Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Int J Cardiol ; 344: 13-24, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34534604

RESUMO

Microvascular dysfunction caused by cardiac ischemia-reperfusion (I/R) leads to multiple severe cardiac adverse events, such as heart failure and ventricular modeling, which plays a critical role in outcomes. Though marrow mesenchymal stem cell (MSC) therapy has been proven effective for attenuating I/R injury, the limitations of clinical feasibility cannot be ignored. Since exosomes are recognized as the main vehicles for MSCs paracrine effects, we assumed that MSC-derived exosomes could prevent microvascular dysfunction and further protect cardiac function. By establishing a rat cardiac I/R model in vivo and a cardiac microvascular endothelial cells (CMECs) hypoxia-reperfusion (H/R) model in vitro, we demonstrated that MSC-derived exosomes enhanced microvascular regeneration under stress, inhibited fibrosis development, and eventually improved cardiac function through platelet-derived growth factor receptor-ß (PDGFR-ß) modulation. Furthermore, we found that MSC-derived exosomes possessed better therapeutic effects than MSCs themselves.


Assuntos
Cardiomiopatias , Exossomos , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Animais , Medula Óssea , Células Endoteliais , Fibrose , Isquemia , Células-Tronco Mesenquimais , Microcirculação , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Reperfusão
2.
Am J Med Genet A ; 185(3): 999-1003, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33382209

RESUMO

Skeletal overgrowth accompanied by de novo heterozygous activating mutations in PDGFRB (platelet-derived growth factor receptor beta), that is, p.Pro584Arg and p.Trp566Arg, defines Kosaki overgrowth syndrome (OMIM #616592). Emerging evidence suggests a role of PDGFRB in the genesis of cerebral aneurysms. The delineation of the range and progression of the vascular phenotype of Kosaki overgrowth syndrome is urgently needed. Herein, we conducted subsequent analyses of serial neurovascular imaging studies of two original patients with a de novo heterozygous mutation in PDGFRB, that is, p.Pro584Arg. The analysis showed the progressive dilation of basilar and vertebral arteries and coronary arteries commencing during the teenage years and early 20s. The radiographic appearance of the basilar vertebral aneurysms showed signs of arterial wall dilation, compatible with the known vascular pathology of vascular-type Ehlers-Danlos syndrome and Loeys-Dietz syndrome. The dolichoectasia in cerebrovascular arteries can lead to fatal complications, even with neurosurgical interventions. To prevent the progression of artery dilation, preventative and therapeutic medical measures using tyrosine kinase inhibitors may be necessary in addition to optimal control of the systemic blood pressure. Kosaki overgrowth syndrome is a clinically recognizable syndrome that can exhibit progressive dilatory and tortuous vascular changes in basilar/vertebral and coronary arteries as early as in the teenage years. We recommend careful counseling regarding the risk of future vascular complications, optimal blood pressure control, and regular systemic vascular screening during follow-up examinations.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Aneurisma Coronário/genética , Mutação com Ganho de Função , Aneurisma Intracraniano/genética , Mutação de Sentido Incorreto , Mutação Puntual , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Insuficiência Vertebrobasilar/genética , Adolescente , Idade de Início , Substituição de Aminoácidos , Aneurisma/genética , Cegueira/etiologia , Calcinose/etiologia , Doenças das Artérias Carótidas/genética , Aneurisma Coronário/diagnóstico por imagem , Progressão da Doença , Feminino , Perda Auditiva Unilateral/etiologia , Humanos , Aneurisma Intracraniano/diagnóstico por imagem , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Síndrome , Insuficiência Vertebrobasilar/diagnóstico por imagem
3.
Theranostics ; 10(3): 1122-1135, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31938055

RESUMO

PDGF-BB/PDGFRß signaling plays an important role during vascularization by mediating pericyte recruitment to the vasculature, promoting the integrity and function of vessels. Until now it has not been possible to assess the specific role of PDGFRß signaling in tumor progression and angiogenesis due to lack of appropriate animal models and molecular tools. Methods: In the present study, we used a transgenic knock-in mouse strain carrying a silent mutation in the PDGFRß ATP binding site that allows specific targeting of PDGFRß using the compound 1-NaPP1. To evaluate the impact of selective PDGFRß inhibition of stromal cells on tumor growth we investigated four tumor cell lines with no or low PDGFRß expression, i.e. Lewis lung carcinoma (LLC), EO771 breast carcinoma, B16 melanoma and a version of B16 that had been engineered to overexpress PDGF-BB (B16/PDGF-BB). Results: We found that specific impairment of PDGFRß kinase activity by 1-NaPP1 treatment efficiently suppressed growth in tumors with high expression of PDGF-BB, i.e. LLC and B16/PDGF-BB, while the clinically used PDGFRß kinase inhibitor imatinib did not suppress tumor growth. Notably, tumors with low levels of PDGF-BB, i.e. EO771 and B16, neither responded to 1-NaPP1 nor to imatinib treatment. Inhibition of PDGFRß by either drug impaired tumor vascularization and also affected pericyte coverage; however, specific targeting of PDGFRß by 1-NaPP1 resulted in a more pronounced decrease in vessel function with increased vessel apoptosis in high PDGF-BB expressing tumors, compared to treatment with imatinib. In vitro analysis of PDGFRß ASKA mouse embryo fibroblasts and the mesenchymal progenitor cell line 10T1/2 revealed that PDGF-BB induced NG2 expression, consistent with the in vivo data. Conclusion: Specific targeting of PDGFRß signaling significantly inhibits tumor progression and angiogenesis depending on PDGF-BB expression. Our data suggest that targeting PDGFRß in the tumor stroma could have therapeutic value in patients with high tumor PDGF-BB expression.


Assuntos
Antineoplásicos/uso terapêutico , Mesilato de Imatinib/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Embrião de Mamíferos/citologia , Humanos , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias/metabolismo , Neovascularização Patológica , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Células Estromais
4.
J Cereb Blood Flow Metab ; 39(7): 1369-1380, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-29480757

RESUMO

Platelet-derived growth factor receptor-ß (PDGFR-ß) has been reported to promote phenotypic transformation of vascular smooth muscle cells (VSMCs). The purpose of this study was to investigate the role of the PDGFR-ß/IRF9/SIRT-1/NF-κB pathway in VSMC phenotypic transformation after subarachnoid hemorrhage (SAH). SAH was induced using the endovascular perforation model in Sprague-Dawley rats. PDGFR-ß small interfering RNA (siRNA) and IRF9 siRNA were injected intracerebroventricularly 48 h before SAH. SIRT1 activator (resveratrol) and inhibitor (EX527) were administered intraperitoneally 1 h after SAH induction. Twenty-four hours after SAH, the VSMC contractile phenotype marker α-smooth muscle actin (α-SMA) decreased, whereas the VSMC synthetic phenotype marker embryonic smooth muscle myosin heavy chain (Smemb) increased. Both PDGFR-ß siRNA and IRF9 siRNA attenuated the induction of nuclear factor-κB (NF-κB) and enhanced the expression of α-SMA. The SIRT1 activator (resveratrol) preserved VSMC contractile phenotype, significantly alleviated neurological dysfunction, and reduced brain edema. However, these beneficial effects of PDGFR-ß siRNA, IRF9 siRNA and resveratrol were abolished by the SIRT1 inhibitor (EX527). This study shows that PDGFR-ß/IRF9/SIRT-1/NF-κB signaling played a role in the VSMC phenotypic transformation after SAH. Inhibition of this signaling cascade preserved the contractile phenotype of VSMCs, thereby improving neurological outcomes following SAH.


Assuntos
Fator Gênico 3 Estimulado por Interferon, Subunidade gama/fisiologia , Músculo Liso Vascular/fisiopatologia , NF-kappa B/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Sirtuína 1/fisiologia , Hemorragia Subaracnóidea/fisiopatologia , Actinas/análise , Animais , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Masculino , Músculo Liso Vascular/química , Cadeias Pesadas de Miosina/análise , Fenótipo , RNA Interferente Pequeno/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Resveratrol/farmacologia , Transdução de Sinais/fisiologia , Sirtuína 1/antagonistas & inibidores , Hemorragia Subaracnóidea/etiologia
5.
J Assoc Res Otolaryngol ; 19(4): 363-379, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29869048

RESUMO

Normal blood supply to the cochlea is critical for hearing. Noise damages auditory sensory cells and has a marked effect on the microvasculature in the cochlear lateral wall. Pericytes in the stria vascularis (strial pericytes) are particularly vulnerable and sensitive to acoustic trauma. Exposure of NG2DsRedBAC transgenic mice (6-8 weeks old) to wide-band noise at a level of 120 dB for 3 h per day for 2 consecutive days produced a significant hearing threshold shift and caused pericytes to protrude and migrate from their normal endothelial attachment sites. The pericyte migration was associated with increased expression of platelet-derived growth factor beta (PDGF-BB). Blockade of PDGF-BB signaling with either imatinib, a potent PDGF-BB receptor (PDGFR) inhibitor, or APB5, a specific PDGFRß blocker, significantly attenuated the pericyte migration from strial vessel walls. The PDGF-BB-mediated strial pericyte migration was further confirmed in an in vitro cell migration assay, as well as in an in vivo live animal model used in conjunction with confocal fluorescence microscopy. Pericyte migration took one of two different forms, here denoted protrusion and detachment. The protrusion is characterized by pericytes with a prominent triangular shape, or pericytes extending fine strands to neighboring capillaries. The detachment is characterized by pericyte detachment and movement away from vessels. We also found the sites of pericyte migration highly associated with regions of vascular leakage. In particular, under transmission electron microscopy (TEM), multiple vesicles at the sites of endothelial cells with loosely attached pericytes were observed. These data show that cochlear pericytes are markedly affected by acoustic trauma, causing them to display abnormal morphology. The effect of loud sound on pericytes is mediated by upregulation of PDGF-BB. Normal functioning pericytes are required for vascular stability.


Assuntos
Becaplermina/fisiologia , Ruído , Pericitos/fisiologia , Transdução de Sinais/fisiologia , Estria Vascular/fisiologia , Animais , Permeabilidade Capilar , Movimento Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia
6.
Proc Natl Acad Sci U S A ; 114(35): E7262-E7271, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28808001

RESUMO

The dimeric 44-residue E5 protein of bovine papillomavirus is the smallest known naturally occurring oncoprotein. This transmembrane protein binds to the transmembrane domain (TMD) of the platelet-derived growth factor ß receptor (PDGFßR), causing dimerization and activation of the receptor. Here, we use Rosetta membrane modeling and all-atom molecular dynamics simulations in a membrane environment to develop a chemically detailed model of the E5 protein/PDGFßR complex. In this model, an active dimer of the PDGFßR TMD is sandwiched between two dimers of the E5 protein. Biochemical experiments showed that the major PDGFßR TMD complex in mouse cells contains two E5 dimers and that binding the PDGFßR TMD to the E5 protein is necessary and sufficient to recruit both E5 dimers into the complex. These results demonstrate how E5 binding induces receptor dimerization and define a molecular mechanism of receptor activation based on specific interactions between TMDs.


Assuntos
Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular , Transformação Celular Viral , Dimerização , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Conformação Molecular , Papillomaviridae/metabolismo , Infecções por Papillomavirus , Multimerização Proteica , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
7.
Int J Hematol ; 105(5): 697-701, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28000100

RESUMO

Abnormal platelet-derived growth factor receptor (PDGFR)-mediated signaling may cause hematologic neoplasm. The PDGFR beta (PDGFRB) gene, located at chromosome band 5q31-33, forms a fusion gene as a result of chromosome translocation. Although patients with PDGFRB rearrangement mostly present with myeloproliferative neoplasm and eosinophilia, acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) have also been reported in this population. Treatment with imatinib mesylate alone has been shown to have excellent long-term efficacy against myeloproliferative neoplasms; however, its long-term effects on ALL and AML have not been elucidated. A 75-year-old man was diagnosed with acute myeloid leukemia having the PDGFRB and cGMP-dependent protein kinase 2 fusion gene with additional genetic abnormalities. Continuous therapy with single-agent imatinib mesylate resulted in cytogenetic remission and decreased molecular burden for 9 months; however, the leukemia subsequently recurred, and the patient died 1 year after initiation of treatment. This case report supports the importance of cytogenetic analysis during patient screening.


Assuntos
Antineoplásicos/uso terapêutico , Mesilato de Imatinib/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Idoso , Cromossomos Humanos Par 5/genética , Proteína Quinase Dependente de GMP Cíclico Tipo II/genética , Análise Citogenética , Evolução Fatal , Fusão Gênica/genética , Rearranjo Gênico/genética , Humanos , Masculino , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Indução de Remissão , Transdução de Sinais/genética , Translocação Genética/genética , Resultado do Tratamento
8.
Anticancer Res ; 36(5): 2291-5, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27127135

RESUMO

BACKGROUND/AIM: Studies developed in the field of platelet-derived growth factors/platelet-derived growth factor receptors (PDGFs/PDGFRs) inhibition have focused on the therapeutic effects on tumor cells, neglecting their potential effects on tumor blood vessels. We herein propose a differential and critic assessment of platelet-derived growth factor B (PDGF-B) and platelet-derived growth factor receptor ß (PDGFRß) in renal cell carcinoma, correlated with the four main vascular patterns previously reported by our team. MATERIALS AND METHODS: PDGF-B and PDGFRß were evaluated on 50 archival paraffin embedded specimens related to vascular endothelial growth factor (VEGF), its inhibitory isoform VEGF165b and vascular patterns. RESULTS AND CONCLUSION: Our results support the involvement of VEGF165b in the phosphorylation of PDGFRß with an inhibitory effect on endothelial proliferation and migration. The simultaneous action of PDGF-B/PDGFRß and VEGF165b on the same type of receptor may explain the resistance to antiangiogenic therapy, which depends on the degree of modulation of PDGFRß phosphorylation.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Renais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Proteínas Proto-Oncogênicas c-sis/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Becaplermina , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/química , Humanos , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/química , Neovascularização Patológica/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-sis/análise , Receptor beta de Fator de Crescimento Derivado de Plaquetas/análise , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Estudos Retrospectivos , Fator A de Crescimento do Endotélio Vascular/análise
9.
Cardiology ; 133(4): 242-56, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26752520

RESUMO

OBJECTIVE: To explore the role of platelet-derived growth factor-B (PDGF-B)/platelet-derived growth factor receptor-ß (PDGFR-ß) signaling in chronic atrial fibrillation (AF). METHODS: Thirty-nine AF patients and 33 patients with sinus rhythm (SR) were enrolled. Twenty canines were randomized into 5 groups: control, sham and AF lasting 1, 2 or 4 weeks. The AF canine models were made by rapid atrial pacing. Rat atrial fibroblasts were treated with PDGF-BB or PDGF-BB + PDGFR inhibitor AG1295, respectively. Gene expression in the right atrial appendage of patients, the left atrium of canines and rat atrial fibroblasts was measured by quantitative real-time PCR and Western blot, respectively. The degree of atrial fibrosis was evaluated by Masson trichrome staining. RESULTS: The degree of atrial fibrosis and the expression of PDGF-B, PDGFR-ß and collagen type I (COL1) in AF patients significantly increased compared to patients with SR. The degree of atrial fibrosis and the expression of PDGF-B and COL1 in canines increased progressively with the increased duration of AF. The expression of PDGFR-ß increased progressively 2 weeks after AF. PDGF-BB promoted the proliferation and COL1 secretion of rat atrial fibroblasts. AG1295 attenuated these effects. CONCLUSIONS: Our study suggests that PDGF-B/PDGFR-ß signaling, which promotes the proliferation and COL1 secretion of atrial fibroblasts, is an important contributor to atrial fibrosis in AF and may represent a novel target for the intervention of AF.


Assuntos
Fibrilação Atrial/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais , Adulto , Animais , Proliferação de Células , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Cães , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Átrios do Coração/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-sis/genética , RNA Mensageiro/metabolismo , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Regulação para Cima
10.
Yi Chuan ; 37(8): 731-40, 2015 08.
Artigo em Chinês | MEDLINE | ID: mdl-26353387

RESUMO

Idiopathic basal ganglia calcification (IBGC), also known as Fahr's disease, is an inheritable neurodegenerative syndrome characterized by mineral deposits in the basal ganglia and other brain regions. Patients with IBGC are often accompanied with movement disorders, cognitive impairment as well as psychiatric abnormalities. So far, no therapeutic drug has been developed for the treatment of IBGC. Recently, genetic studies have identified several genes associated with IBGC, including SLC20A2, PDGFRB, PDGFB, ISG15 and XPR1. Loss-of-function mutations in these genes have been associated with disturbance in phosphate homeostasis in brain regions, the dysfunction of blood-brain barrier as well as enhanced IFN-α/ß immunity. In this review, we summarize the latest research progress in the studies on molecular genetics of IBGC, and discuss the molecular mechanisms underlying the pathophysiology of mutations of different genes.


Assuntos
Doenças dos Gânglios da Base/genética , Calcinose/genética , Doenças Neurodegenerativas/genética , Citocinas/genética , Humanos , Mutação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Virais/genética , Transdução de Sinais , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Ubiquitinas/genética , Receptor do Retrovírus Politrópico e Xenotrópico
11.
Exp Dermatol ; 24(6): 468-70, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25708924

RESUMO

Embryonic hair follicle (HF) induction and formation is dependent on signalling crosstalk between the dermis and specialized dermal condensates on the mesenchymal side and epidermal cells and incipient placodes on the epithelial side, but the precise nature and succession of signals remain unclear. Platelet-derived growth factor (PDGF) signalling is involved in the development of several organs and the maintenance of adult tissues, including HF regeneration in the hair cycle. As both PDGF receptors, PDGFRα and PDGFRß, are expressed in embryonic dermis and dermal condensates, we explored in this study the role of PDGF signalling in HF induction and formation in the developing skin mesenchyme. We conditionally ablated both PDGF receptors with Tbx18(Cre) in early dermal condensates before follicle formation, and with Prx1-Cre broadly in the ventral dermis prior to HF induction. In both PDGFR double mutants, HF induction and formation ensued normally, and the pattern of HF formation and HF numbers were unaffected. These data demonstrate that mesenchymal PDGF signalling, either in the specialized niche or broadly in the dermis, is dispensable for HF induction and formation.


Assuntos
Derme/embriologia , Folículo Piloso/embriologia , Morfogênese/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/fisiologia , Animais , Derme/citologia , Derme/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Folículo Piloso/citologia , Folículo Piloso/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/fisiologia , Camundongos , Camundongos Mutantes , Modelos Animais , Morfogênese/genética , Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/genética
12.
Br J Anaesth ; 111(6): 1004-12, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23820675

RESUMO

BACKGROUND: Pain and renal dysfunction occur in sickle cell disease. Morphine used to treat pain also co-activates platelet-derived growth factor receptor-ß (PDGFR-ß), which can adversely affect renal disease. We examined the influence of morphine in mesangial cells in vitro and in mouse kidneys in vivo. METHODS: > Mouse mesangial cells treated with 1 µM morphine in vitro or kidneys of transgenic homozygous or hemizygous sickle or control mice (n=3 for each), treated with morphine (0.75, 1.4, 2.14, 2.8, 3.6, and 4.3 mg kg(-1) day(-1) in two divided doses during the first, second, third, fourth, fifth, and sixth weeks, respectively), were used. Western blotting, bromylated deoxy uridine incorporation-based cell proliferation assay, reverse transcriptase-polymerase chain reaction, immunofluorescent microscopy, and blood/urine chemistry were used to analyse signalling, cell proliferation, opioid receptor (OP) expression, and renal function. RESULTS: Morphine stimulated phosphorylation of PDGFR-ß and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) to the same extent as induced by platelet-derived growth factor-BB (PDGF-BB) and promoted a two-fold increase in mesangial cell proliferation. The PDGFR-ß inhibitor, AG1296, OP antagonists, and silencing of µ- and κ-OP abrogated morphine-induced MAPK/ERK phosphorylation and proliferation by ~100%. Morphine treatment of transgenic mice resulted in phosphorylation of PDGFR-ß, MAPK/ERK, and signal transducer and activator of transcription 3 (Stat3) in the kidneys. Morphine inhibited micturition and blood urea nitrogen (BUN) clearance and increased BUN and urinary protein in sickle mice. CONCLUSION: Morphine stimulates mitogenic signalling leading to mesangial cell proliferation and promotes renal dysfunction in sickle mice.


Assuntos
Analgésicos Opioides/farmacologia , Anemia Falciforme/metabolismo , Rim/efeitos dos fármacos , Morfina/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/toxicidade , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Rim/metabolismo , Rim/fisiopatologia , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/metabolismo , Camundongos , Camundongos Transgênicos , Morfina/administração & dosagem , Morfina/toxicidade , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptores Opioides/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transtornos Urinários/induzido quimicamente , Transtornos Urinários/fisiopatologia
13.
Virchows Arch ; 463(1): 57-65, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23748876

RESUMO

Platelet derived growth factor receptor (PDGFR) is a membrane tyrosine-kinase receptor required for fibroblast activation in stromal proliferations. In order to assess the role of PDGFR in myelofibrosis (MF) we determined in 60 bone marrow biopsies the occurrence and distribution of its α and ß subunits in normal and fibrotic bone marrow stroma using immunohistochemistry, and compared this with the grade of MF determined by Gömöri's silver impregnation. PDGF receptor subunits were found to be differentially expressed in the marrow parenchyma. PDGFRα expression identified megakaryocytes, endosteal and endothelial cells while PDGFRß was virtually absent from inter-trabecular spaces in normal marrow. Activated fibroblasts characteristic for MF intensely expressed PDGFRß but only a moderate increase in PDGFRα expression was seen. Semi-quantitative PDGFRß immunoreactivity scores correlated well with the grade of MF in the vast majority of the MF cases (Spearman r= 0.83). Altogether, 21/60 (35.0%) cases showed a relative increase of PDGFRß expression, compared to the MF grade, suggesting that increased stromal PDGFRß expression occurs early and precedes reticulin and collagen fiber production during fibroblast activation. In conclusion, bone marrow PDGFRß expression closely correlates with the grade of MF. Increased immunoreactivity for PDGFRß occurs already in the prefibrotic stage of the disease and might allow a functional classification of the bone marrow stromal reaction.


Assuntos
Células-Tronco Mesenquimais/química , Mielofibrose Primária/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Antígenos CD34/análise , Humanos , Imuno-Histoquímica , Mielofibrose Primária/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/análise
14.
Angiogenesis ; 16(4): 889-902, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23793459

RESUMO

Mice lacking histidine-rich glycoprotein (HRG) display an accelerated angiogenic switch and larger tumors-a phenotype caused by enhanced platelet activation in the HRG-deficient mice. Here we show that platelets induce molecular changes in the pre-tumorigenic environment in HRG-deficient mice, promoting cell survival, angiogenesis and epithelial-to-mesenchymal transition (EMT) and that these effects involved signaling via TBK1, Akt2 and PDGFRß. These early events subsequently translate into an enhanced rate of spontaneous metastasis to distant organs in mice lacking HRG. Later in tumor development characteristic features of pathological angiogenesis, such as decreased perfusion and pericyte coverage, are more pronounced in HRG-deficient mice. At this stage, platelets are essential to support the larger tumor volumes formed in mice lacking HRG by keeping their tumor vasculature sufficiently functional. We conclude that HRG-deficiency promotes tumor progression via enhanced platelet activity and that platelets play a dual role in this process. During early stages of transformation, activated platelets promote tumor cell survival, the angiogenic switch and invasiveness. In the more progressed tumor, platelets support the enhanced pathological angiogenesis and hence increased tumor growth seen in the absence of HRG. Altogether, our findings strengthen the notion of HRG as a potent tumor suppressor, with capacity to attenuate the angiogenic switch, tumor growth, EMT and subsequent metastatic spread, by regulating platelet activity.


Assuntos
Plaquetas/fisiologia , Carcinoma/irrigação sanguínea , Transição Epitelial-Mesenquimal/fisiologia , Metástase Neoplásica/fisiopatologia , Neovascularização Patológica/fisiopatologia , Neoplasias Pancreáticas/irrigação sanguínea , Proteínas/fisiologia , Transdução de Sinais/fisiologia , Proteínas Supressoras de Tumor/deficiência , Animais , Carcinoma/genética , Carcinoma/fisiopatologia , Carcinoma/secundário , Transformação Celular Neoplásica , Microambiente Celular , Progressão da Doença , Insulinoma/genética , Insulinoma/patologia , Insulinoma/fisiopatologia , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Metástase Neoplásica/genética , Neovascularização Patológica/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/fisiopatologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas/genética , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Carga Tumoral , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia
15.
Res Vet Sci ; 94(3): 596-601, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23122810

RESUMO

Bovine cutaneous fibropapillomas are benign skin tumours formed by proliferation of epidermal keratinocytes and dermal fibroblasts caused by bovine papillomaviruses (BPVs). BPV E5 oncoprotein plays a key role in neoplastic cell transformation by specifically binding to the platelet derived growth factor beta receptor (PDGFßR) causing its phosphorylation and activation of proliferation and survival signal transduction pathways, among these phosphatidyl inositol-3-kinase (PI3K)/Akt and Ras-mitogen-activated-protein-kinase-Erk (Ras-MAPK-Erk) pathways. The aim of this study was to investigate the expression of PDGFßR, its phosphorylation status and expression of the downstream molecules phospho-Akt (pAkt) and phospho-Erk (pErk), in naturally occurring bovine cutaneous fibropapillomas. By immunohistochemistry on serial sections we showed cytoplasmic co-expression of the PDGFßR and E5 protein in neoplastic tissue. Western blot analysis revealed that PDGFßR was phosphorylated in higher amount in tumour samples compared to normal skin. pAkt, but not pErk, was also overexpressed in tumour samples. These findings may provide new insights into the aetiopathogenic mechanisms underlying naturally occurring bovine fibropapillomas and contribute to understanding the molecular scenario underlying BPV induced tumourigenesis.


Assuntos
Papillomavirus Bovino 1/metabolismo , Doenças dos Bovinos/virologia , Infecções por Papillomavirus/veterinária , Receptor beta de Fator de Crescimento Derivado de Plaquetas/biossíntese , Transdução de Sinais/fisiologia , Animais , Western Blotting/veterinária , Bovinos , Doenças dos Bovinos/metabolismo , Eletroforese em Gel de Poliacrilamida/veterinária , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Oncogênica v-akt/metabolismo , Proteína Oncogênica v-akt/fisiologia , Proteínas Oncogênicas Virais/metabolismo , Proteínas Oncogênicas Virais/fisiologia , Infecções por Papillomavirus/metabolismo , Fosforilação , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia
16.
Am J Pathol ; 180(6): 2590-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22507839

RESUMO

The acute response to vascular cell injury, which underpins vasculo-occlusive pathologies such as atherogenesis and restenosis after percutaneous coronary intervention, involves a complex series of molecular events that alter patterns of gene expression and favor a synthetic phenotype. One transcription factor that has been implicated in this process is the evolutionarily conserved mammalian stress response pathway regulator activating transcription factor 4 (ATF-4). Here, we show for the first time that both mRNA and protein levels of ATF-4 are induced in smooth muscle cells (SMCs) by the potent migratory factor PDGF-BB through PDGFR-ß. PDGF-BB also stimulates the expression of tenascin-C (TN-C), an extracellular matrix glycoprotein that regulates the activity of focal adhesion complexes, facilitating the SMC migration that underlies negative vascular remodeling in response to injury. Overexpression of ATF-4 increased transcript levels of the four TN-C isoforms in rat vascular SMCs, and ATF-4 knockdown inhibited PDGF-BB-inducible TN-C expression in vitro and injury-inducible TN-C protein expression in the balloon-injured rat artery wall. Furthermore, we show that ATF-4 is required for PDGF-BB-inducible SMC migration in response to injury. PDGF-BB-induced migration was also compromised in ATF-4 null mEFs, and this effect was rescued by the addition of TN-C. Our findings thus demonstrate the role of ATF-4 in both injury- and PDGF-BB-inducible TN-C expression and cell migration.


Assuntos
Fator 4 Ativador da Transcrição/biossíntese , Proteínas Proto-Oncogênicas c-sis/fisiologia , Tenascina/biossíntese , Fator 4 Ativador da Transcrição/deficiência , Fator 4 Ativador da Transcrição/genética , Animais , Becaplermina , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/fisiopatologia , Artéria Carótida Primitiva/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Inativação Gênica , Camundongos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Tenascina/genética , Cicatrização/fisiologia
17.
Dev Cell ; 22(3): 651-9, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22387002

RESUMO

The placenta is a hematopoietic organ that supports hematopoietic stem/progenitor cell (HSPC) generation and expansion without promoting differentiation. We identified PDGF-B signaling in trophoblasts as a key component of the unique placental hematopoietic microenvironment that protects HSPCs from premature differentiation. Loss of PDGF-B or its receptor, PDGFRß, induced definitive erythropoiesis in placental labyrinth vasculature. This was evidenced by accumulation of CFU-Es and actively proliferating definitive erythroblasts that clustered around central macrophages, highly reminiscent of erythropoiesis in the fetal liver. Ectopic erythropoiesis was not due to a requirement of PDGF-B signaling in hematopoietic cells but rather in placental trophoblasts, which upregulated Epo in the absence of PDGF-B signaling. Furthermore, overexpression of hEPO specifically in the trophoblasts in vivo was sufficient to convert the placenta into an erythropoietic organ. These data provide genetic evidence of a signaling pathway that is required to restrict erythroid differentiation to specific anatomical niches during development.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Placenta/fisiologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Transdução de Sinais/fisiologia , Trofoblastos/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Precursoras Eritroides/fisiologia , Eritropoese/fisiologia , Eritropoetina/fisiologia , Feminino , Células-Tronco Hematopoéticas/citologia , Humanos , Macrófagos/citologia , Macrófagos/fisiologia , Camundongos , Placenta/citologia , Gravidez , Proteínas Proto-Oncogênicas c-sis/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Trofoblastos/citologia
18.
Leukemia ; 26(4): 728-35, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22015771

RESUMO

Constitutive tyrosine kinase activation by reciprocal chromosomal translocation is a common pathogenetic mechanism in chronic myeloproliferative disorders. Since centrosomal proteins have been recurrently identified as translocation partners of tyrosine kinases FGFR1, JAK2, PDGFRα and PDGFRß in these diseases, a role for the centrosome in oncogenic transformation has been hypothesized. In this study, we addressed the functional role of centrosomally targeted tyrosine kinase activity. First, centrosomal localization was not routinely found for all chimeric fusion proteins tested. Second, targeting of tyrosine kinases to the centrosome by creating artificial chimeric fusion kinases with the centrosomal targeting domain of AKAP450 failed to enhance the oncogenic transforming potential in both Ba/F3 and U2OS cells, although phospho-tyrosine-mediated signal transduction pathways were initiated at the centrosome. We conclude that the centrosomal localization of constitutively activated tyrosine kinases does not contribute to disease pathogenesis in chronic myeloproliferative disorders.


Assuntos
Transformação Celular Neoplásica , Centrossomo/fisiologia , Transtornos Mieloproliferativos/complicações , Proteínas Tirosina Quinases/fisiologia , Linhagem Celular Tumoral , Doença Crônica , Humanos , Fosforilação , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Tirosina/metabolismo
20.
Clin Hemorheol Microcirc ; 48(1): 5-13, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21876230

RESUMO

Recently, it was shown that mesenchymal stem cells (MSCs) are capable of differentiating into endothelial cells which highlights the potential role of MSCs in neovascularization. In the present study, we investigated the paracrine factors responsible for tube formation in human adipose-tissue derived stem cells (ASCs). Moreover, we analyzed ASC's migration towards PDGF-BB and altered levels of proteins involved in different pathways. Freshly isolated human adipose tissue-derived stem cells were seeded onto wells coated with Matrigel and cultured in endothelial growth medium. Capillary-like tube formation was observed after 18 hours culture. Tube formation was significantly reduced in the presence of antibodies against platelet-derived growth factor receptor beta (PDGF) or basic fibroblast growth factor (bFGF). Reverse phase proteomic assay (RPPA) was used to interrogate the expression of 139 phosphorylated or native proteins after incubation with PDGF-BB protein for 24 hours. The present data suggest, that freshly isolated ASCs contain a subpopuplation of stem cells that can form capillary like tubes which is dependent on PDGF and bFGF signaling pathway. Furthermore, Migration of human ASCs significantly increased in response to increased concentrations of PDGF-BB. In addition, incubation of ASCs with PDGF-BB altered phosphorylation of several transcription proteins that are widely expressed throughout the hematopoietic system, targeting genes that have been associated with proliferation, anti-apoptosis or differentiation.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Mesenquimais/citologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Tecido Adiposo/metabolismo , Becaplermina , Diferenciação Celular/fisiologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Neovascularização Fisiológica/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-sis/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Fatores de Transcrição STAT/metabolismo , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...