Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
PLoS One ; 16(6): e0252868, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34185781

RESUMO

Regulation of dendritic cell functions is a complex process in which several mediators play diverse roles as a network in a context-dependent manner. The precise mechanisms underlying dendritic cell functions have remained to be addressed. Semaphorins play crucial roles in regulation of various cell functions. We previously revealed that Semaphorin 3E (Sema3E) contributes to regulation of allergen-induced airway pathology partly mediated by controlling recruitment of conventional dendritic cell subsets in vivo, though the underlying mechanism remained elusive. In this study, we investigate the potential regulatory role of Sema3E in dendritic cells. We demonstrated that bone marrow-derived dendritic cells differentiated from Sema3e-/- progenitors have an enhanced migration capacity both at the baseline and in response to CCL21. The enhanced migration ability of Sema3E dendritic cells was associated with an overexpression of the chemokine receptor (CCR7), elevated Rac1 GTPase activity and F-actin polymerization. Using a mouse model of allergic airway sensitization, we observed that genetic deletion of Sema3E leads to a time dependent upregulation of CCR7 on CD11b+ conventional dendritic cells in the lungs and mediastinal lymph nodes. Furthermore, aeroallergen sensitization of Sema3e-/- mice lead to an enhanced expression of PD-L2 and IRF-4 as well as enhanced allergen uptake in pulmonary CD11b+ DC, compared to wild type littermates. Collectively, these data suggest that Sema3E implicates in regulation of dendritic cell functions which could be considered a basis for novel immunotherapeutic strategies for the diseases associated with defective dendritic cells in the future.


Assuntos
Alérgenos/imunologia , Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Pneumonia/imunologia , Semaforinas/fisiologia , Actinas/metabolismo , Animais , Movimento Celular , Quimiocina CCL21/metabolismo , Camundongos , Camundongos Knockout , Neuropeptídeos/metabolismo , Pneumonia/metabolismo , Pneumonia/patologia , Receptores CCR7/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
2.
Neuroendocrinology ; 111(5): 421-441, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32365351

RESUMO

INTRODUCTION: Gonadotropin-releasing hormone (GnRH) deficiency causes hypogonadotropic hypogonadism (HH), a rare genetic disorder that impairs sexual reproduction. HH can be due to defective GnRH-secreting neuron development or function and may be associated with other clinical signs in overlapping genetic syndromes. With most of the cases being idiopathic, genetics underlying HH is still largely unknown. OBJECTIVE: To assess the contribution of mutated Semaphorin 3G (SEMA3G) in the onset of a syndromic form of HH, characterized by intellectual disability and facial dysmorphic features. METHOD: By combining homozygosity mapping with exome sequencing, we identified a novel variant in the SEMA3G gene. We then applied mouse as a model organism to examine SEMA3Gexpression and its functional requirement in vivo. Further, we applied homology modelling in silico and cell culture assays in vitro to validate the pathogenicity of the identified gene variant. RESULTS: We found that (i) SEMA3G is expressed along the migratory route of GnRH neurons and in the developing pituitary, (ii) SEMA3G affects GnRH neuron development, but is redundant in the adult hypothalamic-pituitary-gonadal axis, and (iii) mutated SEMA3G alters binding properties in silico and in vitro to its PlexinA receptors and attenuates its effect on the migration of immortalized GnRH neurons. CONCLUSION: In silico, in vitro, and in vivo models revealed that SEMA3G regulates GnRH neuron migration and that its mutation affecting receptor selectivity may be responsible for the HH-related defects.


Assuntos
Hormônio Liberador de Gonadotropina/deficiência , Hipogonadismo/genética , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistema Hipotálamo-Hipofisário/metabolismo , Semaforinas/fisiologia , Animais , Células Cultivadas , Consanguinidade , Anormalidades Craniofaciais/etiologia , Deficiências do Desenvolvimento/etiologia , Homozigoto , Humanos , Hipogonadismo/complicações , Deficiência Intelectual/etiologia , Masculino , Camundongos , Linhagem , Irmãos , Síndrome
3.
Biochem Soc Trans ; 48(6): 2875-2890, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33258873

RESUMO

In vertebrates, the semaphorin family of proteins is composed of 21 members that are divided into five subfamilies, i.e. classes 3 to 7. Semaphorins play crucial roles in regulating multiple biological processes, such as neural remodeling, tissue regeneration, cancer progression, and, especially, in immunological regulation. Semaphorin 4D (SEMA4D), also known as CD100, is an important member of the semaphorin family and was first characterized as a lymphocyte-specific marker. SEMA4D has diverse effects on immunologic processes, including immune cell proliferation, differentiation, activation, and migration, through binding to its specific membrane receptors CD72, PLXNB1, and PLXNB2. Furthermore, SEMA4D and its underlying signaling have been increasingly linked with several immunological diseases. This review focuses on the significant immunoregulatory role of SEMA4D and the associated underlying mechanisms, as well as the potential application of SEMA4D as a diagnostic marker and therapeutic target for the treatment of immunological diseases.


Assuntos
Antígenos CD/fisiologia , Doenças do Sistema Imunitário/metabolismo , Linfócitos/metabolismo , Regeneração , Semaforinas/fisiologia , Animais , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos B/biossíntese , Artrite Reumatoide/metabolismo , Linfócitos B/imunologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Dermatite Alérgica de Contato/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Humanos , Sistema Imunitário , Ligantes , Linfócitos/citologia , Glicoproteínas de Membrana/química , Esclerose Múltipla/metabolismo , Neoplasias/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Ligação Proteica , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/metabolismo , Semaforinas/imunologia , Transdução de Sinais/imunologia
4.
Cell Mol Biol Lett ; 23: 2, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29308068

RESUMO

BACKGROUND: Anti-angiogenesis therapy that targets VEGF is one of the important treatment strategies in advanced ovarian cancer. However, depending on the pharmaceutical agent, treatment can have undesirable side effects. SEMA4D has recently gained interest for its role in promoting angiogenesis. Here, we try to further understand the mechanism by which SEMA4D promotes angiogenesis in ovarian cancer. METHODS: Correlation and western blot assaya were used to detect the relationship between VEGF and SEMA4D in clinical tissues and cells. Vasculogenic mimicry and transwell migration analyses were used to detect the roles of VEGF, SEMA4D and plexin-B1 on vasculogenic mimicry and migration. Vascular density and SEMA4D expression was determined using immunofluorescence staining in clinical tissues of EOC. Western blot was used to detect the expressions of CD31, MMP2 and VE-cadherin. We also analyzed the relationship between VEGF-SEMA4D and malignant tumor prognosis. RESULTS: We found that knockdown of VEGF could suppress SEMA4D expression and that the expressions of VEGF and SEMA4D have a positive correlation in EOC cancer tissues. Vasculogenic mimicry and transwell migration analyses showed that SEMA4D and VEGF have a synergistic effect on the promotion of angiogenesis in A2780 and HUVEC cells. Soluble SEMA4D (sSEMA4D) could promote VM and migration in A2780 and HUVEC cells via the SEMA4D/plexin-B1 pathway, but the effect was not noted in stably transfected shR-plexin-B1 cells. In clinical tissues of EOC, the vascular density and SEMA4D/plexin-B1 expression were higher. When VEGF, SEMA4D and plexin-B1 was knocked down, the expression of CD31, MMP2 and VE-cadherin, which are the markers and initiators of angiogenesis and the epithelial-mesenchymal transition (EMT) process were reduced. VEGF and SEMA4D had a positive correlation with the malignant degree of ovarian cancer, and SEMA4D can serve as an independent prognostic factor. CONCLUSIONS: VEGF and SEMA4D have synergistic effects on the promotion of angiogenesis in epithelial ovarian cancer. Targeting VEGF and the SEMA4D signaling pathway could be important for the therapy for EOC.


Assuntos
Antígenos CD/metabolismo , Neoplasias Epiteliais e Glandulares/irrigação sanguínea , Neovascularização Patológica , Neoplasias Ovarianas/irrigação sanguínea , Semaforinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Antígenos CD/genética , Antígenos CD/fisiologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/mortalidade , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Prognóstico , Semaforinas/genética , Semaforinas/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia
5.
Toxicol Pathol ; 45(7): 894-903, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29113558

RESUMO

Although the brain is well established as a master regulator of homeostasis in peripheral tissues, central regulation of bone mass represents a novel and rapidly expanding field of study. This review examines the current understanding of central regulation of the skeleton, exploring several of the key pathways connecting brain to bone and their implications both in mice and the clinical setting. Our understanding of central bone regulation has largely progressed through examination of skeletal responses downstream of nutrient regulatory pathways in the hypothalamus. Mutations and modulation of these pathways, in cases such as leptin deficiency, induce marked bone phenotypes, which have provided vital insights into central bone regulation. These studies have identified several central neuropeptide pathways that stimulate well-defined changes in bone cell activity in response to changes in energy homeostasis. In addition, this work has highlighted the endocrine nature of the skeleton, revealing a complex cross talk that directly regulates other organ systems. Our laboratory has studied bone-active neuropeptide pathways and defined osteoblast-based actions that recapitulate central pathways linking bone, fat, and glucose homeostasis. Studies of neural control of bone have produced paradigm-shifting changes in our understanding of the skeleton and its relationship with the wider array of organ systems.


Assuntos
Remodelação Óssea , Neurônios/fisiologia , Animais , Osso e Ossos/fisiologia , Homeostase , Humanos , Hipotálamo/fisiologia , Leptina/fisiologia , Músculo Esquelético/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neuropeptídeo Y/fisiologia , Polipeptídeo Pancreático/fisiologia , Peptídeo YY/fisiologia , Pró-Opiomelanocortina/fisiologia , Receptores de Canabinoides/fisiologia , Semaforinas/fisiologia , Sistema Nervoso Simpático/fisiologia
6.
Cancer Cell ; 32(4): 427-443.e8, 2017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-29017055

RESUMO

Neuroblastoma (NB) is a childhood cancer arising from sympatho-adrenal neural crest cells. Disseminated forms have high frequency of multiple tumoral foci whose etiology remains unknown; NB embryonic origin limits investigations in patients and current models. We developed an avian embryonic model driving human NB tumorigenesis in tissues homologous to patients. We found that aggressive NBs display a metastatic mode, secondary dissemination via peripheral nerves and aorta. Through tumor transcriptional profiling, we found that NB dissemination is induced by the shutdown of a pro-cohesion autocrine signal, SEMA3C, which constrains the tumoral mass. Lowering SEMA3C levels shifts the balance toward detachment, triggering NB cells to collectively evade the tumor. Together with patient cohort analysis, this identifies a microenvironment-driven pro-metastatic switch for NB.


Assuntos
Neuroblastoma/secundário , Microambiente Tumoral , Adolescente , Adulto , Animais , Adesão Celular , Embrião de Galinha , Criança , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Estadiamento de Neoplasias , Proteínas do Tecido Nervoso/fisiologia , Neuroblastoma/etiologia , Neuroblastoma/patologia , Receptores de Superfície Celular/fisiologia , Semaforinas/genética , Semaforinas/fisiologia
7.
Methods Mol Biol ; 1493: 1-25, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787839

RESUMO

Semaphorins are extracellular signaling proteins that are essential for the development and maintenance of many organs and tissues. The more than 20-member semaphorin protein family includes secreted, transmembrane and cell surface-attached proteins with diverse structures, each characterized by a single cysteine-rich extracellular sema domain, the defining feature of the family. Early studies revealed that semaphorins function as axon guidance molecules, but it is now understood that semaphorins are key regulators of morphology and motility in many different cell types including those that make up the nervous, cardiovascular, immune, endocrine, hepatic, renal, reproductive, respiratory and musculoskeletal systems, as well as in cancer cells. Semaphorin signaling occurs predominantly through Plexin receptors and results in changes to the cytoskeletal and adhesive machinery that regulate cellular morphology. While much remains to be learned about the mechanisms underlying the effects of semaphorins, exciting work has begun to reveal how semaphorin signaling is fine-tuned through different receptor complexes and other mechanisms to achieve specific outcomes in various cellular contexts and physiological systems. These and future studies will lead to a more complete understanding of semaphorin-mediated development and to a greater understanding of how these proteins function in human disease.


Assuntos
Semaforinas/fisiologia , Transdução de Sinais , Animais , Humanos , Semaforinas/metabolismo
8.
Methods Mol Biol ; 1493: 345-361, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787863

RESUMO

Angiogenesis identifies the process of endothelial cell sprouting and remodeling leading to the formation of new and functional blood vessels. Vascular expansion during development and in the adult mammal provides nutrients and oxygen to areas with increased need. Although many molecules and pathways have been identified as regulators of angiogenesis, aspects of this complex process remain unclear. Particularly undefined are the signals that orchestrate vessel survival and pruning once new blood vessels have sprouted. These poorly characterized aspects of angiogenesis need exploration. This chapter describes the experiments and methods enabling the characterization of Semaphorin 6A as a critical regulator of endothelial cell survival and vessel function.


Assuntos
Neovascularização Fisiológica/fisiologia , Semaforinas/fisiologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Sobrevivência Celular , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos
9.
Oncotarget ; 7(52): 87431-87448, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-27911862

RESUMO

Semaphorin-3E (Sema3E) is a member of an axon guidance gene family, and has recently been reported to contribute to tumor progression and metastasis. However, its role in pancreatic cancer is yet unknown and uncharacterized. In this study, we showed that Sema3E is overexpressed in human pancreatic cancer, and that high Sema3E levels are associated with tumor progression and poor survival. Interestingly, we also observed Sema3E expression in the nucleus, even though Sema3E is reported to be a secreted protein. Overexpression of Sema3E in pancreatic cancer cells promoted cell proliferation and migration in vitro, and increased tumor incidence and growth in vivo. Conversely, knockout of Sema3E suppressed cancer cell proliferation and migration in vitro, and reduced tumor incidence and size in vivo. Moreover, Sema3E induced cell proliferation via acting through the MAPK/ERK pathway. Collectively, these results reveal an undiscovered role of Sema3E in promoting pancreatic cancer pathogenesis, suggesting that Sema3E may be a suitable prognostic marker and therapeutic target for pancreatic cancer.


Assuntos
Neoplasias Pancreáticas/patologia , Semaforinas/fisiologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Neoplasias Pancreáticas/mortalidade , Semaforinas/análise
10.
Biochem Biophys Res Commun ; 479(4): 610-614, 2016 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-27697528

RESUMO

Semaphorins are essential for the functions in the regulation of cell migration. SEMA 4A has been proven to play a prominent role in immune function and angiogenesis. However, whether SEMA 4A is involved in HCC chemoresistance is unclear. We investigated the role of SEMA 4A in HCC chemoresistance and the underlying mechanisms. We tested the doxorubicin sensitivity of the Huh7, and Hep-G2 HCC cell lines. Immunofluorescence and Western blot were used to detect the location and expression of EMT-related protein, such as, E-cadherin, Vimentin, and SEMA4A expression. Microarray data showed that SEMA 4A and SEMA 3F increased most dramatically under DOX treatment. Kncokdown of SEMA 4A in hepatoma cells can reduce EMT process. Expectedly, depletion of SEMA 4A also reversed EMT and increased the DOX sensitivity. SEMA 4A confers doxorubicin resistance on HCC by inducing epithelial-mesenchymal transition (EMT).


Assuntos
Antibióticos Antineoplásicos/farmacologia , Carcinoma Hepatocelular/patologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/patologia , Semaforinas/fisiologia , Antígenos CD , Caderinas/metabolismo , Movimento Celular , Técnicas de Silenciamento de Genes , Células Hep G2 , Humanos , Semaforinas/genética , Vimentina/metabolismo
11.
Panminerva Med ; 58(3): 197-205, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27120047

RESUMO

BACKGROUND: MicroRNAs play an important role in cancer development. Deregulation of microRNAs can lead to tumorigenesis. Class 3 semaphorin, semaphorin 3E (Sema3E), has been shown to be implicated in tumor growth and metastasis. The role of miR-4282 in regulating colorectal carcinoma and its correlation to Sema3E remain uncertain. METHODS: Real-time quantitative reverse transcription polymerase chain reaction was used to detect the levels of miR-4282 and Sema3E in colorectal carcinoma cells and colorectal tumor tissues. Sema3E protein level in cell lines and human tissues was analyzed by western blot Transient transfections of miR-4282 inhibitor or mimics were conducted to silence or overexpress miR-4282. Sema3E siRNA was transfected to knockdown Sema3E in tumor cell lines. MTT assay was employed to measure colorectal tumor cell growth. Migration and invasion of the cells were examined by trans-well assays. Luciferase reporter assays were performed to confirm miR-4282 targeted at Sema3E. RESULTS: In the present study, reduced miR-4282 expression was observed in the colorectal carcinoma cell lines and human carcinoma tissues in comparison with normal human colon cells (P<0.05) or matched non-tumor tissues (P<0.05), whereas, Sema3E was up-regulated in colorectal carcinoma cells lines (P<0.05) and human colorectal tumor tissues (P<0.05). MiR-4282 was then reduced by the inhibitor and overexpressed by its mimics transfection. It was found that miR-4282 inhibition promoted cell growth, migration and invasion (P<0.05) of HT29 and HCT116 colorectal carcinoma cells while miR-4282 overexpression suppressed cell growth and mobility (P<0.05). Sema3E was predicted as a target of miR-4282 in miRDB database. We found that miR-4282 overexpression significantly reduced luciferase activity of pRL-Sema3E-3'-UTR (P<0.05), but failed to alter the activity of pRL-sema3E-3'-UTR-mutation. Also, miR4282 overexpression suppressed Sema3E expression in the colorectal carcinoma cell lines. To further confirm the role of Sema3E suppression in the function of the colorectal carcinoma cells by miR-4282, HT29 and HCT116 cells were transfected with Sema3E siRNA. We found that cell growth, migration and invasion of HT29 and HCT116 cells were dramatically inhibited by Sema3E knockdown (P<0.05). CONCLUSIONS: Our findings suggested that miR-4282 is a tumor suppressor in colorectal carcinoma cells and exerted its inhibitory effect on the tumor cells through targeting Sema3E by inhibiting Sema3E translation or enhancing Sema3E mRNA degradation. Thus, manipulation of miR-4282 and interfere with Sema3E might represent a potential target for the treatment of colorectal cancer.


Assuntos
Proliferação de Células , Neoplasias Colorretais/patologia , Genes Supressores de Tumor/fisiologia , MicroRNAs/fisiologia , Semaforinas/antagonistas & inibidores , Linhagem Celular Tumoral , Movimento Celular , Neoplasias Colorretais/genética , Humanos , Invasividade Neoplásica , Semaforinas/análise , Semaforinas/genética , Semaforinas/fisiologia
12.
J Immunol ; 196(1): 459-68, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26597008

RESUMO

Dendritic cell (DC) migration is essential for efficient host defense against pathogens and cancer, as well as for the efficacy of DC-based immunotherapies. However, the molecules that induce the migratory phenotype of DCs are poorly defined. Based on a large-scale proteome analysis of maturing DCs, we identified the GPI-anchored protein semaphorin 7A (Sema7A) as being highly expressed on activated primary myeloid and plasmacytoid DCs in human and mouse. We demonstrate that Sema7A deficiency results in impaired chemokine CCL21-driven DC migration in vivo. Impaired formation of actin-based protrusions, resulting in slower three-dimensional migration, was identified as the mechanism underlying the DC migration defect. Furthermore, we show, by atomic force microscopy, that Sema7A decreases adhesion strength to extracellular matrix while increasing the connectivity of adhesion receptors to the actin cytoskeleton. This study demonstrates that Sema7A controls the assembly of actin-based protrusions that drive DC migration in response to CCL21.


Assuntos
Citoesqueleto de Actina/metabolismo , Antígenos CD/fisiologia , Movimento Celular/fisiologia , Quimiocina CCL21/metabolismo , Células Dendríticas/fisiologia , Matriz Extracelular/metabolismo , Semaforinas/fisiologia , Animais , Antígenos CD/genética , Adesão Celular , Movimento Celular/genética , Células Cultivadas , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/fisiologia , Humanos , Camundongos , Camundongos Knockout , Microscopia de Força Atômica , Interferência de RNA , RNA Interferente Pequeno , Semaforinas/genética
13.
J Immunol ; 195(3): 934-43, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26116513

RESUMO

Mammalian target of rapamycin (mTOR) plays crucial roles in activation and differentiation of diverse types of immune cells. Although several lines of evidence have demonstrated the importance of mTOR-mediated signals in CD4(+) T cell responses, the involvement of mTOR in CD8(+) T cell responses is not fully understood. In this study, we show that a class IV semaphorin, SEMA4A, regulates CD8(+) T cell activation and differentiation through activation of mTOR complex (mTORC) 1. SEMA4A(-/-) CD8(+) T cells exhibited impairments in production of IFN-γ and TNF-α and induction of the effector molecules granzyme B, perforin, and FAS-L. Upon infection with OVA-expressing Listeria monocytogenes, pathogen-specific effector CD8(+) T cell responses were significantly impaired in SEMA4A(-/-) mice. Furthermore, SEMA4A(-/-) CD8(+) T cells exhibited reduced mTORC1 activity and elevated mTORC2 activity, suggesting that SEMA4A is required for optimal activation of mTORC1 in CD8(+) T cells. IFN-γ production and mTORC1 activity in SEMA4A(-/-) CD8(+) T cells were restored by administration of recombinant Sema4A protein. In addition, we show that plexin B2 is a functional receptor of SEMA4A in CD8(+) T cells. Collectively, these results not only demonstrate the role of SEMA4A in CD8(+) T cells, but also reveal a novel link between a semaphorin and mTOR signaling.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Ativação Linfocitária/imunologia , Complexos Multiproteicos/imunologia , Proteínas do Tecido Nervoso/fisiologia , Semaforinas/fisiologia , Serina-Treonina Quinases TOR/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Proliferação de Células , Proteína Ligante Fas/biossíntese , Granzimas/biossíntese , Interferon gama/biossíntese , Listeria monocytogenes/imunologia , Listeriose/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Citotóxicas Formadoras de Poros/biossíntese , Ligação Proteica/imunologia , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Semaforinas/genética , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
14.
Congenit Anom (Kyoto) ; 55(1): 26-30, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385160

RESUMO

Semaphorins and their receptors, neuropilins and plexins, were initially characterized as a modulator of axonal guidance during development, but are now recognized as a regulator of a wide range of developmental events including morphogenesis and angiogenesis, and activities of the immune system. Owing to the development of next-generation sequencing technologies together with other useful DNA assays, it has also become clear that semaphorin signaling plays a crucial role in many congenital diseases such as retinal degeneration and congenital heart defects. This review summarizes the recent knowledge about the relationship between a variety of congenital diseases and semaphorin signaling.


Assuntos
Semaforinas/fisiologia , Transdução de Sinais , Animais , Síndrome CHARGE/metabolismo , Doença de Hirschsprung/metabolismo , Humanos , Síndrome de Kallmann/metabolismo , Degeneração Retiniana/metabolismo , Síndrome de Cimitarra/metabolismo , Tetralogia de Fallot/metabolismo , Persistência do Tronco Arterial/metabolismo
15.
Cell Rep ; 9(5): 1812-1826, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25464848

RESUMO

Different cancer cell compartments often communicate through soluble factors to facilitate tumor growth. Glioma stem cells (GSCs) are a subset of tumor cells that resist standard therapy to contribute to disease progression. How GSCs employ a distinct secretory program to communicate with and nurture each other over the nonstem tumor cell (NSTC) population is not well defined. Here, we show that GSCs preferentially secrete Sema3C and coordinately express PlexinA2/D1 receptors to activate Rac1/nuclear factor (NF)-κB signaling in an autocrine/paracrine loop to promote their own survival. Importantly, Sema3C is not expressed in neural progenitor cells (NPCs) or NSTCs. Disruption of Sema3C induced apoptosis of GSCs, but not NPCs or NSTCs, and suppressed tumor growth in orthotopic models of glioblastoma. Introduction of activated Rac1 rescued the Sema3C knockdown phenotype in vivo. Our study supports the targeting of Sema3C to break this GSC-specific autocrine/paracrine loop in order to improve glioblastoma treatment, potentially with a high therapeutic index.


Assuntos
Sobrevivência Celular , Glioblastoma/patologia , Células-Tronco Neoplásicas/fisiologia , Semaforinas/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Apoptose , Carcinogênese/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Ativação Enzimática , Expressão Gênica , Glioblastoma/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Glicoproteínas de Membrana , Camundongos Transgênicos , Transplante de Neoplasias , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular/metabolismo
16.
Nat Rev Drug Discov ; 13(8): 603-21, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25082288

RESUMO

Semaphorins are membrane-bound or diffusible factors that regulate key cellular functions and are involved in cell-cell communication. Most of the effects of semaphorins are mediated by plexins. Work over the past decade has revealed crucial functions of the semaphorin-plexin system in mammalian physiology. It has also become clear that semaphorins and plexins have important roles in many pathophysiological processes, including cancer, immunological diseases and bone disorders, and that they represent novel targets for drugs to prevent or treat various diseases. This Review summarizes the functions of the mammalian semaphorin-plexin system as well as its role in diseases and discusses emerging strategies to pharmacologically target semaphorin-plexin signalling.


Assuntos
Moléculas de Adesão Celular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Semaforinas/fisiologia , Animais , Doenças Ósseas/fisiopatologia , Osso e Ossos/fisiologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/fisiologia , Sistema Nervoso Central/fisiologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/fisiopatologia , Regeneração Nervosa/fisiologia , Receptores de Superfície Celular/efeitos dos fármacos , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Clin Calcium ; 24(6): 853-61, 2014 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-24870836

RESUMO

Bone is constantly renewed by the balanced action of osteoblastic bone formation and osteoclastic bone resorption both of which mainly occur at the bone surface. This restructuring process called "bone remodeling" is important not only for normal bone mass and strength, but also for mineral homeostasis. Coupling has been understood as a balanced induction of osteoblastic bone formation in response to osteoclastic bone resorption. An imbalance of this coupling is often linked to various bone diseases. TGF-ß and IGF released from bone matrix during osteoclastic bone resorption are the favored candidates as classical coupling factor. Recently, several reports suggest that osteoclast-derived molecules/cytokines (clastokine) mediate directional signaling between osteoblasts and osteoclasts into the bone microenvironment. Thus, the elucidation of the regulatory mechanisms involved in bone cell communication and coupling is critical for a deeper understanding of the skeletal system in health and disease.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/fisiologia , Comunicação Celular/genética , Comunicação Celular/fisiologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Animais , Antígenos CD/fisiologia , Proteína Morfogenética Óssea 6/fisiologia , Remodelação Óssea/genética , Remodelação Óssea/fisiologia , Microambiente Celular/genética , Microambiente Celular/fisiologia , Homeostase/genética , Homeostase/fisiologia , Humanos , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Camundongos , Fatores de Transcrição NFATC/metabolismo , Osteoprotegerina/fisiologia , Pró-Proteína Convertases/fisiologia , Ligante RANK/fisiologia , Semaforina-3A/fisiologia , Semaforinas/fisiologia , Serina Endopeptidases/fisiologia , Somatomedinas/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Via de Sinalização Wnt/fisiologia
18.
Tumour Biol ; 35(5): 4949-55, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24474252

RESUMO

Sema domain of semaphorin 4B (SEMA4B), which is an interacting protein of LNM35, plays an important role in lung cancer invasion. However, the regulation mechanism of this protein is completely unknown. Here, we report that hypoxia and hypoxia mimic reagent could downregulate the expression of SEMA4B in human non-small cell lung cancer (NSCLC) lines. We provide evidences that SEMA4B is a direct target of hypoxia-inducible factor 1 (HIF-1). Silencing the expression of HIF-1α in cancer cells by RNA interference abolished hypoxia-repressed SEMA4B expression. Using luciferase reporter assay, we showed that HIF-1α recognized a hypoxia-responsive element (HRE) of SEMA4B gene, which is required for HIF-1-repressed SEMA4B expression. Moreover, ectopic expression of SEMA4B abolished invasion of hypoxia-induced NSCLC cells. Taken together, these data would shed novel insights on the mechanisms for invasion of hypoxia-induced NSCLC cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Hipóxia Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias Pulmonares/patologia , Semaforinas/fisiologia , Linhagem Celular Tumoral , Humanos , Invasividade Neoplásica , Regiões Promotoras Genéticas , Semaforinas/genética
19.
Oncology ; 86(1): 46-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24401553

RESUMO

Since the approval of the first antiangiogenic agent bevacizumab, a neutralizing antibody against the vascular endothelial growth factor (VEGF), antiangiogenic therapies augmented the standard armamentarium of anticancer therapies and proved their clinical efficacy. Nevertheless, antiangiogenic strategies could not fulfill the expected hopes. In clinical routine, therapy responses to antiangiogenic therapies were mostly transient and most of the patients developed evasive resistance mechanisms during therapy. Further, no predictive biomarker for therapy response could be developed, hampering the clinical development of these agents and triggering skepticism. In the past years, knowledge on the biology of angiogenesis increased and the role of tumor hypoxia was better characterized and identified as the driver for angiogenic regulation mechanisms. Besides VEGF, new angiogenic and antiangiogenic factors were characterized and the process of endothelial cell migration, proliferation and vessel formation was better elucidated. Thus, a strong connection to neural development and axon guidance molecules like netrins, Slit proteins, semaphorins, ephrins and their cognate receptors UNC5, Robo1-4, neuropilin and EphB was identified. The aim of this review is to present the importance of these axon guidance molecules with special focus on Robo4 and semaphorins in tumor angiogenesis and to highlight their value as potential targets for new antiangiogenic therapies.


Assuntos
Inibidores da Angiogênese/farmacologia , Axônios/metabolismo , Semaforinas/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Neoplasias/irrigação sanguínea , Fatores de Crescimento Neural/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Netrina-1 , Receptores de Superfície Celular/fisiologia , Transdução de Sinais , Proteínas Supressoras de Tumor/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
20.
Cancer Immunol Immunother ; 63(2): 81-99, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24263240

RESUMO

Neuropilin 1 (NRP1) is a transmembrane glycoprotein that acts as a co-receptor for a number of extracellular ligands including class III/IV semaphorins, certain isoforms of vascular endothelial growth factor and transforming growth factor beta. An exact understanding of the role of NRP1 in the immune system has been obscured by the differences in NRP1 expression observed between mice and humans. In mice, NRP1 is selectively expressed on thymic-derived Tregs and greatly enhances immunosuppressive function. In humans, NRP1 is expressed on plasmacytoid dendritic cells (pDCs) where it aids in priming immune responses and on a subset of T regulatory cells (Tregs) isolated from secondary lymph nodes. Preliminary studies that show NRP1 expression on T cells confers enhanced immunosuppressive activity. However, the mechanism by which this activity is mediated remains unclear. NRP1 expression has also been identified on activated T cells and Tregs isolated from inflammatory microenvironments, suggesting NRP1 might represent a novel T cell activation marker. Of clinical interest, NRP1 may enhance Treg tumour infiltration and a decrease in NRP1+ Tregs correlates with successful chemotherapy, suggesting a specific role for NRP1 in cancer pathology. As a therapeutic target, NRP1 allows simultaneous targeting of NRP1-expressing tumour vasculature, NRP1+ Tregs and pDCs. With the development of anti-NRP1 monoclonal antibodies and cell-penetrating peptides, NRP1 represents a promising new target for cancer therapies. This paper reviews current knowledge on the role and function of NRP1 in Tregs and pDCs, both in physiological and cancer settings, as well as its potential as a therapeutic target in cancer.


Assuntos
Neoplasias/terapia , Neuropilina-1/fisiologia , Animais , Células Dendríticas/imunologia , Humanos , Ativação Linfocitária , Neoplasias/imunologia , Neuropilina-1/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/fisiologia , Semaforina-3A/fisiologia , Semaforinas/fisiologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA