral prion disease susceptibility. Mice were therefore treated with RANKL to enhance M cell density in the gut. We show that prion uptake from the gut lumen was enhanced in RANKL-treated mice, resulting in shortened survival times and increased disease susceptibility, equivalent to a 10-fold higher infectious titre of prions. Together these data demonstrate that M cells are the critical gatekeepers of oral prion infection, whose density in the gut epithelium directly limits or enhances disease susceptibility. Our data suggest that factors which alter M cell-density in the gut epithelium may be important risk factors which influence host susceptibility to orally acquired prion diseases.


Assuntos
Suscetibilidade a Doenças , Células Epiteliais , Mucosa Intestinal , Doenças Priônicas/metabolismo , Animais , Modelos Animais de Doenças , Imunofluorescência , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real , Transcitose/fisiologia
6.
Int Immunol ; 29(10): 471-478, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29186424

RESUMO

Nasopharynx-associated lymphoid tissue (NALT) is one of the major constituents of the mucosa-associated lymphoid tissue (MALT), and has the ability to induce antigen-specific immune responses. However, the molecular mechanisms responsible for antigen uptake from the nasal cavity into the NALT remain largely unknown. Immunohistochemical analysis showed that CCL9 and CCL20 were co-localized with glycoprotein 2 (GP2) in the epithelium covering NALT, suggesting the existence of M cells in NALT. In analogy with the reduced number of Peyer's patch M cells in CCR6-deficient mice, the number of NALT M cells was drastically decreased in CCR6-deficient mice compared with the wild-type mice. Translocation of nasally administered Salmonella enterica serovar Typhimurium into NALT via NALT M cells was impaired in CCR6-deficient mice, whereas S. Typhimurium demonstrated consistent co-localization with NALT M cells in wild-type mice. When wild-type mice were nasally administered with an attenuated vaccine strain of S. Typhimurium, the mice were protected from a subsequent challenge with wild-type S. Typhimurium. Antigen-specific fecal and nasal IgA was detected after nasal immunization with the attenuated vaccine strain of S. Typhimurium only in wild-type mice but not in CCR6-deficient mice. Taken together, these observations demonstrate that NALT M cells are important as a first line of defense against infection by enabling activation of the common mucosal immune system (CMIS).


Assuntos
Células Epiteliais/imunologia , Imunidade nas Mucosas/imunologia , Tecido Linfoide/imunologia , Nasofaringe/imunologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
8.
Immunology ; 152(4): 613-627, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28746740

RESUMO

The intestinal lamina propria (LP) contains antigen-presenting cells with features of dendritic cells and macrophages, collectively referred to as mononuclear phagocytes (MNPs). Association of MNPs with the epithelium is thought to play an important role in multiple facets of intestinal immunity including imprinting MNPs with the ability to induce IgA production, inducing the expression of gut homing molecules on T cells, facilitating the capture of luminal antigens and microbes, and subsequent immune responses in the mesenteric lymph node (MLN). However, the factors promoting this process in the steady state are largely unknown, and in vivo models to test and confirm the importance of LP-MNP association with the epithelium for these outcomes are unexplored. Evaluation of epithelial expression of chemoattractants in mice where MNP-epithelial associations were impaired suggested CCL20 as a candidate promoting epithelial association. Expression of CCR6, the only known receptor for CCL20, was required for MNPs to associate with the epithelium. LP-MNPs from CCR6-/- mice did not display defects in acquiring antigen and stimulating T-cell responses in ex vivo assays or in responses to antigen administered systemically. However, LP-MNPs from CCR6-deficient mice were impaired at acquiring luminal and epithelial antigens, inducing IgA production in B cells, inducing immune responses in the MLN, and capturing and trafficking luminal commensal bacteria to the MLN. These findings identify a crucial role for CCR6 in promoting LP-MNPs to associate with the intestinal epithelium in the steady state to perform multiple functions promoting gut immune homeostasis.


Assuntos
Células Dendríticas/imunologia , Impressão Genômica/imunologia , Vigilância Imunológica , Mucosa Intestinal/imunologia , Macrófagos/imunologia , Receptores CCR6/imunologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Quimiocina CCL20/genética , Quimiocina CCL20/imunologia , Células Dendríticas/citologia , Humanos , Macrófagos/citologia , Camundongos , Camundongos Knockout , Receptores CCR6/genética , Linfócitos T/citologia , Linfócitos T/imunologia
9.
Immunity ; 29(3): 451-63, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18799151

RESUMO

Medullary thymic epithelial cells (mTECs) are specialized for inducing central immunological tolerance to self-antigens. To accomplish this, mTECs must adopt a mature phenotype characterized by expression of the autoimmune regulator Aire, which activates the transcription of numerous genes encoding tissue-restricted self-antigens. The mechanisms that control mature Aire(+) mTEC development in the postnatal thymus remain poorly understood. We demonstrate here that, although either CD4(+) or CD8(+) thymocytes are sufficient to sustain formation of a well-defined medulla, expansion of the mature mTEC population requires autoantigen-specific interactions between positively selected CD4(+) thymocytes bearing autoreactive T cell receptor (TCR) and mTECs displaying cognate self-peptide-MHC class II complexes. These interactions also involve the engagement of CD40 on mTECs by CD40L induced on the positively selected CD4(+) thymocytes. This antigen-specific TCR-MHC class II-mediated crosstalk between CD4(+) thymocytes and mTECs defines a unique checkpoint in thymic stromal development that is pivotal for generating a mature mTEC population competent for ensuring central T cell tolerance.


Assuntos
Autoantígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Epiteliais/imunologia , Timo/imunologia , Fatores de Transcrição/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Ligante de CD40/imunologia , Ligante de CD40/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Tolerância a Antígenos Próprios , Timo/citologia , Timo/metabolismo , Transativadores/imunologia , Transativadores/metabolismo , Fatores de Transcrição/imunologia , Proteína AIRE
10.
Am J Physiol Cell Physiol ; 311(3): C498-507, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27413168

RESUMO

Microfold (M) cells are phagocytic intestinal epithelial cells in the follicle-associated epithelium of Peyer's patches that transport particulate antigens from the gut lumen into the subepithelial dome. Differentiation of M cells from epithelial stem cells in intestinal crypts requires the cytokine receptor activator of NF-κB ligand (RANKL) and the transcription factor Spi-B. We used three-dimensional enteroid cultures established with small intestinal crypts from mice as a model system to investigate signaling pathways involved in M cell differentiation and the influence of other cytokines on RANKL-induced M cell differentiation. Addition of RANKL to enteroids induced expression of multiple M cell-associated genes, including Spib, Ccl9 [chemokine (C-C motif) ligand 9], Tnfaip2 (TNF-α-induced protein 2), Anxa5 (annexin A5), and Marcksl1 (myristoylated alanine-rich protein kinase C substrate) in 1 day. The mature M cell marker glycoprotein 2 (Gp2) was strongly induced by 3 days and expressed by 11% of cells in enteroids. The noncanonical NF-κB pathway was required for RANKL-induced M cell differentiation in enteroids, as addition of RANKL to enteroids from mice with a null mutation in the mitogen-activated protein kinase kinase kinase 14 (Map3k14) gene encoding NF-κB-inducing kinase failed to induce M cell-associated genes. While the cytokine TNF-α alone had little, if any, effect on expression of M cell-associated genes, addition of TNF-α to RANKL consistently resulted in three- to sixfold higher levels of multiple M cell-associated genes than RANKL alone. One contributing mechanism is the rapid induction by TNF-α of Relb and Nfkb2 (NF-κB subunit 2), genes encoding the two subunits of the noncanonical NF-κB heterodimer. We conclude that endogenous activators of canonical NF-κB signaling present in the gut-associated lymphoid tissue microenvironment, including TNF-α, can play a supportive role in the RANKL-dependent differentiation of M cells in the follicle-associated epithelium.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiologia , Intestinos/fisiologia , Ligante RANK/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Biomarcadores/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Feminino , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
11.
J Virol ; 88(12): 6934-43, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24696493

RESUMO

UNLABELLED: Microfold (M) cells are specialized intestinal epithelial cells that internalize particulate antigens and aid in the establishment of immune responses to enteric pathogens. M cells have also been suggested as a portal for pathogen entry into the host. While virus particles have been observed in M cells, it is not known whether viruses use M cells to initiate a productive infection. Noroviruses (NoVs) are single-stranded RNA viruses that infect host organisms via the fecal-oral route. Murine NoV (MNV) infects intestinal macrophages and dendritic cells and provides a tractable experimental system for understanding how an enteric virus overcomes the intestinal epithelial barrier to infect underlying target cells. We found that replication of two divergent MNV strains was reduced in mice depleted of M cells. Reoviruses are double-stranded RNA viruses that infect hosts via respiratory or enteric routes. In contrast to MNV, reovirus infects enterocytes in the intestine. Despite differences in cell tropism, reovirus infection was also reduced in M cell-depleted mice. These data demonstrate that M cells are required for the pathogenesis of two unrelated enteric viruses that replicate in different cell types within the intestine. IMPORTANCE: To successfully infect their hosts, pathogens that infect via the gastrointestinal tract must overcome the multilayered system of host defenses. Microfold (M) cells are specialized intestinal epithelial cells that internalize particulate antigens and aid in the establishment of immune responses to enteric pathogens. Virus particles have been observed within M cells. However, it is not known whether viruses use M cells to initiate a productive infection. To address this question, we use MNV and reovirus, two enteric viruses that replicate in different cell types in the intestine, intestinal epithelial cells for reovirus and intestinal mononuclear phagocytes for MNV. Interestingly, MNV- and reovirus-infected mice depleted of M cells showed reduced viral loads in the intestine. Thus, our work demonstrates the importance of M cells in the pathogenesis of enteric viruses irrespective of the target cell type in which the virus replicates.


Assuntos
Infecções por Caliciviridae/virologia , Células Epiteliais/virologia , Intestinos/virologia , Norovirus/fisiologia , Infecções por Reoviridae/virologia , Reoviridae/fisiologia , Replicação Viral , Animais , Linhagem Celular , Humanos , Intestinos/citologia , Camundongos , Camundongos Endogâmicos BALB C
12.
Proc Natl Acad Sci U S A ; 108(13): 5342-7, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21402940

RESUMO

Receptor activator of NF-κB (RANK), known for controlling bone mass, has been recognized for its role in epithelial cell activation of the mammary gland. Because bone and the epidermo-pilosebaceous unit of the skin share a lifelong renewal activity where similar molecular players operate, and because mammary glands and hair follicles are both skin appendages, we have addressed the function of RANK in the hair follicle and the epidermis. Here, we show that mice deficient in RANK ligand (RANKL) are unable to initiate a new growth phase of the hair cycle and display arrested epidermal homeostasis. However, transgenic mice overexpressing RANK in the hair follicle or administration of recombinant RANKL both activate the hair cycle and epidermal growth. RANK is expressed by the hair follicle germ and bulge stem cells and the epidermal basal cells, cell types implicated in the renewal of the epidermo-pilosebaceous unit. RANK signaling is dispensable for the formation of the stem cell compartment and the inductive hair follicle mesenchyme, and the hair cycle can be rescued by Rankl knockout skin transplantation onto nude mice. RANKL is actively transcribed by the hair follicle at initiation of its growth phase, providing a mechanism for stem cell RANK engagement and hair-cycle entry. Thus, RANK-RANKL regulates hair renewal and epidermal homeostasis and provides a link between these two activities.


Assuntos
Proliferação de Células , Células Epidérmicas , Células Epiteliais/fisiologia , Folículo Piloso/citologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Epiderme/fisiologia , Células Epiteliais/citologia , Folículo Piloso/fisiologia , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , NF-kappa B/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Transplante de Pele , Células-Tronco/citologia , Células-Tronco/fisiologia
13.
Gut ; 62(1): 53-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22267601

RESUMO

BACKGROUND: The earliest endoscopically-evident lesion in Crohn's disease is the aphthous ulcer, which develops over ectopic lymphoid tissues (ie, inducible lymphoid follicles (ILF), tertiary lymphoid tissue (TLT)) in the chronically inflamed intestine. ILF/TLT are induced within effector sites by homeostatic lymphoid chemokines, but their role in the development of intestinal ILF/TLT and in the pathogenesis of Crohn's disease is poorly understood. DESIGN: Using a mouse model of Crohn's-like ileitis (TNFARE) which develops florid induction of ILF/TLT within its terminal ileum, the contribution of the CCR7/CCL19/CCL21 chemokine axis during the development of TLT and its role in disease pathogenesis were assessed. RESULTS: Both CCL19 and CCL21 were increased within the inflamed ileum of TNFARE mice, which resulted in CCR7 internalisation and impaired T cell chemotaxis. ILF/TLT were a major source of CCL19 and CCL21 and increased local synthesis, augmented recruitment/retention of effector, naïve and central memory T cell subsets within the inflamed ileum. Immunoblockade of CCR7 resulted in further effector T cell retention and exacerbation of ileitis. CONCLUSIONS: Induction of ILF/TLT in the chronically inflamed intestine alters the homeostatic CCL19-CCL21 lymphoid-chemokine gradient and increases recruitment/retention of effector CCR7+ T cell subsets within the terminal ileum, contributing to the perpetuation of chronic inflammation. Thus, blockade of CCR7 or its ligands might result in deleterious consequences for subjects with chronic inflammatory diseases.


Assuntos
Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Coristoma/imunologia , Doença de Crohn/imunologia , Ileíte/imunologia , Receptores CCR7/metabolismo , Subpopulações de Linfócitos T/metabolismo , Animais , Biomarcadores/metabolismo , Quimiotaxia de Leucócito , Coristoma/patologia , Doença de Crohn/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Humanos , Ileíte/patologia , Tecido Linfoide , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Blood ; 117(25): 6885-94, 2011 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-21527514

RESUMO

p90 ribosomal S6 kinase 2 (p90RSK2) is important in diverse cellular processes including gene expression, cell proliferation, and survival. We found that p90RSK2 is commonly activated in diverse leukemia cell lines expressing different leukemogenic tyrosine kinases, including BCR-ABL and FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD). Interestingly, in a murine BM transplantation (BMT) model, genetic deficiency of RSK2 did not affect the pathogenesis or disease progression of BCR-ABL-induced myeloproliferative neoplasm (PN). In contrast, FLT3-ITD induced a T-cell acute lymphoblastic leukemia in BMT mice receiving RSK2 knockout (KO) BM cells, phenotypically distinct from the myeloproliferative neoplasm induced by FLT3-ITD using wild-type BM cells. In consonance with these results, inhibition of RSK2 by an RSK inhibitor, fmk, did not effectively induce apoptosis in BCR-ABL-expressing murine Ba/F3 cells, human K562 cells or primary tissue samples from CML patients, whereas fmk treatment induced significant apoptotic cell death not only in FLT3-ITD-positive Ba/F3 cells, human Molm14 and Mv(4;11) leukemia cells, but also in primary tissue samples from AML patients. These results suggest that RSK2 is dispensable for BCR-ABL-induced myeloid leukemia, but may be required for pathogenesis and lineage determination in FLT3-ITD-induced hematopoietic transformation. RSK2 may thus represent an alternative therapeutic target in the treatment of FLT3-ITD-positive leukemia.


Assuntos
Proteínas de Fusão bcr-abl/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mieloide/enzimologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Proteínas de Fusão bcr-abl/metabolismo , Técnicas de Inativação de Genes , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mieloide/genética , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
15.
Cancer Cell ; 7(2): 179-91, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15710330

RESUMO

The SH2-containing tyrosine phosphatase Shp2 (PTPN11) is required for growth factor and cytokine signaling. Germline Shp2 mutations cause Noonan Syndrome (NS), which is associated with increased risk of juvenile myelomonocytic leukemia (JMML). Somatic Shp2 mutations occur in sporadic JMML and other leukemias. We found that Shp2 mutants associated with sporadic leukemias transform murine bone marrow cells, whereas NS mutants are less potent in this assay. Transformation requires multiple domains within Shp2 and the Shp2 binding protein Gab2, and is associated with hyperactivation of the Erk, Akt, and Stat5 pathways. Mutant Shp2-transduced BM causes a fatal JMML-like disorder or, less commonly, lymphoproliferation. Shp2 mutants also cause myeloproliferation in Drosophila. Mek or Tor inhibitors potently inhibit transformation, suggesting new approaches to JMML therapy.


Assuntos
Leucemia/genética , Mutação , Proteínas Tirosina Fosfatases/genética , Alelos , Animais , Animais Geneticamente Modificados , Células da Medula Óssea/citologia , Proliferação de Células , DNA Complementar/metabolismo , Modelos Animais de Doenças , Drosophila , Drosophila melanogaster , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Interleucina-3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Leucemia/metabolismo , Camundongos , Modelos Genéticos , Neoplasias Experimentais , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Retroviridae/genética , Transdução de Sinais , Fatores de Tempo
16.
Am J Pathol ; 179(4): 1861-71, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21854748

RESUMO

Cryptopatches (CPs) and isolated lymphoid follicles (ILFs) are organized intestinal lymphoid tissues that develop postnatally in mice and include stromal cells expressing the receptor activator of nuclear factor kappa-B ligand (RANKL). We investigated how stromal RANKL influences the development and differentiation of CPs and ILFs by analyzing the development of these lymphoid structures in knockout mice lacking RANKL. We found that RANKL(-/-) mice had a fourfold reduction in the overall density of CPs in the small intestine compared to control mice, with the largest decrease in the proximal small intestine. No B cells were present in CPs from the small intestine of RANKL(-/-) mice and ILF formation was completely blocked. In sharp contrast, colonic ILFs containing B cells were present in RANKL(-/-) mice. Stromal cells within CPs in the small intestine of RANKL(-/-) mice did not express CXCL13 (originally called B lymphocyte chemoattractant) and often lacked other normally expressed stromal cell antigens, whereas colonic lymphoid aggregates in RANKL(-/-) mice retained stromal CXCL13 expression. The CXCL13-dependent maturation of precursor CPs into ILFs is differentially regulated in the small intestine and colon, with an absolute requirement for RANKL only in the small intestine.


Assuntos
Intestino Grosso/embriologia , Intestino Grosso/imunologia , Intestino Delgado/embriologia , Intestino Delgado/imunologia , Tecido Linfoide/embriologia , Organogênese , Ligante RANK/metabolismo , Animais , Antígenos/imunologia , Linfócitos B/patologia , Antígeno CD11c/metabolismo , Contagem de Células , Quimiocina CXCL13/metabolismo , Humanos , Intestino Grosso/patologia , Intestino Delgado/patologia , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , Receptor beta de Linfotoxina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Ligante RANK/deficiência , Células Estromais/imunologia , Células Estromais/patologia
17.
Int Immunol ; 23(4): 261-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21422150

RESUMO

M cells are responsible for uptake of mucosal antigens in Peyer's patches (PPs). Differentiation of M cells is thought to be induced by interactions between follicle-associated epithelium and PP cells; however, it remains elusive what types of immune cells function as M-cell inducers. Here, we attempted to identify the cells that serve as an M-cell inducer in PP. We found that a unique B-cell subset characterized by CCR6(hi)CD11c(int) resided in the subepithelial dome (SED) in mouse PP. CCR6(hi)CD11c(int) B cells showed chemotactic migration in response to CCL20. Furthermore, this unique B-cell subset substantially decreased in PP of CCR6-deficient mice, indicating that the SED localization of CCR6(hi)CD11c(int) B cells is most likely regulated by the CCL20-CCR6 system. Concomitantly, CCR6 deficiency caused remarkable decrement of M cells. Moreover, adoptive transfer of CCR6(hi)CD11c(int) B cells from wild-type mice restored the M-cell decrement in CCR6-deficient mice. Collectively, the spatial regulation of CCR6(hi)CD11c(int) B cells via the CCL20-CCR6 system may play a vital role in M-cell differentiation in mice.


Assuntos
Linfócitos B/metabolismo , Células Epiteliais/metabolismo , Subpopulações de Linfócitos/metabolismo , Nódulos Linfáticos Agregados/citologia , Receptores CCR6/metabolismo , Transferência Adotiva , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Antígeno CD11c/biossíntese , Diferenciação Celular , Movimento Celular/genética , Células Cultivadas , Quimiocina CCL20/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CCR6/genética , Receptores CCR6/imunologia
18.
Cancer Cell ; 6(6): 587-96, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15607963

RESUMO

To better understand the origin of leukemic stem cells, we tested the hypothesis that all leukemia oncogenes could transform committed myeloid progenitor cells lacking the capacity for self-renewal, as has recently been reported for MLL-ENL. Flow-sorted populations of common myeloid progenitors and granulocyte-monocyte progenitors were transduced with the oncogenes MOZ-TIF2 and BCR-ABL, respectively. MOZ-TIF2-transduced progenitors could be serially replated in methylcellulose cultures and continuously propagated in liquid culture, and resulted in an acute myeloid leukemia in vivo that could be serially transplanted. In contrast, BCR-ABL transduction conferred none of these properties to hematopoietic progenitors. These data demonstrate that some, but not all, leukemia oncogenes can confer properties of leukemic stem cells to hematopoietic progenitors destined to undergo apoptotic cell death.


Assuntos
Transformação Celular Neoplásica/genética , Genes abl/fisiologia , Células-Tronco Hematopoéticas/patologia , Proteínas de Fusão Oncogênica/fisiologia , Doença Aguda , Animais , Southern Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Diferenciação Celular/genética , Linhagem da Célula , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Ensaio de Unidades Formadoras de Colônias , Citometria de Fluxo , Genes abl/genética , Células Precursoras de Granulócitos/metabolismo , Células Precursoras de Granulócitos/patologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunofenotipagem , Interleucina-3/farmacologia , Leucemia Mieloide/genética , Leucemia Mieloide/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mutação , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas de Fusão Oncogênica/genética
19.
Cancer Cell ; 3(3): 259-71, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12676584

RESUMO

The MOZ-TIF2 fusion is associated with acute myeloid leukemia (AML) with inv(8)(p11q13). MOZ is a MYST family histone acetyltransferase (HAT), whereas TIF2 is a nuclear receptor coactivator that associates with CREB binding protein (CBP). Here we demonstrate that MOZ-TIF2 has transforming properties in vitro and causes AML in a murine bone marrow transplant assay. The C2HC nucleosome recognition motif of MOZ is essential for transformation, whereas MOZ HAT activity is dispensable. However, MOZ-TIF2 interaction with CBP through the TIF2 CBP interaction domain (CID) is essential for transformation. These results indicate that nucleosomal targeting by MOZ and recruitment of CBP by TIF2 are critical requirements for MOZ-TIF2 transformation and indicate that MOZ gain of function contributes to leukemogenesis.


Assuntos
Acetiltransferases/metabolismo , Leucemia Mieloide Aguda/genética , Nucleossomos/metabolismo , Fatores de Transcrição/metabolismo , Acetiltransferases/química , Acetiltransferases/genética , Animais , Transplante de Medula Óssea , Linhagem Celular , Transformação Celular Viral , Inversão Cromossômica , Cromossomos Humanos Par 8 , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Transplante de Células-Tronco Hematopoéticas , Histona Acetiltransferases , Humanos , Camundongos , Modelos Moleculares , Proteínas Nucleares/genética , Coativador 2 de Receptor Nuclear , Nucleossomos/química , Mutação Puntual , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Dedos de Zinco/genética
20.
Cancer Cell ; 3(5): 459-69, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12781364

RESUMO

FIP1L1-PDGFRalpha causes hypereosinophilic syndrome (HES) and is inhibited by the tyrosine kinase inhibitor imatinib (Gleevec). Imatinib is a potent inhibitor of ABL, ARG, PDGFRalpha, PDGFRbeta, and KIT and induces durable hematologic responses in HES patients. However, we observed relapse with resistance to imatinib as consequence of a T674I mutation in FIP1L1-PDGFRalpha, analogous to the imatinib-resistant T315I mutation in BCR-ABL. We developed a murine bone marrow transplant model of FIP1L1-PDGFRalpha-induced myeloproliferative disease to evaluate the efficacy of PKC412, an alternative inhibitor of PDGFRalpha, for the treatment of HES. PKC412 is effective for treatment of FIP1L1-PDGFRalpha-induced disease and of imatinib-induced resistance due to the T674I mutation. Our data establish PKC412 as molecularly targeted therapy for HES and other diseases expressing activated PDGFRalpha and demonstrate the potential of alternative kinase inhibitors to overcome resistance in target tyrosine kinases.


Assuntos
Transtornos Mieloproliferativos/tratamento farmacológico , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Estaurosporina/análogos & derivados , Estaurosporina/uso terapêutico , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo , Animais , Antineoplásicos/farmacologia , Benzamidas , Western Blotting , Medula Óssea/patologia , Transplante de Medula Óssea , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Proteínas de Fusão bcr-abl/metabolismo , Vetores Genéticos , Humanos , Mesilato de Imatinib , Imunofenotipagem , Camundongos , Modelos Genéticos , Mutação , Testes de Precipitina , Recidiva , Retroviridae/genética , Baço/citologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA