Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(35): e2405746121, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39172787

RESUMO

While macrophage heterogeneity during metabolic dysfunction-associated steatohepatitis (MASH) has been described, the fate of these macrophages during MASH regression is poorly understood. Comparing macrophage heterogeneity during MASH progression vs regression, we identified specific macrophage subpopulations that are critical for MASH/fibrosis resolution. We elucidated the restorative pathways and gene signatures that define regression-associated macrophages and establish the importance of TREM2+ macrophages during MASH regression. Liver-resident Kupffer cells are lost during MASH and are replaced by four distinct monocyte-derived macrophage subpopulations. Trem2 is expressed in two macrophage subpopulations: i) monocyte-derived macrophages occupying the Kupffer cell niche (MoKC) and ii) lipid-associated macrophages (LAM). In regression livers, no new transcriptionally distinct macrophage subpopulation emerged. However, the relative macrophage composition changed during regression compared to MASH. While MoKC was the major macrophage subpopulation during MASH, they decreased during regression. LAM was the dominant macrophage subtype during MASH regression and maintained Trem2 expression. Both MoKC and LAM were enriched in disease-resolving pathways. Absence of TREM2 restricted the emergence of LAMs and formation of hepatic crown-like structures. TREM2+ macrophages are functionally important not only for restricting MASH-fibrosis progression but also for effective regression of inflammation and fibrosis. TREM2+ macrophages are superior collagen degraders. Lack of TREM2+ macrophages also prevented elimination of hepatic steatosis and inactivation of HSC during regression, indicating their significance in metabolic coordination with other cell types in the liver. TREM2 imparts this protective effect through multifactorial mechanisms, including improved phagocytosis, lipid handling, and collagen degradation.


Assuntos
Células de Kupffer , Cirrose Hepática , Macrófagos , Glicoproteínas de Membrana , Receptores Imunológicos , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Animais , Camundongos , Macrófagos/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/genética , Células de Kupffer/metabolismo , Fígado/metabolismo , Fígado/patologia , Metabolismo dos Lipídeos , Camundongos Endogâmicos C57BL , Masculino , Lipídeos , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/genética , Camundongos Knockout
2.
Mol Cancer Res ; 19(3): 507-515, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33234577

RESUMO

Hematopoietic-derived cells are integral components of the tumor microenvironment and serve as critical mediators of tumor-host interactions. Host cells derived from myeloid and lymphoid lineages perform well-established functions linked to cancer development, progression, and response to therapy. It is unclear whether host erythroid cells also contribute to shaping the path that cancer can take, but emerging evidence points to this possibility. Here, we show that tumor-promoting environmental stress and tumor-induced hemodynamic changes trigger renal erythropoietin production and erythropoietin-dependent expansion of splenic erythroid cell populations in mice. These erythroid cells display molecular features indicative of an immature erythroid phenotype, such as the expression of both CD71 and TER119 and the retention of intact nuclei, and express genes encoding immune checkpoint molecules. Nucleated erythroid cells with similar properties are present in mouse and human tumor tissues. Antibody-mediated erythropoietin blockade reduces tumor-responsive erythroid cell induction and tumor growth. These findings reveal the potential of tumor-induced erythropoietin and erythroid cells as targets for cancer treatment. IMPLICATIONS: : Our study identifies erythropoietin and erythroid cells as novel players in tumor-host interactions and highlights the involvement of multiorgan signaling events in their induction in response to environmental stress and tumor growth.


Assuntos
Células Eritroides/metabolismo , Proteínas de Checkpoint Imunológico/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos , Transdução de Sinais
3.
J Immunol ; 196(5): 2368-76, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26792803

RESUMO

The protein kinase p38α mediates cellular responses to environmental and endogenous cues that direct tissue homeostasis and immune responses. Studies of mice lacking p38α in several different cell types have demonstrated that p38α signaling is essential to maintaining the proliferation-differentiation balance in developing and steady-state tissues. The mechanisms underlying these roles involve cell-autonomous control of signaling and gene expression by p38α. In this study, we show that p38α regulates gut-associated lymphoid tissue (GALT) formation in a noncell-autonomous manner. From an investigation of mice with intestinal epithelial cell-specific deletion of the p38α gene, we find that p38α serves to limit NF-κB signaling and thereby attenuate GALT-promoting chemokine expression in the intestinal epithelium. Loss of this regulation results in GALT hyperplasia and, in some animals, mucosa-associated B cell lymphoma. These anomalies occur independently of luminal microbial stimuli and are most likely driven by direct epithelial-lymphoid interactions. Our study illustrates a novel p38α-dependent mechanism preventing excessive generation of epithelial-derived signals that drive lymphoid tissue overgrowth and malignancy.


Assuntos
Mucosa Intestinal/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Colite/genética , Colite/imunologia , Colite/metabolismo , Colite/patologia , Colo/imunologia , Colo/metabolismo , Colo/microbiologia , Colo/patologia , Células Epiteliais/metabolismo , Expressão Gênica , Hiperplasia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microbiota/imunologia , Proteína Quinase 14 Ativada por Mitógeno/genética , Nódulos Linfáticos Agregados/patologia
4.
J Biol Chem ; 290(1): 284-95, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25406311

RESUMO

An improved understanding of the molecular pathways that drive tooth morphogenesis and enamel secretion is needed to generate teeth from organ cultures for therapeutic implantation or to determine the pathogenesis of primary disorders of dentition (Abdollah, S., Macias-Silva, M., Tsukazaki, T., Hayashi, H., Attisano, L., and Wrana, J. L. (1997) J. Biol. Chem. 272, 27678-27685). Here we present a novel ectodermal dysplasia phenotype associated with conditional deletion of p38α MAPK in ectodermal appendages using K14-cre mice (p38α(K14) mice). These mice display impaired patterning of dental cusps and a profound defect in the production and biomechanical strength of dental enamel because of defects in ameloblast differentiation and activity. In the absence of p38α, expression of amelogenin and ß4-integrin in ameloblasts and p21 in the enamel knot was significantly reduced. Mice lacking the MAP2K MKK6, but not mice lacking MAP2K MKK3, also show the enamel defects, implying that MKK6 functions as an upstream kinase of p38α in ectodermal appendages. Lastly, stimulation with BMP2/7 in both explant culture and an ameloblast cell line confirm that p38α functions downstream of BMPs in this context. Thus, BMP-induced activation of the p38α MAPK pathway is critical for the morphogenesis of tooth cusps and the secretion of dental enamel.


Assuntos
Ameloblastos/metabolismo , Esmalte Dentário/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Incisivo/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Odontogênese/genética , Ameloblastos/citologia , Amelogenina/genética , Amelogenina/metabolismo , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Diferenciação Celular , Proliferação de Células , Esmalte Dentário/citologia , Esmalte Dentário/crescimento & desenvolvimento , Incisivo/citologia , Incisivo/crescimento & desenvolvimento , Integrina beta4/genética , Integrina beta4/metabolismo , MAP Quinase Quinase 3/genética , MAP Quinase Quinase 3/metabolismo , MAP Quinase Quinase 6/genética , MAP Quinase Quinase 6/metabolismo , Camundongos , Camundongos Transgênicos , Proteína Quinase 14 Ativada por Mitógeno/genética , Transdução de Sinais , Técnicas de Cultura de Tecidos , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
5.
J Invest Dermatol ; 134(8): 2231-2240, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24662766

RESUMO

UVB is a component of solar radiation primarily responsible for causing damage and cancer in irradiated skin, and disrupting immune homeostasis. The immediate harm and long-term health risks of excessive sunlight exposure are affecting the lives of nearly all people worldwide. Inflammation is a key mechanism underlying UVB's various detrimental effects. Here we show that activation of the protein kinase p38α is restricted to the epidermis in UVB-exposed skin, and that p38α ablation targeted to the epithelial compartment is sufficient to suppress UVB-induced inflammation. Mechanistically, loss of epithelial p38α signaling attenuates the expression of genes required to induce vascular leakage and edema, and also increases the steady-state abundance of epidermal γδ T cells, which are known to promote the repair of damaged epidermis. These effects of p38α deficiency delineate a molecular network operating at the organism-environment interface, and reveal conditions crucial to preventing the pathology resulting from sun-damaged skin.


Assuntos
Dermatite/prevenção & controle , Epiderme/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Raios Ultravioleta/efeitos adversos , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Ciclo-Oxigenase 2/fisiologia , Dermatite/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T gama-delta/fisiologia
6.
J Biol Chem ; 288(33): 23788-97, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23836897

RESUMO

The epithelium of mucosal and skin surfaces serves as a permeability barrier and affords mechanisms for local immune defense. Crucial to the development and maintenance of a properly functioning epithelium is the balance of cell proliferation, differentiation, and death. Here we show that this balance depends on cross-regulatory interactions among multiple protein kinase-mediated signals and their coordinated transmission. From an investigation of conditional gene knock-out mice, we find that epithelial-specific loss of the protein kinase p38α leads to aberrant activation of TAK1, JNK, EGF receptor, and ERK in distinct microanatomical areas of the intestines and skin. Consequently, the epithelial tissues display excessive proliferation, inadequate differentiation, and sensitivity to apoptosis. These anomalies leave the tissue prone to damage and collapse at the trigger of an environmental insult. The vulnerability of p38α-deficient epithelium predicts adverse effects of long term pharmacological p38α inhibition; yet such limitations could be overcome by concomitant blockade of one or more of the dysregulated protein kinase signaling pathways.


Assuntos
Epitélio/enzimologia , Homeostase , Sistema de Sinalização das MAP Quinases , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Ativação Enzimática , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Epitélio/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Inflamação/enzimologia , Inflamação/patologia , Mucosa Intestinal/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Pele/patologia , Ubiquitinação , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
7.
Mol Immunol ; 54(2): 157-63, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23287596

RESUMO

The cytokine interleukin-4 (IL-4) exerts pleiotropic effects on macrophages as it plays a key role in the immune response to infectious agents, allergens, and vaccines. Macrophages exposed to IL-4 drastically change their gene expression and metabolic state to adjust to new functional requirements. IL-4 also induces macrophages to fuse together and form multinucleated giant cells (MGCs). MGC formation is associated with chronic inflammation resulting from persistence of pathogenic microorganisms or foreign materials in tissues. Very little is known, however, about the mechanisms regulating IL-4-induced macrophage-to-MGC conversion. We observed a dramatic increase in ß-catenin protein but not mRNA amount in mouse macrophages following exposure to IL-4. To investigate the role of ß-catenin in macrophages, we generated mice with a myeloid cell-specific deletion of the ß-catenin gene. Ablation of ß-catenin expression did not affect the viability of macrophages or impair expression of known IL-4-inducible genes. Intriguingly, ß-catenin-deficient macrophages incubated with IL-4 formed MGCs with markedly greater efficiency than wild-type macrophages. Similar increases in multinucleated cell formation were detected in the peritoneal cavity of myeloid cell-specific ß-catenin knockout mice injected with chitin, which is known to induce endogenous IL-4 production. Our findings reveal ß-catenin as a novel regulator of macrophage responses to IL-4, and suggest that therapeutic modulation of its expression or function may help enhance the effectiveness or ameliorate the pathology of IL-4-driven immune responses.


Assuntos
Células Gigantes/efeitos dos fármacos , Células Gigantes/metabolismo , Interleucina-4/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , beta Catenina/metabolismo , Animais , Células Cultivadas , Quitina/efeitos adversos , Quitina/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Peritonite/induzido quimicamente , Peritonite/imunologia , beta Catenina/genética
8.
Cancer Discov ; 1(6): 496-507, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22389871

RESUMO

UNLABELLED: The transcription factor NF-κB promotes survival of cancer cells exposed to doxorubicin and other chemotherapeutic agents. IκB kinase is essential for chemotherapy-induced NF-κB activation and considered a prime target for anticancer treatment. An IκB kinase inhibitor sensitized human melanoma xenografts in mice to killing by doxorubicin, yet also exacerbated treatment toxicity in the host animals. Using mouse models that simulate cell-selective targeting, we found that impaired NF-κB activation in melanoma and host myeloid cells accounts for the therapeutic and the adverse effects, respectively. Ablation of tumor-intrinsic NF-κB activity resulted in apoptosis-driven tumor regression following doxorubicin treatment. By contrast, chemotherapy in mice with myeloid-specific loss of NF-κB activation led to a massive intratumoral recruitment of interleukin-1ß-producing neutrophils and necrotic tumor lesions, a condition associated with increased host mortality but not accompanied by tumor regression. Therefore, a molecular target-based therapy may be steered toward different clinical outcomes depending on the drug's cell-specific effects. SIGNIFICANCE: Our findings show that the IκB kinase­NF-κB signaling pathway is important for both promoting treatment resistance and preventing host toxicity in cancer chemotherapy; however, the two functions are exerted by distinct cell type­specific mechanisms and can therefore be selectively targeted to achieve an improved therapeutic outcome.


Assuntos
Melanoma/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/metabolismo , Imidazóis/farmacologia , Interleucina-1beta/metabolismo , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , NF-kappa B/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinoxalinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Clin Invest ; 120(7): 2457-73, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20551513

RESUMO

Nearly every extracellular ligand that has been found to play a role in regulating bone biology acts, at least in part, through MAPK pathways. Nevertheless, much remains to be learned about the contribution of MAPKs to osteoblast biology in vivo. Here we report that the p38 MAPK pathway is required for normal skeletogenesis in mice, as mice with deletion of any of the MAPK pathway member-encoding genes MAPK kinase 3 (Mkk3), Mkk6, p38a, or p38b displayed profoundly reduced bone mass secondary to defective osteoblast differentiation. Among the MAPK kinase kinase (MAP3K) family, we identified TGF-beta-activated kinase 1 (TAK1; also known as MAP3K7) as the critical activator upstream of p38 in osteoblasts. Osteoblast-specific deletion of Tak1 resulted in clavicular hypoplasia and delayed fontanelle fusion, a phenotype similar to the cleidocranial dysplasia observed in humans haploinsufficient for the transcription factor runt-related transcription factor 2 (Runx2). Mechanistic analysis revealed that the TAK1-MKK3/6-p38 MAPK axis phosphorylated Runx2, promoting its association with the coactivator CREB-binding protein (CBP), which was required to regulate osteoblast genetic programs. These findings reveal an in vivo function for p38beta and establish that MAPK signaling is essential for bone formation in vivo. These results also suggest that selective p38beta agonists may represent attractive therapeutic agents to prevent bone loss associated with osteoporosis and aging.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Animais , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Humanos , MAP Quinase Quinase Quinases/genética , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Transgênicos , Proteína Quinase 14 Ativada por Mitógeno/genética , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , Fosforilação , Proteínas Proto-Oncogênicas , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
BMC Immunol ; 10: 57, 2009 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-19863805

RESUMO

BACKGROUND: Selenium, a micronutrient whose deficiency in diet causes immune dysfunction and inflammatory disorders, is thought to exert its physiological effects mostly in the form of selenium-containing proteins (selenoproteins). Incorporation of selenium into the amino acid selenocysteine (Sec), and subsequently into selenoproteins is mediated by Sec tRNA([Ser]Sec). RESULTS: To define macrophage-specific selenoprotein functions, we generated mice with the Sec tRNA([Ser]Sec) gene specifically deleted in myeloid cells. These mutant mice were devoid of the "selenoproteome" in macrophages, yet exhibited largely normal inflammatory responses. However, selenoprotein deficiency led to aberrant expression of extracellular matrix-related genes, and diminished migration of macrophages in a protein gel matrix. CONCLUSION: Selenium status may affect immune defense and tissue homeostasis through its effect on selenoprotein expression and the trafficking of tissue macrophages.


Assuntos
Movimento Celular/genética , Dermatite Irritante/imunologia , Proteínas da Matriz Extracelular/metabolismo , Macrófagos/metabolismo , Peritonite/imunologia , Selenoproteínas/metabolismo , Animais , Citocinas/metabolismo , Dermatite Irritante/genética , Dermatite Irritante/metabolismo , Dermatite Irritante/patologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/imunologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Lipopolissacarídeos/administração & dosagem , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Oxirredução , Peritonite/induzido quimicamente , Peritonite/genética , Peritonite/metabolismo , Peritonite/patologia , Ésteres de Forbol/administração & dosagem , RNA de Transferência/genética , Selenoproteínas/genética , Selenoproteínas/imunologia , Zimosan/administração & dosagem
11.
Nat Immunol ; 9(9): 1019-27, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18677317

RESUMO

The mitogen-activated protein kinase p38 mediates cellular responses to injurious stress and immune signaling. Among the many p38 isoforms, p38 alpha is the most widely expressed in adult tissues and can be targeted by various pharmacological inhibitors. Here we investigated how p38 alpha activation is linked to cell type-specific outputs in mouse models of cutaneous inflammation. We found that both myeloid and epithelial p38 elicit inflammatory responses, yet p38 alpha signaling in each cell type served distinct inflammatory functions and varied depending on the mode of skin irritation. In addition, myeloid p38 alpha limited acute inflammation via activation of anti-inflammatory gene expression dependent on mitogen- and stress-activated kinases. Our results suggest a dual function for p38 alpha in the regulation of inflammation and show mixed potential for its inhibition as a therapeutic strategy.


Assuntos
Mediadores da Inflamação/metabolismo , Inflamação/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Células Cultivadas/metabolismo , Modelos Animais de Doenças , Células Epiteliais , Expressão Gênica/efeitos dos fármacos , Camundongos , Células Mieloides , Inibidores de Proteínas Quinases/farmacologia , Dermatopatias/genética , Dermatopatias/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
12.
Proc Natl Acad Sci U S A ; 105(16): 6150-5, 2008 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-18427110

RESUMO

Bacillus anthracis, the etiologic agent of anthrax, avoids immune surveillance and commandeers host macrophages as a vehicle for lymphatic spreading. Here, we show that B. anthracis edema toxin (ET), via its adenylyl cyclase activity, dramatically increases the motility of infected macrophages and the expression of vascular endothelial growth factor. The transcription factor CREB and the syndecan-1 gene, a CREB target, play crucial roles in ET-induced macrophage migration. These molecular and cellular responses occur in macrophages engaged in antiinflammatory G protein-coupled receptor activation, thus illustrating a common signaling circuitry controlling resolution of inflammation and host cell hijacking by B. anthracis.


Assuntos
Antraz/imunologia , Bacillus anthracis/enzimologia , Movimento Celular/genética , AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Macrófagos/imunologia , Adenilil Ciclases/metabolismo , Animais , Antraz/genética , Antraz/microbiologia , Antígenos de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Sindecana-1/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA