Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 13(11): 1072-82, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22983360

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) are regulated by various bone marrow stromal cell types. Here we identified rare activated bone marrow monocytes and macrophages with high expression of α-smooth muscle actin (α-SMA) and the cyclooxygenase COX-2 that were adjacent to primitive HSPCs. These myeloid cells resisted radiation-induced cell death and further upregulated COX-2 expression under stress conditions. COX-2-derived prostaglandin E(2) (PGE(2)) prevented HSPC exhaustion by limiting the production of reactive oxygen species (ROS) via inhibition of the kinase Akt and higher stromal-cell expression of the chemokine CXCL12, which is essential for stem-cell quiescence. Our study identifies a previously unknown subset of α-SMA(+) activated monocytes and macrophages that maintain HSPCs and protect them from exhaustion during alarm situations.


Assuntos
Actinas/imunologia , Medula Óssea/imunologia , Células-Tronco Hematopoéticas/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Actinas/genética , Animais , Medula Óssea/metabolismo , Medula Óssea/efeitos da radiação , Comunicação Celular/genética , Comunicação Celular/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Sobrevivência Celular/efeitos da radiação , Quimiocina CXCL12/genética , Quimiocina CXCL12/imunologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Dinoprostona/biossíntese , Dinoprostona/imunologia , Raios gama , Regulação da Expressão Gênica/imunologia , Regulação da Expressão Gênica/efeitos da radiação , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos da radiação , Macrófagos/citologia , Macrófagos/efeitos da radiação , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/efeitos da radiação , Camundongos , Monócitos/citologia , Monócitos/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transdução de Sinais/efeitos da radiação
2.
Immunity ; 43(4): 776-87, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26384546

RESUMO

Emerging evidence suggests that immunological mechanisms underlie metabolic control of adipose tissue. Here, we have shown the regulatory impact of a rare subpopulation of dendritic cells, rich in perforin-containing granules (perf-DCs). Using bone marrow transplantation to generate animals selectively lacking perf-DCs, we found that these chimeras progressively gained weight and exhibited features of metabolic syndrome. This phenotype was associated with an altered repertoire of T cells residing in adipose tissue and could be completely prevented by T cell depletion in vivo. A similar impact of perf-DCs on inflammatory T cells was also found in a well-defined model of multiple sclerosis, experimental autoimmune encephlalomyelitis (EAE). Thus, perf-DCs probably represent a regulatory cell subpopulation critical for protection from metabolic syndrome and autoimmunity.


Assuntos
Autoimunidade/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Inflamação/imunologia , Síndrome Metabólica/imunologia , Proteínas Citotóxicas Formadoras de Poros/análise , Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Transferência Adotiva , Animais , Antígenos de Diferenciação/análise , Antígeno CD11c/análise , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/transplante , Células Clonais/imunologia , Grânulos Citoplasmáticos/química , Células Dendríticas/classificação , Células Dendríticas/ultraestrutura , Dieta Hiperlipídica/efeitos adversos , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Inflamação/patologia , Depleção Linfocítica , Masculino , Síndrome Metabólica/etiologia , Síndrome Metabólica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/imunologia , Obesidade/patologia , Fenótipo , Proteínas Citotóxicas Formadoras de Poros/deficiência , Proteínas Citotóxicas Formadoras de Poros/genética , Quimera por Radiação , Tolerância a Antígenos Próprios/imunologia
3.
Blood ; 121(7): 1220-8, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23223359

RESUMO

Transplantation of T cell-depleted BM (TDBM) under mild conditioning, associated with minimal toxicity and reduced risk of GVHD, offers an attractive therapeutic option for patients with nonmalignant hematologic disorders and can mediate immune tolerance to subsequent organ transplantation. However, overcoming TDBM rejection after reduced conditioning remains a challenge. Here, we address this barrier using donorderived central memory CD8(+) T cells (Tcms), directed against third-party antigens. Our results show that fully allogeneic or (hostXdonor)F1-Tcm, support donor chimerism (> 6 months) in sublethally irradiated (5.5Gy) mice, without GVHD symptoms. Chimerism under yet lower irradiation (4.5Gy) was achieved by combining Tcm with short-term administration of low-dose Rapamycin. Importantly, this chimerism resulted in successful donor skin acceptance, whereas third-party skin was rejected. Tracking of host anti-donor T cells (HADTCs), that mediate TDBMT rejection, in a novel bioluminescence-imaging model revealed that Tcms both induce accumulation and eradicate HADTCs in the LNs,concomitant with their elimination from other organs, including the BM. Further analysis with 2-photon microcopy revealed that Tcms form conjugates with HADTCs, resulting in decelerated and confined movement of HADTCs within the LNs in an antigen-specific manner. Thus, anti-third-party Tcms support TDBMT engraftment under reduced-conditioning through lymph-node sequestration and deletion of HADTCs, offering a novel and potentially safe approach for attaining stable hematopoietic chimerism.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Quimeras de Transplante/imunologia , Condicionamento Pré-Transplante/métodos , Animais , Transplante de Medula Óssea/imunologia , Feminino , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Doenças Hematológicas/imunologia , Doenças Hematológicas/terapia , Humanos , Memória Imunológica , Imunossupressores/administração & dosagem , Isoantígenos , Linfonodos/imunologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Sirolimo/administração & dosagem , Transplante de Pele/imunologia , Linfócitos T/imunologia , Doadores de Tecidos
4.
Blood ; 122(15): 2562-71, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23982175

RESUMO

Analysis of hematopoietic stem cells (HSCs) in factor VIII knockout (FVIIIKO) mice revealed a novel regulatory role for the coagulation cascade in hematopoiesis. Thus, HSCs in FVIIIKO mice had reduced proportions of CD34(low) cells within Lin(-)Sca(+)Kit(+) progenitors, and exhibited reduced long-term repopulating capacity as well as hyper granulocyte-colony-stimulating factor (G-CSF)-induced mobilization. This disregulation of HSCs is likely caused by reduced levels of thrombin, and is associated with altered protease-activated receptor 1 (PAR1) signaling, as PAR1 KO mice also exhibited enhanced G-CSF-induced mobilization. Analysis of reciprocal bone marrow (BM) chimera (FVIIIKO BM into wild-type recipients and vice versa) and the detection of PAR1 expression on stromal elements indicates that this phenotype is likely controlled by stromal elements. Micro-computed tomography analysis of distal tibia metaphyses also revealed for the first time a major impact of the FVIII/thrombin/PAR1 axis on the dynamic bone structure, showing reduced bone:tissue volume ratio and trabecular number in FVIIIKO and PAR1KO mice. Taken together, these results show a critical and novel role for the coagulation cascade, mediated in part by thrombin-PAR1 interaction, and regulates HSC maintenance and a reciprocal interplay between HSCs and the dynamic bone structure.


Assuntos
Osso e Ossos/fisiologia , Fator VIII/fisiologia , Hematopoese/fisiologia , Receptor PAR-1/fisiologia , Trombina/fisiologia , Animais , Coagulação Sanguínea/fisiologia , Osso e Ossos/diagnóstico por imagem , Fator VIII/genética , Fator VIII/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Transdução de Sinais/fisiologia , Células Estromais/citologia , Células Estromais/fisiologia , Trombina/metabolismo , Microtomografia por Raio-X
5.
Blood ; 120(9): 1843-55, 2012 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-22645180

RESUMO

Cytokine-induced expansion of hematopoietic stem and progenitor cells (HSPCs) is not fully understood. In the present study, we show that whereas steady-state hematopoiesis is normal in basic fibroblast growth factor (FGF-2)-knockout mice, parathyroid hormone stimulation and myeloablative treatments failed to induce normal HSPC proliferation and recovery. In vivo FGF-2 treatment expanded stromal cells, including perivascular Nestin(+) supportive stromal cells, which may facilitate HSPC expansion by increasing SCF and reducing CXCL12 via mir-31 up-regulation. FGF-2 predominantly expanded a heterogeneous population of undifferentiated HSPCs, preserving and increasing durable short- and long-term repopulation potential. Mechanistically, these effects were mediated by c-Kit receptor activation, STAT5 phosphorylation, and reduction of reactive oxygen species levels. Mice harboring defective c-Kit signaling exhibited abrogated HSPC expansion in response to FGF-2 treatment, which was accompanied by elevated reactive oxygen species levels. The results of the present study reveal a novel mechanism underlying FGF-2-mediated in vivo expansion of both HSPCs and their supportive stromal cells, which may be used to improve stem cell engraftment after clinical transplantation.


Assuntos
Proliferação de Células , Quimiocina CXCL12/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células Estromais/metabolismo , Animais , Sequência de Bases , Transplante de Medula Óssea , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL12/genética , Regulação para Baixo/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Hormônio Paratireóideo/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/genética , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/metabolismo , Células Estromais/efeitos dos fármacos
6.
Blood ; 120(8): 1647-57, 2012 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-22776817

RESUMO

Immature dendritic cells (imDCs) can have a tolerizing effect under normal conditions or after transplantation. However, because of the significant heterogeneity of this cell population, it is extremely difficult to study the mechanisms that mediate the tolerance induced or to harness the application of imDCs for clinical use. In the present study, we describe the generation of a highly defined population of imDCs from hematopoietic progenitors and the direct visualization of the fate of TCR-transgenic alloreactive CD4(+) and CD8(+) T cells after encountering cognate or noncognate imDCs. Whereas CD4(+) T cells were deleted via an MHC-independent mechanism through the NO system, CD8(+) T-cell deletion was found to occur through a unique MHC-dependent, perforin-based killing mechanism involving activation of TLR7 and signaling through Triggering Receptor-1 Expressed on Myeloid cells (TREM-1). This novel subpopulation of perforin-expressing imDCs was also detected in various lymphoid tissues in normal animals and its frequency was markedly enhanced after GM-CSF administration.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Granzimas/imunologia , Células-Tronco Hematopoéticas/imunologia , Glicoproteínas de Membrana/imunologia , Perforina/imunologia , Receptores Imunológicos/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Técnicas de Cultura de Células/métodos , Células Cultivadas , Células Dendríticas/citologia , Feminino , Células-Tronco Hematopoéticas/citologia , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos C57BL , Receptor Gatilho 1 Expresso em Células Mieloides , Quinases da Família src/imunologia
7.
Proc Natl Acad Sci U S A ; 108(19): 7956-61, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21518888

RESUMO

Rearrangements of the MLL (ALL1) gene are very common in acute infant and therapy-associated leukemias. The rearrangements underlie the generation of MLL fusion proteins acting as potent oncogenes. Several most consistently up-regulated targets of MLL fusions, MEIS1, HOXA7, HOXA9, and HOXA10 are functionally related and have been implicated in other types of leukemias. Each of the four genes was knocked down separately in the human precursor B-cell leukemic line RS4;11 expressing MLL-AF4. The mutant and control cells were compared for engraftment in NOD/SCID mice. Engraftment of all mutants into the bone marrow (BM) was impaired. Although homing was similar, colonization by the knockdown cells was slowed. Initially, both types of cells were confined to the trabecular area; this was followed by a rapid spread of the WT cells to the compact bone area, contrasted with a significantly slower process for the mutants. In vitro and in vivo BrdU incorporation experiments indicated reduced proliferation of the mutant cells. In addition, the CXCR4/SDF-1 axis was hampered, as evidenced by reduced migration toward an SDF-1 gradient and loss of SDF-1-augmented proliferation in culture. The very similar phenotype shared by all mutant lines implies that all four genes are involved and required for expansion of MLL-AF4 associated leukemic cells in mice, and down-regulation of any of them is not compensated by the others.


Assuntos
Genes Homeobox , Proteínas de Homeodomínio/genética , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Técnicas de Silenciamento de Genes , Rearranjo Gênico , Histona-Lisina N-Metiltransferase , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteína Meis1 , Transplante de Neoplasias , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , RNA Interferente Pequeno/genética , Transplante Heterólogo
8.
Blood ; 118(22): e156-67, 2011 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21951685

RESUMO

The CX3C chemokine family is composed of only one member, CX3CL1, also known as fractalkine, which in mice is the sole ligand of the G protein-coupled, 7-transmembrane receptor CX3CR1. Unlike classic small peptide chemokines, CX3CL1 is synthesized as a membrane-anchored protein that can promote integrin-independent adhesion. Subsequent cleavage by metalloproteases, either constitutive or induced, can generate shed CX3CL1 entities that potentially have chemoattractive activity. To study the CX3C interface in tissues of live animals, we generated transgenic mice (CX3CL1cherry:CX3CR1gfp), which express red and green fluorescent reporter genes under the respective control of the CX3CL1 and CX3CR1 promoters. Furthermore, we performed a structure/function analysis to differentiate the in vivo functions of membrane-tethered versus shed CX3CL1 moieties by comparing their respective ability to correct established defects in macrophage function and leukocyte survival in CX3CL1-deficient mice. Specifically, expression of CX3CL1(105Δ), an obligatory soluble CX3CL1 isoform, reconstituted the formation of transepithelial dendrites by intestinal macrophages but did not rescue circulating Ly6Clo CX3CR1hi blood monocytes in CX3CR1gfp/gfp mice. Instead, monocyte survival required the full-length membrane-anchored CX3CL1, suggesting differential activities of tethered and shed CX3CL1 entities.


Assuntos
Quimiocina CX3CL1/química , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/fisiologia , Animais , Células Cultivadas , Quimiocina CX3CL1/metabolismo , Quimiocinas CX3C/química , Quimiocinas CX3C/genética , Quimiocinas CX3C/metabolismo , Quimiocinas CX3C/fisiologia , Feminino , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiologia , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Relação Estrutura-Atividade
9.
Blood ; 117(3): 1042-52, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21045195

RESUMO

Cytotoxic T lymphocytes (CTLs) suppress T cell responses directed against their antigens regardless of their own T cell receptor (TCR) specificity. This makes the use of CTLs promising for tolerance induction in autoimmunity and transplantation. It has been established that binding of the CTL CD8 molecule to the major histocompatibility complex (MHC) class I α3 domain of the recognizing T cell must be permitted for death of the latter cell to ensue. However, the signaling events triggered in the CTL by this molecular interaction in the absence of TCR recognition have never been clarified. Here we use single-cell imaging to study the events occurring in CTLs serving as targets for recognition by specific T cells. We demonstrate that CTLs actively respond to recognition by polarizing their cytotoxic granules to the contact area, releasing their lethal cargo, and vigorously proliferating. Using CTLs from perforin knockout (KO) mice and lymphocyte specific kinase (Lck) knockdown with specific small interfering RNA (siRNA), we show that the killing of the recognizing CD8 T cell is perforin dependent and is initiated by Lck signaling in the CTL. Collectively, these data suggest a novel mechanism in which the entire cascade generally triggered by TCR engagement is "hijacked" in CTLs serving as targets for T cell recognition without TCR ligation.


Assuntos
Grânulos Citoplasmáticos/imunologia , Ativação Linfocitária/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Grânulos Citoplasmáticos/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/imunologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Citometria de Fluxo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Camundongos Knockout , Modelos Imunológicos , Ovalbumina/farmacologia , Fragmentos de Peptídeos/farmacologia , Perforina/genética , Perforina/metabolismo , Interferência de RNA , Linfócitos T/metabolismo , Linfócitos T Citotóxicos/metabolismo
10.
Blood ; 117(2): 419-28, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20585044

RESUMO

Mechanisms governing stress-induced hematopoietic progenitor cell mobilization are not fully deciphered. We report that during granulocyte colony-stimulating factor-induced mobilization c-Met expression and signaling are up-regulated on immature bone marrow progenitors. Interestingly, stromal cell-derived factor 1/CXC chemokine receptor-4 signaling induced hepatocyte growth factor production and c-Met activation. We found that c-Met inhibition reduced mobilization of both immature progenitors and the more primitive Sca-1(+)/c-Kit(+)/Lin(-) cells and interfered with their enhanced chemotactic migration to stromal cell-derived factor 1. c-Met activation resulted in cellular accumulation of reactive oxygen species by mammalian target of rapamycin inhibition of Forkhead Box, subclass O3a. Blockage of mammalian target of rapamycin inhibition or reactive oxygen species signaling impaired c-Met-mediated mobilization. Our data show dynamic c-Met expression and function in the bone marrow and show that enhanced c-Met signaling is crucial to facilitate stress-induced mobilization of progenitor cells as part of host defense and repair mechanisms.


Assuntos
Movimento Celular/fisiologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Animais , Quimiocina CXCL12/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Fator de Crescimento de Hepatócito/metabolismo , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Proc Natl Acad Sci U S A ; 106(21): 8659-64, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19433788

RESUMO

Xenotransplantation of pig tissues has great potential to overcome the shortage of organ donors. One approach to address the vigorous immune rejection associated with xenotransplants is the use of embryonic precursor tissue, which induces and utilizes host vasculature upon its growth and development. Recently, we showed in mice that embryonic pig pancreatic tissue from embryonic day 42 (E42) exhibits optimal properties as a beta cell replacement therapy. We now demonstrate the proof of concept in 2 diabetic Cynomolgus monkeys, followed for 393 and 280 days, respectively. A marked reduction of exogenous insulin requirement was noted by the fourth month after transplantation, reaching complete independence from exogenous insulin during the fifth month after transplantation, with full physiological control of blood glucose levels. The porcine origin of insulin was documented by a radioimmunoassay specific for porcine C-peptide. Furthermore, the growing tissue was found to be predominantly vascularized with host blood vessels, thereby evading hyperacute or acute rejection, which could potentially be mediated by preexisting anti-pig antibodies. Durable graft protection was achieved, and most of the late complications could be attributed to the immunosuppressive protocol. While fine tuning of immune suppression, tissue dose, and implantation techniques are still required, our results demonstrate that porcine E-42 embryonic pancreatic tissue can normalize blood glucose levels in primates. Its long-term proliferative capacity, its revascularization by host endothelium, and its reduced immunogenicity, strongly suggest that this approach could offer an attractive replacement therapy for diabetes.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Pâncreas/embriologia , Pâncreas/cirurgia , Suínos/embriologia , Suínos/cirurgia , Transplante Heterólogo , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Rejeição de Enxerto/imunologia , Macaca fascicularis , Masculino , Pâncreas/irrigação sanguínea , Pâncreas/imunologia , Transplante de Pâncreas , Estreptozocina/farmacologia , Transplante Heterólogo/imunologia
12.
Stem Cells Transl Med ; 11(2): 178-188, 2022 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-35298657

RESUMO

Over the last decades, several studies demonstrated the possibility of lung regeneration through transplantation of various lung progenitor populations. Recently, we showed in mice that fetal or adult lung progenitors could potentially provide donor cells for transplantation, provided that the lung stem cell niche in the recipient is vacated of endogenous lung progenitors by adequate conditioning. Accordingly, marked lung regeneration could be attained following i.v. infusion of a single cell suspension of lung cells into recipient mice conditioned with naphthalene (NA) and 6Gy total body irradiation (TBI). As clinical translation of this approach requires the use of allogenic donors, we more recently developed a novel transplantation modality based on co-infusion of hematopoietic and lung progenitors from the same donor. Thus, by virtue of hematopoietic chimerism, which leads to immune tolerance toward donor antigens, the lung progenitors can be successfully engrafted without any need for post-transplant immune suppression. In the present study, we demonstrate that it is possible to replace NA in the conditioning regimen with Cyclophosphamide (CY), approved for the treatment of many diseases and that a lower dose of 2 GY TBI can successfully enable engraftment of donor-derived hematopoietic and lung progenitors when CY is administered in 2 doses after the stem cell infusion. Taken together, our results suggest a feasible and relatively safe protocol that could potentially be translated to clinical transplantation of lung progenitors across major MHC barriers in patients with terminal lung diseases.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Condicionamento Pré-Transplante , Animais , Ciclofosfamida , Humanos , Indicadores e Reagentes , Pulmão , Camundongos , Quimeras de Transplante , Condicionamento Pré-Transplante/métodos
13.
Cell Rep ; 30(3): 807-819.e4, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31968255

RESUMO

Induction of lung regeneration by transplantation of lung progenitor cells is a critical preclinical challenge. Recently, we demonstrated that robust lung regeneration can be achieved if the endogenous stem cell niches in the recipient's lung are vacated by sub-lethal pre-conditioning. However, overcoming MHC barriers is an additional requirement for clinical application of this attractive approach. We demonstrate here that durable tolerance toward mis-matched lung progenitors and their derivatives can be achieved without any chronic immune suppression, by virtue of co-transplantation with hematopoietic progenitors from the same donor. Initial proof of concept of this approach was attained by transplantation of fetal lung cells comprising both hematopoietic and non-hematopoietic progenitors. Furthermore, an even higher rate of blood and epithelial lung chimerism was attained by using adult lung cells supplemented with bone marrow hematopoietic progenitors. These results lay the foundation for repair of lung injury through a procedure akin to bone marrow transplantation.


Assuntos
Linhagem da Célula/genética , Pulmão/fisiologia , Regeneração/genética , Transplante de Células-Tronco , Células-Tronco Adultas/citologia , Animais , Autorrenovação Celular , Quimera , Feto/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Tolerância Imunológica , Pulmão/embriologia , Camundongos Endogâmicos C57BL , Doadores de Tecidos , Condicionamento Pré-Transplante
14.
Stem Cells ; 26(5): 1347-55, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18339772

RESUMO

Cell therapy as an alternative to orthotopic liver transplantation represents a major challenge, since negligible proliferation of isolated hepatocytes occurs after transplantation because of the stringent homeostatic control displayed by the host liver. Thus, different modalities of liver injury as part of the pretransplant conditioning are a prerequisite for this approach. The major objective of the present study was to test whether xenotransplantation of pig fetal liver fragments, in which potential cell-cell and cell-stroma interactions are spared, might afford more robust growth and proliferation compared with isolated pig fetal hepatoblasts. After transplantation into SCID mice, fetal liver tissue fragments exhibited marked growth and proliferation, in the setting of a quiescent host liver, compared with isolated fetal hepatoblasts harvested at the same gestational age (embryonic day 28). The proliferative advantage of fetal pig liver fragments was clearly demonstrated by immunohistochemical and morphometric assays and was observed not only after implantation into the liver but also into extrahepatic sites, such as the spleen and the subrenal capsule. The presence of all types of nonparenchymal liver cells that is crucial for normal liver development and regeneration was demonstrated in the implants. Preservation of the three-dimensional structure in pig fetal liver fragments enables autonomous proliferation of transplanted hepatic cells in the setting of a quiescent host liver, without any requirement for liver injury in the pretransplant conditioning. The marked proliferation and functional maturation exhibited by the pig fetal liver fragments suggests that it could afford a preferable source for transplantation.


Assuntos
Hepatócitos/citologia , Homeostase , Transplante de Fígado , Fígado/citologia , Fígado/embriologia , Animais , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Epiteliais/citologia , Regulação da Expressão Gênica , Fígado/irrigação sanguínea , Mesoderma/citologia , Camundongos , Camundongos SCID , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células-Tronco/citologia , Suínos , Transplante Heterólogo
15.
Stem Cells Transl Med ; 7(1): 68-77, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29266820

RESUMO

Repair of injured lungs represents a longstanding therapeutic challenge. We recently demonstrated that human and mouse embryonic lung tissue from the canalicular stage of development are enriched with lung progenitors, and that a single cell suspension of canalicular lungs can be used for transplantation, provided that lung progenitor niches in the recipient mice are vacated by strategies similar to those used in bone marrow transplantation. Considering the ethical limitations associated with the use of fetal cells, we investigated here whether adult lungs could offer an alternative source of lung progenitors for transplantation. We show that intravenous infusion of a single cell suspension of adult mouse lungs from GFP+ donors, following conditioning of recipient mice with naphthalene and subsequent sublethal irradiation, led to marked colonization of the recipient lungs, at 6-8 weeks post-transplant, with donor derived structures including epithelial, endothelial, and mesenchymal cells. Epithelial cells within these donor-derived colonies expressed markers of functionally distinct lung cell types, and lung function, which is significantly compromised in mice treated with naphthalene and radiation, was found to be corrected following transplantation. Dose response analysis suggests that the frequency of patch forming cells in adult lungs was about threefold lower compared to that found in E16 fetal lungs. However, as adult lungs are much larger, the total number of patch forming cells that can be collected from this source is significantly greater. Our study provides proof of concept for lung regeneration by adult lung cells after preconditioning to vacate the pulmonary niche. Stem Cells Translational Medicine 2018;7:68-77.


Assuntos
Células Epiteliais/transplante , Regeneração Tecidual Guiada/métodos , Lesão Pulmonar/terapia , Pulmão/citologia , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Células Cultivadas , Células Epiteliais/citologia , Feminino , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Naftalenos/toxicidade
16.
PLoS Med ; 3(7): e215, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16768546

RESUMO

BACKGROUND: Transplantation of embryonic pig pancreatic tissue as a source of insulin has been suggested for the cure of diabetes. However, previous limited clinical trials failed in their attempts to treat diabetic patients by transplantation of advanced gestational age porcine embryonic pancreas. In the present study we examined growth potential, functionality, and immunogenicity of pig embryonic pancreatic tissue harvested at different gestational ages. METHODS AND FINDINGS: Implantation of embryonic pig pancreatic tissues of different gestational ages in SCID mice reveals that embryonic day 42 (E42) pig pancreas can enable a massive growth of pig islets for prolonged periods and restore normoglycemia in diabetic mice. Furthermore, both direct and indirect T cell rejection responses to the xenogeneic tissue demonstrated that E42 tissue, in comparison to E56 or later embryonic tissues, exhibits markedly reduced immunogenicity. Finally, fully immunocompetent diabetic mice grafted with the E42 pig pancreatic tissue and treated with an immunosuppression protocol comprising CTLA4-Ig and anti-CD40 ligand (anti-CD40L) attained normal blood glucose levels, eliminating the need for insulin. CONCLUSIONS: These results emphasize the importance of selecting embryonic tissue of the correct gestational age for optimal growth and function and for reduced immunogenicity, and provide a proof of principle for the therapeutic potential of E42 embryonic pig pancreatic tissue transplantation in diabetes.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 1/cirurgia , Transplante de Pâncreas , Transplante Heterólogo , Abatacepte , Tirosina Quinase da Agamaglobulinemia , Aloxano , Animais , Glicemia/análise , Ligante de CD40/antagonistas & inibidores , Feminino , Idade Gestacional , Rejeição de Enxerto/prevenção & controle , Humanos , Imunocompetência , Imunoconjugados/uso terapêutico , Imunossupressores/uso terapêutico , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/ultraestrutura , Rim , Leucócitos Mononucleares/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos Nus , Camundongos SCID , Pâncreas/embriologia , Transplante de Pâncreas/imunologia , Pâncreas Exócrino/ultraestrutura , Gravidez , Proteínas Tirosina Quinases/deficiência , Sus scrofa/embriologia , Transplante Heterólogo/imunologia , Transplante Heterotópico/imunologia
17.
J Neuroimmunol ; 301: 7-11, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27836179

RESUMO

The lack of biomarkers is a major obstacle for investigating myelin repair. We used metabolic incorporation of the choline analog - propargyl-choline (P-Cho) to label and visualize newly synthesized myelin in the CNS of mice induced with experimental autoimmune encephalomyelitis (EAE). We further developed unbiased colocalization analysis to quantify P-Cho incorporation specifically into the myelin. Our findings indicate that P-Cho injection to mice recovering from EAE, either spontaneously or following glatiramer acetate treatment, results in significant elevation of its incorporation into the myelin, offering a novel strategy for assessing remyelination in animal models and the remyelination potential of candidate drugs.


Assuntos
Encefalomielite Autoimune Experimental/patologia , Bainha de Mielina/metabolismo , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/patologia , Alcinos/metabolismo , Animais , Nucléolo Celular/metabolismo , Nucléolo Celular/patologia , Colina/análogos & derivados , Colina/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/diagnóstico por imagem , Encefalomielite Autoimune Experimental/tratamento farmacológico , Acetato de Glatiramer/uso terapêutico , Imunossupressores/uso terapêutico , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Imunoeletrônica , Proteína Básica da Mielina/metabolismo , Proteína Proteolipídica de Mielina/toxicidade , Bainha de Mielina/ultraestrutura , Fragmentos de Peptídeos/toxicidade , Recuperação de Função Fisiológica/efeitos dos fármacos , Medula Espinal/ultraestrutura
18.
Inflamm Bowel Dis ; 22(2): 257-67, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26529559

RESUMO

The continuous recirculation of mature lymphocytes and their entry into the peripheral lymph nodes are crucial for the development of an immune response to foreign antigens. Occasionally, the entry and the subsequent response of T lymphocytes in these sites lead to severe inflammation and pathological conditions. Here, we characterized the tetraspanin molecule, CD151, as a regulator of T cell motility in health and in models of inflammatory bowel disease. CD151 formed a cell surface complex with VLA-4 and LFA-1 integrins, and its activation led to enhanced migration of T cells. Picomolar levels of CCL2 that were previously shown to inhibit T-cell migration to lymph nodes suppressed CD151 expression and dissociated CD151-integrin complexes in T lymphocytes, resulting in attenuated migration toward T-cell attractant chemokines. To directly inhibit CD151 function, a truncated CD151 peptide fragment mimicking of the CD151 extracellular loop was designed. CD151 extracellular loop inhibited T-cell migration in vitro and in vivo and attenuated the development of dextrane sulfate sodium-induced colitis. Thus, CD151 is a key orchestrator of T cell motility; interference with its proper function results in attenuated progression of inflammatory bowel disease.


Assuntos
Movimento Celular/imunologia , Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Inflamação/imunologia , Linfócitos T/imunologia , Tetraspanina 24/fisiologia , Animais , Estudos de Casos e Controles , Movimento Celular/fisiologia , Seguimentos , Proteínas de Homeodomínio/fisiologia , Humanos , Antígenos Comuns de Leucócito/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prognóstico , Receptores CCR2/fisiologia
19.
J Thorac Oncol ; 10(1): 46-58, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25654727

RESUMO

BACKGROUND: The existing shortage of animal models that properly mimic the progression of early-stage human lung cancer from a solitary confined tumor to an invasive metastatic disease hinders accurate characterization of key interactions between lung cancer cells and their stroma. We herein describe a novel orthotopic animal model that addresses these concerns and consequently serves as an attractive platform to study tumor-stromal cell interactions under conditions that reflect early-stage lung cancer. METHODS: Unlike previous methodologies, we directly injected small numbers of human or murine lung cancer cells into murine's left lung and longitudinally monitored disease progression. Next, we used green fluorescent protein-tagged tumor cells and immuno-fluorescent staining to determine the tumor's microanatomic distribution and to look for tumor-infiltrating immune cells and stromal cells. Finally, we compared chemokine gene expression patterns in the tumor and lung microenvironment. RESULTS: We successfully generated a solitary pulmonary nodule surrounded by normal lung parenchyma that grew locally and spread distally over time. Notably, we found that both fibroblasts and leukocytes are recruited to the tumor's margins and that distinct myeloid cell attracting and CCR2-binding chemokines are specifically induced in the tumor microenvironment. CONCLUSION: Our orthotopic lung cancer model closely mimics the pathologic sequence of events that characterizes early-stage human lung cancer propagation. It further introduces new means to monitor tumor-stromal cell interactions and offers unique opportunities to test therapeutic targets under conditions that reflect early-stage lung cancer. We argue that for such purposes our model is superior to lung cancer models that are based either on genetic induction of epithelial transformation or on ectopic transplantation of malignant cells.


Assuntos
Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/terapia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Modelos Animais de Doenças , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Transplante Heterólogo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Nat Med ; 21(8): 869-79, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26168294

RESUMO

Repair of injured lungs represents a longstanding therapeutic challenge. We show that human and mouse embryonic lung tissue from the canalicular stage of development (20-22 weeks of gestation for humans, and embryonic day 15-16 (E15-E16) for mouse) are enriched with progenitors residing in distinct niches. On the basis of the marked analogy to progenitor niches in bone marrow (BM), we attempted strategies similar to BM transplantation, employing sublethal radiation to vacate lung progenitor niches and to reduce stem cell competition. Intravenous infusion of a single cell suspension of canalicular lung tissue from GFP-marked mice or human fetal donors into naphthalene-injured and irradiated syngeneic or SCID mice, respectively, induced marked long-term lung chimerism. Donor type structures or 'patches' contained epithelial, mesenchymal and endothelial cells. Transplantation of differentially labeled E16 mouse lung cells indicated that these patches were probably of clonal origin from the donor. Recipients of the single cell suspension transplant exhibited marked improvement in lung compliance and tissue damping reflecting the energy dissipation in the lung tissues. Our study provides proof of concept for lung reconstitution by canalicular-stage human lung cells after preconditioning of the pulmonary niche.


Assuntos
Células-Tronco Embrionárias/transplante , Pulmão/embriologia , Condicionamento Pré-Transplante , Animais , Bromodesoxiuridina/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos SCID , Regeneração , Quimeras de Transplante , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA