Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 46(5): 773-775, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28514684

RESUMO

Normalization of tumor blood vessels enhances the infiltration and functions of T cells. Tian et al. (2017) report that effector CD4+ T cells, in turn, support vascular normalization, highlighting intertwined roles for blood vessels and T cells in cancer.


Assuntos
Ativação Linfocitária , Linfócitos T/imunologia , Linfócitos T CD4-Positivos/imunologia , Humanos , Neoplasias
2.
Proc Natl Acad Sci U S A ; 120(9): e2210836120, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36821580

RESUMO

Defining the ontogeny of tumor-associated macrophages (TAM) is important to develop therapeutic targets for mesothelioma. We identified two distinct macrophage populations in mouse peritoneal and pleural cavities, the monocyte-derived, small peritoneal/pleural macrophages (SPM), and the tissue-resident large peritoneal/pleural macrophages (LPM). SPM rapidly increased in tumor microenvironment after tumor challenge and contributed to the vast majority of M2-like TAM. The selective depletion of M2-like TAM by conditional deletion of the Dicer1 gene in myeloid cells (D-/-) promoted tumor rejection. Sorted SPM M2-like TAM initiated tumorigenesis in vivo and in vitro, confirming their capacity to support tumor development. The transcriptomic and single-cell RNA sequencing analysis demonstrated that both SPM and LPM contributed to the tumor microenvironment by promoting the IL-2-STAT5 signaling pathway, inflammation, and epithelial-mesenchymal transition. However, while SPM preferentially activated the KRAS and TNF-α/NFkB signaling pathways, LPM activated the IFN-γ response. The importance of LPM in the immune response was confirmed by depleting LPM with intrapleural clodronate liposomes, which abrogated the antitumoral memory immunity. SPM gene signature could be identified in pleural effusion and tumor from patients with untreated mesothelioma. Five genes, TREM2, STAB1, LAIR1, GPNMB, and MARCO, could potentially be specific therapeutic targets. Accordingly, Trem2 gene deletion led to reduced SPM M2-like TAM with compensatory increase in LPM and slower tumor growth. Overall, these experiments demonstrate that SPM M2-like TAM play a key role in mesothelioma development, while LPM more specifically contribute to the immune response. Therefore, selective targeting of monocyte-derived TAM may enhance antitumor immunity through compensatory expansion of tissue-resident TAM.


Assuntos
Mesotelioma Maligno , Mesotelioma , Animais , Camundongos , Mesotelioma Maligno/metabolismo , Mesotelioma Maligno/patologia , Macrófagos Associados a Tumor/patologia , Macrófagos/metabolismo , Mesotelioma/metabolismo , Monócitos/patologia , Microambiente Tumoral , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(1): 541-551, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31889004

RESUMO

Cancer immunotherapies are increasingly combined with targeted therapies to improve therapeutic outcomes. We show that combination of agonistic anti-CD40 with antiangiogenic antibodies targeting 2 proangiogenic factors, vascular endothelial growth factor A (VEGFA) and angiopoietin 2 (Ang2/ANGPT2), induces pleiotropic immune mechanisms that facilitate tumor rejection in several tumor models. On the one hand, VEGFA/Ang2 blockade induced regression of the tumor microvasculature while decreasing the proportion of nonperfused vessels and reducing leakiness of the remaining vessels. On the other hand, both anti-VEGFA/Ang2 and anti-CD40 independently promoted proinflammatory macrophage skewing and increased dendritic cell activation in the tumor microenvironment, which were further amplified upon combination of the 2 treatments. Finally, combined therapy provoked brisk infiltration and intratumoral redistribution of cytotoxic CD8+ T cells in the tumors, which was mainly driven by Ang2 blockade. Overall, these nonredundant synergistic mechanisms endowed T cells with improved effector functions that were conducive to more efficient tumor control, underscoring the therapeutic potential of antiangiogenic immunotherapy in cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Antígenos CD40/agonistas , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Angiopoietina-2/antagonistas & inibidores , Angiopoietina-2/metabolismo , Animais , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígenos CD40/imunologia , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Sinergismo Farmacológico , Feminino , Humanos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Nat Methods ; 15(3): 183-186, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29355847

RESUMO

We describe a lentivirus-encoded chimeric receptor, termed extracellular vesicle (EV)-internalizing receptor (EVIR), which enables the selective uptake of cancer-cell-derived EVs by dendritic cells (DCs). The EVIR enhances DC presentation of EV-associated tumor antigens to CD8+ T cells primarily through MHCI recycling and cross-dressing. EVIRs should facilitate exploring the mechanisms and implications of horizontal transfer of tumor antigens to antigen-presenting cells.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Células Dendríticas/imunologia , Vesículas Extracelulares/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Animais , Células Cultivadas , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Vesículas Extracelulares/imunologia , Feminino , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Camundongos Endogâmicos C57BL
5.
J Pathol ; 250(5): 573-592, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32086811

RESUMO

Macrophages sustain tumour progression by facilitating angiogenesis, promoting immunosuppression, and enhancing cancer cell invasion and metastasis. They also modulate tumour response to anti-cancer therapy in pre-clinical models. This knowledge has motivated the development of agents that target tumour-associated macrophages (TAMs), some of which have been investigated in early clinical trials. Here, we provide a comprehensive overview of the biology and therapeutic targeting of TAMs, highlighting opportunities, setbacks, and new challenges that have emerged after a decade of intense translational and clinical research into these multifaceted immune cells. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Macrófagos/imunologia , Macrófagos/patologia , Neoplasias/patologia , Neovascularização Patológica/patologia , Microambiente Tumoral/imunologia , Humanos , Sistema Imunitário/patologia , Neoplasias/imunologia , Neovascularização Patológica/imunologia , Reino Unido
6.
7.
Angiogenesis ; 21(3): 425-532, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29766399

RESUMO

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Assuntos
Bioensaio/métodos , Neoplasias , Neovascularização Patológica , Animais , Bioensaio/instrumentação , Guias como Assunto , Humanos , Camundongos , Neoplasias/irrigação sanguínea , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
8.
Arterioscler Thromb Vasc Biol ; 36(11): 2203-2212, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27634833

RESUMO

OBJECTIVE: Perivascular cells, including pericytes, macrophages, smooth muscle cells, and other specialized cell types, like podocytes, participate in various aspects of vascular function. However, aside from the well-established roles of smooth muscle cells and pericytes, the contributions of other vascular-associated cells are poorly understood. Our goal was to ascertain the function of perivascular macrophages in adult tissues under nonpathological conditions. APPROACH AND RESULTS: We combined confocal microscopy, in vivo cell depletion, and in vitro assays to investigate the contribution of perivascular macrophages to vascular function. We found that resident perivascular macrophages are associated with capillaries at a frequency similar to that of pericytes. Macrophage depletion using either clodronate liposomes or antibodies unexpectedly resulted in hyperpermeability. This effect could be rescued when M2-like macrophages, but not M1-like macrophages or dendritic cells, were reconstituted in vivo, suggesting subtype-specific roles for macrophages in the regulation of vascular permeability. Furthermore, we found that permeability-promoting agents elicit motility and eventual dissociation of macrophages from the vasculature. Finally, in vitro assays showed that M2-like macrophages attenuate the phosphorylation of VE-cadherin upon exposure to permeability-promoting agents. CONCLUSIONS: This study points to a direct contribution of macrophages to vessel barrier integrity and provides evidence that heterotypic cell interactions with the endothelium, in addition to those of pericytes, control vascular permeability.


Assuntos
Capilares/metabolismo , Permeabilidade Capilar , Comunicação Celular , Células Endoteliais/metabolismo , Macrófagos Peritoneais/metabolismo , Mesentério/irrigação sanguínea , Peritônio/irrigação sanguínea , Pele/irrigação sanguínea , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Movimento Celular , Células Cultivadas , Técnicas de Cocultura , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Ovalbumina/metabolismo , Fenótipo , Fosforilação , Rodaminas/metabolismo , Fatores de Tempo , Transfecção
9.
Nature ; 479(7371): 122-6, 2011 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-21983962

RESUMO

PHD2 serves as an oxygen sensor that rescues blood supply by regulating vessel formation and shape in case of oxygen shortage. However, it is unknown whether PHD2 can influence arteriogenesis. Here we studied the role of PHD2 in collateral artery growth by using hindlimb ischaemia as a model, a process that compensates for the lack of blood flow in case of major arterial occlusion. We show that Phd2 (also known as Egln1) haplodeficient (Phd2(+/-)) mice displayed preformed collateral arteries that preserved limb perfusion and prevented tissue necrosis in ischaemia. Improved arteriogenesis in Phd2(+/-) mice was due to an expansion of tissue-resident, M2-like macrophages and their increased release of arteriogenic factors, leading to enhanced smooth muscle cell (SMC) recruitment and growth. Both chronic and acute deletion of one Phd2 allele in macrophages was sufficient to skew their polarization towards a pro-arteriogenic phenotype. Mechanistically, collateral vessel preconditioning relied on the activation of canonical NF-κB pathway in Phd2(+/-) macrophages. These results unravel how PHD2 regulates arteriogenesis and artery homeostasis by controlling a specific differentiation state in macrophages and suggest new treatment options for ischaemic disorders.


Assuntos
Artérias/crescimento & desenvolvimento , Extremidades/irrigação sanguínea , Isquemia/prevenção & controle , Macrófagos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/deficiência , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Alelos , Animais , Modelos Animais de Doenças , Extremidades/patologia , Feminino , Heterozigoto , Homeostase , Prolina Dioxigenases do Fator Induzível por Hipóxia , Isquemia/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Miócitos de Músculo Liso/citologia , NF-kappa B/metabolismo , Necrose , Fenótipo , Pró-Colágeno-Prolina Dioxigenase/genética
10.
Stem Cells ; 33(5): 1377-89, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25639612

RESUMO

microRNAs (miRNAs) are short noncoding RNAs, which regulate gene expression post-transcriptionally and play crucial roles in relevant biological and pathological processes. Here, we investigated the putative role of miRNAs in modulating the tumor-initiating potential of mouse medulloblastoma (MB)-derived cancer stem cells (CSCs). We first subjected bona fide highly tumorigenic (HT) CSCs as well as lowly tumorigenic MB CSCs and normal neural stem cells to miRNA profiling, which identified a HT CSC-specific miRNA signature. Next, by cross-checking CSC mRNA/miRNA profiles, we pinpointed miR-135a as a potential tumor suppressor gene, which was strongly downregulated in HT CSCs as well as in the highly malignant experimental tumors derived from them. Remarkably, enforced expression of miR-135a in HT CSCs strongly inhibited tumorigenesis by repressing the miR-135a direct target gene Arhgef6. Considering the upregulation of Arhgef6 in human MBs and its involvement in mediating experimental medulloblastomagenesis, its efficient suppression by miR-135a might make available an effective therapeutic strategy to selectively impair the tumorigenic potential of MB CSCs. Stem Cells 2015;33:1377-1389.


Assuntos
Carcinogênese/patologia , Meduloblastoma/patologia , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Animais , Carcinogênese/genética , Agregação Celular , Transformação Celular Neoplásica/genética , Regulação para Baixo , Perfilação da Expressão Gênica , Meduloblastoma/genética , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Trends Immunol ; 34(7): 350-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23498847

RESUMO

Deregulation of microRNAs (miRNAs) can drive oncogenesis, tumor progression, and metastasis by acting cell-autonomously in cancer cells. However, solid tumors are also infiltrated by large amounts of non-neoplastic stromal cells, including macrophages, which express several active miRNAs. Tumor-associated macrophages (TAMs) enhance angiogenic, immunosuppressive, invasive, and metastatic programming of neoplastic tissue and reduce host survival. Here, we review the role of miRNAs (including miR-155, miR-146, and miR-511) in the control of macrophage production and activation, and examine whether reprogramming miRNA activity in TAMs and/or their precursors might be effective for controlling tumor progression.


Assuntos
Diferenciação Celular/imunologia , Macrófagos/imunologia , MicroRNAs/imunologia , Neoplasias/imunologia , Animais , Carcinogênese , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Ativação de Macrófagos
13.
Exp Cell Res ; 319(11): 1626-34, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23542777

RESUMO

The ability of macrophages to promote vascular growth has been associated with the secretion and local delivery of classic proangiogenic factors (e.g., VEGF-A and proteases). More recently, a series of studies have also revealed that physical contact of macrophages with growing blood vessels coordinates vascular fusion of emerging sprouts. Interestingly, the interactions between macrophages and vascular endothelial cells (ECs) appear to be bidirectional, such that activated ECs also support the expansion and differentiation of proangiogenic macrophages from myeloid progenitors. Here, we discuss recent findings suggesting that dynamic angiogenic vascular niches might also exist in vivo, e.g. in tumors, where sprouting blood vessels and immature myeloid cells like monocytes engage in heterotypic interactions that are required for angiogenesis. Finally, we provide an account of emerging mechanisms of cell-to-cell communication that rely on secreted microvesicles, such as exosomes, which can offer a vehicle for the rapid exchange of molecules and genetic information between macrophages and ECs engaged in angiogenesis.


Assuntos
Comunicação Celular , Diferenciação Celular , Endotélio Vascular/citologia , Macrófagos/citologia , Neovascularização Patológica , Neovascularização Fisiológica , Animais , Humanos
14.
Nat Cancer ; 5(6): 827-843, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38918437

RESUMO

Research into the mechanisms and manifestations of solid tumor vascularization was launched more than 50 years ago with the proposition and experimental demonstrations that angiogenesis is instrumental for tumor growth and was, therefore, a promising therapeutic target. The biological knowledge and therapeutic insights forthcoming have been remarkable, punctuated by new concepts, many of which were not foreseen in the early decades. This article presents a perspective on tumor vascularization and its therapeutic targeting but does not portray a historical timeline. Rather, we highlight eight conceptual milestones, integrating initial discoveries and recent progress and posing open questions for the future.


Assuntos
Neoplasias , Neovascularização Patológica , Humanos , Neovascularização Patológica/história , Neoplasias/irrigação sanguínea , Neoplasias/história , Neoplasias/patologia , Inibidores da Angiogênese/uso terapêutico , Animais , História do Século XX , Terapia de Alvo Molecular/métodos
15.
Nat Cancer ; 5(2): 240-261, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37996514

RESUMO

Dendritic cells (DCs) are antigen-presenting myeloid cells that regulate T cell activation, trafficking and function. Monocyte-derived DCs pulsed with tumor antigens have been tested extensively for therapeutic vaccination in cancer, with mixed clinical results. Here, we present a cell-therapy platform based on mouse or human DC progenitors (DCPs) engineered to produce two immunostimulatory cytokines, IL-12 and FLT3L. Cytokine-armed DCPs differentiated into conventional type-I DCs (cDC1) and suppressed tumor growth, including melanoma and autochthonous liver models, without the need for antigen loading or myeloablative host conditioning. Tumor response involved synergy between IL-12 and FLT3L and was associated with natural killer and T cell infiltration and activation, M1-like macrophage programming and ischemic tumor necrosis. Antitumor immunity was dependent on endogenous cDC1 expansion and interferon-γ signaling but did not require CD8+ T cell cytotoxicity. Cytokine-armed DCPs synergized effectively with anti-GD2 chimeric-antigen receptor (CAR) T cells in eradicating intracranial gliomas in mice, illustrating their potential in combination therapies.


Assuntos
Citocinas , Neoplasias , Humanos , Camundongos , Animais , Imunoterapia , Células Dendríticas , Neoplasias/terapia , Interleucina-12
16.
J Pathol ; 227(1): 4-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22367978

RESUMO

Tumour-associated macrophages (TAMs) foster tumour progression by several mechanisms, including the promotion of angiogenesis, tissue remodelling, and immunosuppression. Such pro-tumoural activities are thought to be executed by TAM subtypes that harbour features of alternatively activated (or M2-polarized) macrophages. However, the molecular signals in tumours that induce recruitment and differentiation of M2-like macrophages are not fully defined. In this issue of The Journal of Pathology, Linde et al investigate the role of the tumour-derived cytokines, VEGF and IL-4, in the recruitment and polarization of macrophages in a mouse model of skin cancer. The authors report that while VEGF-A recruits monocytes from the peripheral circulation, IL-4 induces their differentiation into tumour-promoting, M2-like macrophages. IL-4 signalling blockade was sufficient to reprogram TAMs away from the M2-like phenotype and inhibited tumour angiogenesis and growth. This study attests to the potential of reprogramming TAMs to abate their pro-angiogenic and pro-tumoural functions in tumours.


Assuntos
Regulação Neoplásica da Expressão Gênica , Macrófagos/patologia , Neoplasias Cutâneas/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Humanos
17.
J Immunol ; 187(5): 2181-92, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21810604

RESUMO

M1 activation of macrophages promotes inflammation and immunity to intracellular pathogens, whereas M2 macrophage activation promotes resolution of inflammation, wound healing, and tumor growth. These divergent phenotypes are characterized, in part, by the expression of inducible NO synthase and arginase I (Arg1) in M1 versus M2 activated macrophages, respectively. In this study, we demonstrate that the Ron receptor tyrosine kinase tips the balance of macrophage activation by attenuating the M1 phenotype while promoting expression of Arg1 through a Stat6-independent mechanism. Induction of the Arg1 promoter by Ron is mediated by an AP-1 site located 433 bp upstream of the transcription start site. Treatment of primary macrophages with macrophage stimulating protein, the ligand for Ron, induces potent MAPK activation, upregulates Fos, and enhances binding of Fos to the AP-1 site in the Arg1 promoter. In vivo, Arg1 expression in tumor-associated macrophages (TAMs) from Ron(-/-) mice was significantly reduced compared with that in TAMs from control animals. Furthermore, we show that Ron is expressed specifically by Tie2-expressing macrophages, a TAM subset that exhibits a markedly skewed M2 and protumoral phenotype. Decreased Arg1 in TAMs from Ron(-/-) mice was associated with reduced syngeneic tumor growth in these animals. These findings indicate that Ron induces Arg1 expression in macrophages through a previously uncharacterized AP-1 site in the Arg1 promoter and that Ron could be therapeutically targeted in the tumor microenvironment to inhibit tumor growth by targeting expression of Arg1.


Assuntos
Arginase/biossíntese , Regulação da Expressão Gênica/imunologia , Macrófagos/enzimologia , Neoplasias Experimentais/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Arginase/genética , Arginase/imunologia , Separação Celular , Citometria de Fluxo , Expressão Gênica , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/imunologia
18.
J Immunol ; 186(7): 4183-90, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21368233

RESUMO

Angiopoietin 2 (ANGPT2) is a proangiogenic cytokine whose expression is often upregulated by endothelial cells in tumors. Expression of its receptor, TIE2, defines a highly proangiogenic subpopulation of myeloid cells in circulation and tumors called TIE2-expressing monocytes/macrophages (TEMs). Genetic depletion of TEMs markedly reduces tumor angiogenesis in various tumor models, emphasizing their essential role in driving tumor progression. Previously, we demonstrated that ANGPT2 augments the expression of various proangiogenic genes, the potent immunosuppressive cytokine, IL-10, and a chemokine for regulatory T cells (Tregs), CCL17 by TEMs in vitro. We now show that TEMs also express higher levels of IL-10 than TIE2(-) macrophages in tumors and that ANGPT2-stimulated release of IL-10 by TEMs suppresses T cell proliferation, increases the ratio of CD4(+) T cells to CD8(+) T cells, and promotes the expansion of CD4(+)CD25(high)FOXP3(+) Tregs. Furthermore, syngeneic murine tumors expressing high levels of ANGPT2 contained not only high numbers of TEMs but also increased numbers of Tregs, whereas genetic depletion of tumor TEMs resulted in a marked reduction in the frequency of Tregs in tumors. Taken together, our data suggest that ANGPT2-stimulated TEMs represent a novel, potent immunosuppressive force in tumors.


Assuntos
Angiopoietina-2/fisiologia , Proteínas de Ciclo Celular/fisiologia , Diferenciação Celular/imunologia , Proteínas de Ligação a DNA/fisiologia , Ativação Linfocitária/imunologia , Monócitos/imunologia , Neovascularização Patológica/imunologia , Proteínas Repressoras/fisiologia , Linfócitos T Reguladores/imunologia , Fatores de Transcrição/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/patologia , Proteínas de Ciclo Celular/biossíntese , Linhagem Celular Tumoral , Técnicas de Cocultura , Proteínas de Ligação a DNA/biossíntese , Inibidores do Crescimento/biossíntese , Inibidores do Crescimento/fisiologia , Humanos , Interleucina-10/biossíntese , Interleucina-10/metabolismo , Interleucina-10/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Monócitos/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Proteínas Repressoras/biossíntese , Linfócitos T Reguladores/citologia , Fatores de Transcrição/biossíntese
19.
Brain ; 135(Pt 2): 447-60, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22271661

RESUMO

Transplanted neural stem/precursor cells possess peculiar therapeutic plasticity and can simultaneously instruct several therapeutic mechanisms in addition to cell replacement. Here, we interrogated the therapeutic plasticity of neural stem/precursor cells after their focal implantation in the severely contused spinal cord. We injected syngeneic neural stem/precursor cells at the proximal and distal ends of the contused mouse spinal cord and analysed locomotor functions and relevant secondary pathological events in the mice, cell fate of transplanted neural stem/precursor cells, and gene expression and inflammatory cell infiltration at the injured site. We used two different doses of neural stem/precursor cells and two treatment schedules, either subacute (7 days) or early chronic (21 days) neural stem/precursor cell transplantation after the induction of experimental thoracic severe spinal cord injury. Only the subacute transplant of neural stem/precursor cells enhanced the recovery of locomotor functions of mice with spinal cord injury. Transplanted neural stem/precursor cells survived undifferentiated at the level of the peri-lesion environment and established contacts with endogenous phagocytes via cellular-junctional coupling. This was associated with significant modulation of the expression levels of important inflammatory cell transcripts in vivo. Transplanted neural stem/precursor cells skewed the inflammatory cell infiltrate at the injured site by reducing the proportion of 'classically-activated' (M1-like) macrophages, while promoting the healing of the injured cord. We here identify a precise window of opportunity for the treatment of complex spinal cord injuries with therapeutically plastic somatic stem cells, and suggest that neural stem/precursor cells have the ability to re-programme the local inflammatory cell microenvironment from a 'hostile' to an 'instructive' role, thus facilitating the healing or regeneration past the lesion.


Assuntos
Regeneração Nervosa/fisiologia , Células-Tronco Neurais/transplante , Fagócitos/fisiologia , Traumatismos da Medula Espinal/terapia , Animais , Diferenciação Celular , Camundongos , Atividade Motora/fisiologia , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Resultado do Tratamento
20.
Cancer Cell ; 8(3): 211-26, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16169466

RESUMO

Bone marrow-derived cells contribute to tumor angiogenesis. Here, we demonstrate that monocytes expressing the Tie2 receptor (Tie2-expressing monocytes [TEMs]) (1) are a distinct hematopoietic lineage of proangiogenic cells, (2) are selectively recruited to spontaneous and orthotopic tumors, (3) promote angiogenesis in a paracrine manner, and (4) account for most of the proangiogenic activity of myeloid cells in tumors. Remarkably, TEM knockout completely prevented human glioma neovascularization in the mouse brain and induced substantial tumor regression. Besides TEMs and endothelial cells (ECs), Tie2 expression distinguished a rare population of tumor stroma-derived mesenchymal progenitors representing a primary source of tumor pericytes. Therefore, Tie2 expression characterizes three distinct cell types required for tumor neovascularization: ECs, proangiogenic cells of hematopoietic origin, and pericyte precursors of mesenchymal origin.


Assuntos
Glioblastoma/patologia , Monócitos/fisiologia , Pericitos/patologia , Receptor TIE-2/fisiologia , Animais , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Humanos , Mesoderma/citologia , Mesoderma/patologia , Camundongos , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos , Neovascularização Patológica/fisiopatologia , Neoplasias Pancreáticas/patologia , Células-Tronco/patologia , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA