Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Breast Cancer Res Treat ; 195(1): 17-31, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35793004

RESUMO

PURPOSE: Platelet-derived growth factor B (PDGFB) is known to play essential roles in angiogenesis and lymphangiogenesis during development, and tumor growth and vessel stabilization in experimental models. However, whether these findings could be translated to breast cancer patients remains unclear. We hypothesized that PDGFB gene expression is associated with angiogenesis, cell proliferation, and clinical outcomes in breast cancer patients. METHODS: A total of 7635 primary breast cancer patients with full transcriptome and clinical data available from 13 independent cohorts were analyzed using in silico approach. The median value was used to divide each cohort into high and low PDGFB expression groups. RESULTS: High PDGFB gene expression was associated with increased expression of angiogenesis-related genes, higher amount of vascular cell infiltrations, and with enrichment of angiogenesis gene set, lymphangiogenesis-related gene expressions, lymphangiogenesis-related cell infiltrations, and enrichmentof lymphangiogenesis gene set in GSE96058 and validated by TCGA cohorts; however, not with lymphatic metastasis. PDGFB expression was neither associated with cell proliferation as assessed by Ki67 expression nor with Nottingham histological grade, or response to neoadjuvant chemotherapy. We found that PDGFB was most extensively expressed by endothelial and perivascular-like cells in the tumor microenvironment, and minimally by cancer cells consistently in two single-cell sequence cohorts. High PDGFB expression enriched TGFß, epithelial-mesenchymal transition, hypoxia, and cancer stem cell-associated pathways. However, no association with distant metastasis was observed. Disease-specific and disease-free survival were worse in the high PDGFB expression group consistently in TCGA and METABRIC cohorts. CONCLUSION: PDGFB is predominantly expressed in endothelial cells and is associated with angiogenesis and lymphangiogenesis, but not with cellular proliferation or metastasis in breast cancer.


Assuntos
Neoplasias da Mama , Linfangiogênese , Neoplasias da Mama/patologia , Células Endoteliais/metabolismo , Feminino , Genes sis , Humanos , Linfangiogênese/genética , Neovascularização Patológica/genética , Proteínas Proto-Oncogênicas c-sis/genética , Microambiente Tumoral
2.
Genes Chromosomes Cancer ; 60(3): 147-159, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33099837

RESUMO

The landscape of uterine sarcomas has greatly expanded in recent years to include neoplasms with recurrent gene fusions, such as BCOR and YWHAE translocated high-grade endometrial stromal sarcomas. Sophisticated molecular techniques have also resulted in the description of "new" entities associated with recurrent kinase fusions involving NTRK and RET as well as COL1A1-PDGFB rearranged uterine sarcomas. These rare neoplasms will be discussed in this review, highlighting that some of the underlying molecular events are clinically actionable and potentially susceptible to targeted therapy. While relatively few of these neoplasms have been described to date, likely being previously lumped under the spectrum of undifferentiated uterine sarcoma, the number of cases will expand in the future given their recognition and the increasing availability of molecular testing. These neoplasms have overlapping morphology (often with a "fibrosarcoma-like" appearance) and immunohistochemical features, and are characterized by variable clinical outcomes. Although immunohistochemistry may assist in some cases, a definitive subclassification requires confirmatory molecular studies. As these molecular assays may not be routinely available in most laboratories, referral to reference centers may be needed. In order to assist the pathologist, we suggest a diagnostic algorithm for routine practice when dealing with a malignant or potentially malignant uterine spindle cell neoplasm.


Assuntos
Receptor trkA/metabolismo , Sarcoma/metabolismo , Neoplasias Uterinas/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Cadeia alfa 1 do Colágeno Tipo I/genética , Feminino , Fusão Gênica/genética , Rearranjo Gênico , Genes sis , Humanos , Imuno-Histoquímica , Recidiva Local de Neoplasia/enzimologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptor trkA/genética , Sarcoma/enzimologia , Sarcoma/genética , Sarcoma do Estroma Endometrial/enzimologia , Sarcoma do Estroma Endometrial/genética , Sarcoma do Estroma Endometrial/metabolismo , Neoplasias de Tecidos Moles/genética
3.
Mov Disord ; 36(11): 2468-2480, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34432325

RESUMO

This systematic MDSGene review covers individuals with confirmed genetic forms of primary familial brain calcification (PFBC) available in the literature. Data on 516 (47% men) individuals, carrying heterozygous variants in SLC20A2 (solute carrier family 20 member 2, 61%), PDGFB (platelet-derived growth factor subunit B, 12%), XPR1 (xenotropic and polytropic retrovirus receptor, 16%), or PDGFRB (platelet-derived growth factor receptor beta, 5%) or biallelic variants in MYORG (myogenesis-regulating glycosidase, 13%) or JAM2 (junctional adhesion molecule 2, 2%), were extracted from 93 articles. Nearly one-third of the mutation carriers were clinically unaffected. Carriers of PDGFRB variants were more likely to be clinically unaffected (~54%), and the penetrance of SLC20A2 and XPR1 variants (<70%) was lower in comparison to the remaining three genes (>85%). Among the 349 clinically affected patients, 27% showed only motor and 31% only nonmotor symptoms/signs, whereas the remaining 42% had a combination thereof. While parkinsonism and speech disturbance were the most frequently reported motor manifestations, cognitive deficits, headache, and depression were the major nonmotor symptoms/signs. The basal ganglia were always calcified, and the cerebellum, thalamus, and white matter contained calcifications in 58%, 53%, and 43%, respectively, of individuals. In autosomal-dominant PFBC, mutation severity influenced the number of calcified brain areas, which in turn correlated with the clinical status, whereby the risk of developing symptoms/signs more than doubled for each additional region with calcifications. Our systematic analysis provides the most comprehensive insight into genetic, clinical, and neuroimaging features of known PFBC forms, to date. In addition, it puts forth the penetrance estimates and newly discovered genotype-phenotype relations that will improve counseling of individuals with mutations in PFBC genes. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Encefalopatias , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encefalopatias/genética , Genes sis , Heterozigoto , Humanos , Mutação , Fenótipo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética
4.
BMC Musculoskelet Disord ; 22(1): 710, 2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34407802

RESUMO

BACKGROUND: There is variability in individual response to platelet-rich plasma (PRP) therapy in tennis elbow treatment. Genetic variation, especially within genes encoding growth factors may influence the observed inter-individual differences. The purpose of this study was to identify polymorphic variants of the platelet-derived growth factor beta polypeptide gene (PDGFB) that determine an improved individual response to PRP therapy in tennis elbow patients. METHODS: This prospective cohort study was designed in accordance with STROBE and MIBO guidelines. A cohort of 107 patients (132 elbows, 25 bilateral) was studied, including 65 females (77 elbows) and 42 males (55 elbows), aged 24-64 years (median 46.00 ± 5.50), with lateral elbow tendinopathy treated with autologous PRP injection. The effectiveness of PRP therapy was recorded in all subjects at 2, 4, 8, 12, 24 and 52 weeks after PRP injection using the Visual Analog Scale (VAS), quick version of Disabilities of the Arm, Shoulder and Hand score (QDASH) and Patient-Rated Tennis Elbow Evaluation (PRTEE). In order to determine the PDGFB variants with the best response to PRP therapy, patient reported outcome measures were compared between individual genotypes within studied polymorphic variants (rs2285099, rs2285097, rs2247128, rs5757572, rs1800817 and rs7289325). The influence of single nucleotide polymorphisms on blood and PRP parameters, including the concentration of PDGF-AB and PDGF-BB proteins was also analyzed. RESULTS: Our analysis identified genetic variants of the PDGFB gene that lead to a better response to PRP therapy. The TT (rs2285099) and CC (rs2285097) homozygotes had higher concentration of platelets in whole blood than carriers of other genotypes (p = 0.018) and showed significantly (p < 0.05) lower values of VAS (weeks 2-12), QDASH and PRTEE (weeks 2-24). The rs2285099 and rs2285097 variants formed strong haplotype block (r2 = 98, D'=100). The AA homozygotes (rs2247128) had significantly lower values of VAS (weeks 4-52), QDASH and PRTEE (weeks 8, 12). CONCLUSIONS: PDGFB gene's polymorphisms increase the effectiveness of PRP therapy in tennis elbow treatment. Genotyping two polymorphisms of the PDGFB gene, namely rs2285099 (or rs2285097) and rs2247128 may be a helpful diagnostic tool while assessing patients for PRP therapy and modifying the therapy to improve its effectiveness.


Assuntos
Genes sis , Plasma Rico em Plaquetas , Tendinopatia , Cotovelo de Tenista , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Cotovelo de Tenista/diagnóstico , Cotovelo de Tenista/genética , Cotovelo de Tenista/terapia
5.
Hum Mutat ; 40(4): 392-403, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30609140

RESUMO

Primary familial brain calcification (PFBC) is a rare neurodegenerative disorder with four causative genes (SLC20A2, PDGFRB, PDGFB, and XPR1) that have been identified. Here, we aim to describe the mutational spectrum of four causative genes in a series of 226 unrelated Chinese PFBC patients. Mutations in four causative genes were detected in 16.8% (38/226) of PFBC patients. SLC20A2 mutations accounted for 14.2% (32/226) of all patients. Mutations in the other three genes were relatively rare, accounting for 0.9% (2/226) of all patients, respectively. Clinically, 44.8% of genetically confirmed patients (probands and relatives) were considered symptomatic. The most frequent symptoms were chronic headache, followed by movement disorders and vertigo. Moreover, the total calcification score was significantly higher in the symptomatic group compared to the asymptomatic group. Functionally, we observed impaired phosphate transport induced by seven novel missense mutations in SLC20A2 and two novel mutations in XPR1. The mutation p.D164Y in XPR1 might result in low protein expression through an enhanced proteasome pathway. In conclusion, our study further confirms that mutations in SLC20A2 are the major cause of PFBC and provides additional evidence for the crucial roles of phosphate transport impairment in the pathogenies of PFBC.


Assuntos
Encefalopatias/genética , Calcinose/genética , Predisposição Genética para Doença , Mutação , Doenças Neurodegenerativas/genética , Adulto , Idoso , Alelos , Transporte Biológico , Biomarcadores , Encefalopatias/diagnóstico , Encefalopatias/metabolismo , Calcinose/diagnóstico , Calcinose/metabolismo , Linhagem Celular Tumoral , China , Feminino , Genes sis , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/metabolismo , Neuroimagem , Fenótipo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptores Acoplados a Proteínas G/genética , Receptores Virais/genética , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Tomografia Computadorizada por Raios X , Receptor do Retrovírus Politrópico e Xenotrópico
6.
J Biol Chem ; 289(34): 23712-22, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-24973215

RESUMO

The presence of the blood-brain barrier (BBB) is critical for cholesterol metabolism in the brain, preventing uptake of lipoprotein-bound cholesterol from the circulation. The metabolic consequences of a leaking BBB for cholesterol metabolism have not been studied previously. Here we used a pericyte-deficient mouse model, Pdgfb(ret/ret), shown to have increased permeability of the BBB to a range of low-molecular mass and high-molecular mass tracers. There was a significant accumulation of plant sterols in the brains of the Pdgfb(ret/ret) mice. By dietary treatment with 0.3% deuterium-labeled cholesterol, we could demonstrate a significant flux of cholesterol from the circulation into the brains of the mutant mice roughly corresponding to about half of the measured turnover of cholesterol in the brain. We expected the cholesterol flux into the brain to cause a down-regulation of cholesterol synthesis. Instead, cholesterol synthesis was increased by about 60%. The levels of 24(S)-hydroxycholesterol (24S-OHC) were significantly reduced in the brains of the pericyte-deficient mice but increased in the circulation. After treatment with 1% cholesterol in diet, the difference in cholesterol synthesis between mutants and controls disappeared. The findings are consistent with increased leakage of 24S-OHC from the brain into the circulation in the pericyte-deficient mice. This oxysterol is an efficient suppressor of cholesterol synthesis, and the results are consistent with a regulatory role of 24S-OHC in the brain. To our knowledge, this is the first demonstration that a defective BBB may lead to increased flux of a lipophilic compound out from the brain. The relevance of the findings for the human situation is discussed.


Assuntos
Barreira Hematoencefálica , Encéfalo/metabolismo , Colesterol/metabolismo , Homeostase , Animais , Sequência de Bases , Colesterol/biossíntese , Primers do DNA , Genes sis , Homeostase/genética , Camundongos , Camundongos Transgênicos , Plantas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Esteróis/metabolismo
7.
Proc Natl Acad Sci U S A ; 109(9): 3475-80, 2012 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22345562

RESUMO

Insulin-like growth factor-binding protein 2 (IGFBP2) is increasingly recognized as a glioma oncogene, emerging as a target for therapeutic intervention. In this study, we used an integrative approach to characterizing the IGFBP2 network, combining transcriptional profiling of human glioma with validation in glial cells and the replication-competent ASLV long terminal repeat with a splice acceptor/tv-a glioma mouse system. We demonstrated that IGFBP2 expression is closely linked to genes in the integrin and integrin-linked kinase (ILK) pathways and that these genes are associated with prognosis. We further showed that IGFBP2 activates integrin ß1 and downstream invasion pathways, requires ILK to induce cell motility, and activates NF-κB. Most significantly, the IGFBP2/integrin/ILK/NF-κB network functions as a physiologically active signaling pathway in vivo by driving glioma progression; interfering with any point in the pathway markedly inhibits progression. The results of this study reveal a signaling pathway that is both targetable and highly relevant to improving the survival of glioma patients.


Assuntos
Neoplasias Encefálicas/patologia , Terapia Genética , Vetores Genéticos/uso terapêutico , Glioblastoma/patologia , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/fisiologia , Integrina beta1/fisiologia , NF-kappa B/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Astrocitoma/genética , Astrocitoma/metabolismo , Proteínas Aviárias/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Genes Sintéticos , Genes sis , Vetores Genéticos/administração & dosagem , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/toxicidade , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/toxicidade , Proteínas de Filamentos Intermediários/genética , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Inibidor de NF-kappaB alfa , Invasividade Neoplásica , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Nestina , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Prognóstico , Proteínas Serina-Treonina Quinases/toxicidade , Receptores Virais/genética , Retroviridae , Transdução de Sinais/fisiologia
8.
Biochem Biophys Res Commun ; 434(2): 305-10, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23537647

RESUMO

We have reported that the oncoprotein hepatitis B virus X-interacting protein (HBXIP) acts as a novel transcriptional coactivator to promote proliferation and migration of breast cancer cells. Previously, we showed that HBXIP was able to activate nuclear factor-κB (NF-κB) in breast cancer cells. As an oncogene, the platelet-derived growth factor beta polypeptide (PDGFB) plays crucial roles in carcinogenesis. In the present study, we found that both HBXIP and PDGFB were highly expressed in breast cancer cell lines. Interestingly, HBXIP was able to increase transcriptional activity of NF-κB through PDGFB, suggesting that HBXIP is associated with PDGFB in the cells. Moreover, HBXIP was able to upregulate PDGFB at the levels of mRNA, protein and promoter in the cells. Then, we identified that HBXIP stimulated the promoter of PDGFB through activating transcription factor Sp1. In function, HBXIP enhanced the proliferation of breast cancer cells through PDGFB in vitro. Thus, we conclude that HBXIP upregulates PDGFB via activating transcription factor Sp1 to promote proliferation of breast cancer cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Genes sis , Fator de Transcrição Sp1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias da Mama/metabolismo , Imunoprecipitação da Cromatina , Clonagem Molecular , Feminino , Genes Reporter , Humanos , Luciferases/genética , Luciferases/metabolismo , Células MCF-7 , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fator de Transcrição Sp1/genética , Ativação Transcricional , Regulação para Cima
9.
Breast Cancer Res Treat ; 140(3): 587-601, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23912956

RESUMO

Growth factors (GF) stimulate cell proliferation through binding to cell membrane receptors and are thought to be involved in cancer risk and survival. We examined how genetic variation in epidermal growth factor (EGF), neuregulin 2 (NRG2), ERBB2 (HER2/neu), fibroblast growth factors 1 and 2 (FGF1 and FGF2) and its receptor 2 (FGFR2), and platelet-derived growth factor B (PDGFB) independently and collectively influence breast cancer risk and survival. We analyzed data from the Breast Cancer Health Disparities Study which includes Hispanic (2,111 cases, 2,597 controls) and non-Hispanic white (1,481 cases, 1,586 controls) women. Adaptive rank-truncated product (ARTP) analysis was conducted to determine gene significance. Odds ratios (OR) and 95 % confidence intervals were obtained from conditional logistic regression models to estimate breast cancer risk and Cox proportional hazard models were used to estimate hazard ratios (HR) of dying from breast cancer. We assessed Native American (NA) ancestry using 104 ancestry informative markers. We observed few significant associations with breast cancer risk overall or by menopausal status other than for FGFR2 rs2981582. This SNP was significantly associated with ER+/PR+ (OR 1.66, 95 % CI 1.37-2.00) and ER+/PR- (OR 1.54, 95 % CI 1.03-2.31) tumors. Multiple SNPs in FGF1, FGF2, and NRG2 significantly interacted with multiple SNPs in EGFR, ERBB2, FGFR2, and PDGFB, suggesting that breast cancer risk is dependent on the collective effects of genetic variants in other GFs. Both FGF1 and ERBB2 significantly influenced overall survival, especially among women with low levels of NA ancestry (P ARTP = 0.007 and 0.003, respectively). Our findings suggest that genetic variants in growth factors signaling appear to influence breast cancer risk through their combined effects. Genetic variation in ERBB2 and FGF1 appear to be associated with survival after diagnosis with breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Peptídeos e Proteínas de Sinalização Intercelular/genética , Adulto , Idoso , Estudos de Casos e Controles , Fator de Crescimento Epidérmico/genética , Feminino , Fator 1 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/genética , Genes erbB-2 , Genes sis , Hispânico ou Latino/genética , Humanos , Indígenas Norte-Americanos/genética , Pessoa de Meia-Idade , Fatores de Crescimento Neural/genética , Polimorfismo de Nucleotídeo Único , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , População Branca/genética
11.
Glia ; 58(9): 1050-65, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20468047

RESUMO

Glial progenitors in the white matter and the subventricular zone are the major population of cycling cells in the postnatal central nervous system, and thought to be candidates for glioma-initiating cells. However, less is known about the dividing cell populations in the brainstem than those in the cerebrum, leading to the lag of basic understanding of brainstem gliomas. We herein demonstrate much fewer cycling glial progenitors exist in the brainstem than in the cerebrum. We also show that infecting brainstem glial progenitors with PDGFB-green fluorescent protein (GFP)-expressing retrovirus induced tumors that closely resembled human malignant gliomas. Of note, brainstem tumors grew more slowly than cerebral tumors induced by the same retrovirus, and >80% tumor cells in the brainstem consisted of GFP-positive, infected progenitors while GFP-positive cells in the cerebral tumors were <20%. These indicate that cerebral tumors progressed rapidly by recruiting resident progenitors via paracrine mechanism whereas brainstem tumors grew more slowly by clonal expansion of the infected population. The cerebral and brainstem glial progenitors similarly showed reversible dedifferentiation upon PDGF stimulation in vitro and did not show the intrinsic difference in terms of the responsiveness to PDGF. We therefore suggest that slower, monoclonal progression pattern of the brainstem tumors is at least partly due to the environmental factors including the cell density of the glial progenitors. Together, these findings are the first implications regarding the cell-of-origin and the gliomagenesis in the brainstem.


Assuntos
Neoplasias do Tronco Encefálico/fisiopatologia , Tronco Encefálico/fisiopatologia , Glioma/fisiopatologia , Neuroglia/fisiologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células-Tronco/fisiologia , Animais , Neoplasias Encefálicas/fisiopatologia , Tronco Encefálico/patologia , Neoplasias do Tronco Encefálico/patologia , Diferenciação Celular , Células Cultivadas , Cerebelo/fisiologia , Criança , Feminino , Genes sis , Vetores Genéticos , Glioma/patologia , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Fator de Crescimento Derivado de Plaquetas/genética , Ratos , Retroviridae
12.
Histopathology ; 54(7): 860-72, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19635106

RESUMO

AIMS: To analyse the presence of collagen type I alpha 1-platelet-derived growth factor beta (COL1A1-PDGFB) transcripts in 20 cases of dermatofibrosarcoma protuberans (DFSP) and to assess the relationship between COL1A1 breakpoints and clinical and histopathological variables. METHODS AND RESULTS: Multiplex reverse transcriptase-polymerase chain reaction was carried out using frozen tissue. Our series contained 14 men and six women. Histologically, most cases were of conventional type (n = 9), followed by fibrosarcoma (n = 4), Bednar tumour (n = 2), sclerosing (n = 2), myoid (n = 1) and atrophic (n = 1) DFSP, and giant cell fibroblastoma (n = 1). Immunohistochemistry revealed CD34 expression in 90% of cases. COL1A1-PDGFB fusion transcripts were present in 89% of cases (exons 18, 19, 20, 25, 26, 31, 33/34, 39, 40, 46, 47 and 48 of COL1A1 with exon 2 of PDGFB). There was no recurrence of DFSP in any of the 19 patients treated by Mohs surgery. A partial response was obtained in the two patients treated with imatinib. CONCLUSIONS: The COL1A1-PDGFB fusion was present in all histological subtypes of DFSP, but not all cases expressed the fusion transcript. No association was observed between different COL1A1 breakpoints and clinicopathological parameters. Imatinib mesylate can be useful in locally advanced tumours and metastases.


Assuntos
Colágeno Tipo I/genética , Dermatofibrossarcoma/genética , Dermatofibrossarcoma/patologia , Genes sis , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Sequência de Bases , Benzamidas , Cadeia alfa 1 do Colágeno Tipo I , Primers do DNA/genética , Dermatofibrossarcoma/terapia , Feminino , Fusão Gênica , Humanos , Mesilato de Imatinib , Masculino , Pessoa de Meia-Idade , Cirurgia de Mohs , Piperazinas/uso terapêutico , Pirimidinas/uso terapêutico , Neoplasias Cutâneas/terapia , Adulto Jovem
13.
PLoS Biol ; 4(8): e260, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16869712

RESUMO

Mammalian hearts cannot regenerate. In contrast, zebrafish hearts regenerate even when up to 20% of the ventricle is amputated. The mechanism of zebrafish heart regeneration is not understood. To systematically characterize this process at the molecular level, we generated transcriptional profiles of zebrafish cardiac regeneration by microarray analyses. Distinct gene clusters were identified based on temporal expression patterns. Genes coding for wound response/inflammatory factors, secreted molecules, and matrix metalloproteinases are expressed in regenerating heart in sequential patterns. Comparisons of gene expression profiles between heart and fin regeneration revealed a set of regeneration core molecules as well as tissue-specific factors. The expression patterns of several secreted molecules around the wound suggest that they play important roles in heart regeneration. We found that both platelet-derived growth factor-a and -b (pdgf-a and pdgf-b) are upregulated in regenerating zebrafish hearts. PDGF-B homodimers induce DNA synthesis in adult zebrafish cardiomyocytes. In addition, we demonstrate that a chemical inhibitor of PDGF receptor decreases DNA synthesis of cardiomyocytes both in vitro and in vivo during regeneration. Our data indicate that zebrafish heart regeneration is associated with sequentially upregulated wound healing genes and growth factors and suggest that PDGF signaling is required.


Assuntos
Coração/fisiologia , Regeneração/genética , Peixe-Zebra/genética , Peixe-Zebra/fisiologia , Animais , Becaplermina , Proliferação de Células , Perfilação da Expressão Gênica , Genes sis , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metaloproteinases da Matriz/metabolismo , Dados de Sequência Molecular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis , Transdução de Sinais , Regulação para Cima , Proteínas de Peixe-Zebra/metabolismo
14.
Nephrol Dial Transplant ; 24(2): 448-57, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18775895

RESUMO

BACKGROUND: Morphological changes associated with long-term peritoneal dialysis (PD) include increased vascular surface area due to angiogenesis, submesothelial fibrosis and epithelial mesenchymal transition. Platelet-derived growth factor (PDGF) has been associated with all of these phenomena, and is a prototypical 'response to injury' growth factor. METHODS: Rats received an intraperitoneal injection of adenoviral vector expressing PDGF-B. At sacrifice, we analysed the structure and function of the peritoneal membrane. Gene expression in the peritoneal tissue was assessed for changes suggestive of epithelial mesenchymal transition. RESULTS: Over-expression of PDGF in the rat peritoneum led to significant angiogenesis, cellular proliferation and submesothelial thickening. Although PDGF induced expression of transforming growth factor beta, there was a lack of activation of this growth factor, and we believe that this explains the lack of significant collagen accumulation observed by a hydroxyproline assay. Despite evidence of angiogenesis and subsequent increased solute transport, we observed only a transient, non-significant impact on ultrafiltration function. This suggests that increased vascular surface area is necessary, but not sufficient, to produce ultrafiltration dysfunction. There was no evidence of epithelial mesenchymal transition observed either in regulation of associated genes such as Snail or E-Cadherin or in the lack of dual-labelled epithelial and mesenchymal cells on immunofluorescence. Mesothelial cells exposed to PDGF-B demonstrated increased collagen gene expression. CONCLUSIONS: PDGF-B induced angiogenesis without fibrosis in the peritoneum. The lack of significant ultrafiltration dysfunction and epithelial mesenchymal transition, as observed in patients on PD, suggests that PDGF-B may play a role, but is not the integral component, in response to peritoneal injury.


Assuntos
Peritônio/patologia , Peritônio/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Adenoviridae/genética , Animais , Colágeno/genética , Colágeno Tipo I , Epitélio/patologia , Expressão Gênica , Genes sis , Vetores Genéticos , Humanos , Mesoderma/patologia , Neovascularização Patológica , Diálise Peritoneal/efeitos adversos , Peritônio/irrigação sanguínea , Inibidor 1 de Ativador de Plasminogênio/genética , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-sis/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
15.
Genesis ; 46(2): 74-80, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18257043

RESUMO

In recent years, gene-targeting studies in mice have elucidated many molecular mechanisms in vascular biology. However, it has been difficult to apply this approach to the study of postnatal animals because mutations affecting the vasculature are often embryonically lethal. We have therefore generated transgenic mice that express a tamoxifen-inducible form of Cre recombinase (iCreER(T2)) in vascular endothelial cells using a phage artificial chromosome (PAC) containing the Pdgfb gene (Pdgfb-iCreER mice). This allows the genetic targeting of the vascular endothelium in postnatal animals. We tested efficiency of tamoxifen-induced iCre recombinase activity with ROSA26-lacZ reporter mice and found that in newborn animals recombination could be achieved in most capillary and small vessel endothelial cells in most organs including the central nervous system. In adult animals, recombination activity was also widespread in capillary beds of skeletal muscle, heart, skin, and gut but not in the central nervous system where only a subpopulation of endothelial cells was labeled. We also tested recombination efficiency in a subcutaneous tumor model and found recombination activity in all detectable tumor blood vessels. Thus, Pdgfb-iCreER mice are a valuable research tool to manipulate endothelial cells in postnatal mice and study tumor angiogenesis.


Assuntos
Endotélio Vascular/metabolismo , Técnicas Genéticas , Integrases/genética , Animais , Embrião de Mamíferos , Endotélio Vascular/citologia , Regulação da Expressão Gênica no Desenvolvimento , Genes sis , Camundongos , Camundongos Transgênicos , Neovascularização Patológica , Tamoxifeno/metabolismo
16.
Medicine (Baltimore) ; 97(42): e12918, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30335026

RESUMO

Idiopathic basal ganglia calcification (IBGC) is characterized by brain calcification and a wide variety of neurological and psychiatric symptoms. In families displaying an autosomal dominant inheritance pattern, three causative genes have been identified: SLC20A2, PDGFRB, and very recently, PDGFB. While in clinical practice sporadic presentation of IBGC is frequent, well-documented reports of true sporadic occurrences are rare. We report a case of a 61-year-old woman who presented with depressive and dystonic symptoms revealing IBGC. Her 41-year-old daughter was healthy. In the proband, we identified 4 mutations in PDGFB, and 1 exonic mutation in SLC20A2, all of which were absent in the daughter. These mutations may result in a loss-of-function of PDGF-B or SLC20A2, which has been shown to cause IBGC in humans and disrupts the blood-brain barrier in mice resulting in brain calcification. Herein, we present the occurrence of a sporadic patient of IBGC and its causative mutations.


Assuntos
Doenças dos Gânglios da Base/genética , Calcinose/genética , Adulto , Animais , Éxons/genética , Feminino , Genes sis/genética , Humanos , Camundongos , Pessoa de Meia-Idade , Mutação/genética , Núcleo Familiar , Linhagem , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , República da Coreia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/genética , Sequenciamento do Exoma
17.
Clin Nephrol ; 67(2): 65-72, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17338425

RESUMO

BACKGROUND: Few genetic factors have been identified that determine susceptibility to and progression of IgA-nephropathy (IgAN). Given that IgAN is usually characterized by mesangioproliferative glomerulonephritis and that PDGF-B is of central pathophysiological relevance in this process, we analyzed four single-nucleotide polymorphisms (SNPs) of the PDGF-B gene to evaluate a possible association of these SNPs with disease onset and progression, histological grading and responses to ACE inhibitor (ACEi) therapy. METHODS: The total study population consisted of 195 IgAN patients (127 from southern Italy and 68 from northern Germany) and 200 healthy controls (100 from each region). All four SNPs were in Hardy-Weinberg equilibrium and genotype distributions did not differ between patients and controls in either region. RESULTS: SNP distribution in Italian patients reaching end-stage renal disease (n=45) also was not significantly different from patients maintaining a serum creatinine below 1.2 mg/dl (n=60) during 5.6 +/- 5.5 years of follow-up. Furthermore, we failed to detect significant effects of any SNP on the slope of 1/serum creatinine, proteinuria level or the antiproteinuric response to ACEi. Additionally, particular PDGF-B genotypes did not correlate with histological grading using the Lee classification. CONCLUSION: We conclude that none of the four PDGF-B SNPs is related to the onset of IgAN in two different populations and that none of them has a major influence on the course of IgAN.


Assuntos
Genes sis , Glomerulonefrite por IGA/genética , Polimorfismo de Nucleotídeo Único , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Biópsia , Progressão da Doença , Alemanha , Glomerulonefrite por IGA/tratamento farmacológico , Glomerulonefrite por IGA/etnologia , Glomerulonefrite por IGA/patologia , Humanos , Itália , Fator de Crescimento Derivado de Plaquetas/genética , Reação em Cadeia da Polimerase , Valor Preditivo dos Testes , Estudos Retrospectivos , Índice de Gravidade de Doença
18.
J Clin Oncol ; 23(4): 866-73, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15681532

RESUMO

PURPOSE: The cutaneous malignant tumor dermatofibrosarcoma protuberans (DFSP) is typically associated with a translocation between chromosomes 17 and 22 that places the platelet-derived growth factor-B (PDGFB) under the control of the collagen 1A1 promoter. The purpose of this study was to evaluate molecular, cytogenetic, and kinase activation profiles in a series of DFSPs and to determine whether these biologic parameters are correlated with the clinical responses of DFSP to imatinib. PATIENTS AND METHODS: We analyzed the objective radiologic and clinical response to imatinib at 400 mg twice daily in eight patients with locally advanced DFSP and two patients with metastatic disease. RESULTS: Each of eight patients with locally advanced DFSP had evidence of t(17;22) and showed a clinical response to imatinib. Four of these patients had complete clinical responses. The two patients with metastatic disease had fibrosarcomatous histology and karyotypes that were substantially more complex than those typically associated with localized DFSP. One patient with metastatic DFSP and an associated t(17;22) had a partial response to imatinib but experienced disease progression after 7 months of therapy. In contrast, the other patient with metastatic disease had a tumor lacking t(17;22), and there was no clinical response to imatinib. Unexpectedly, there was minimal platelet-derived growth factor receptor-beta phosphorylation in the untreated DFSP, despite the documented presence of a PDGFB autocrine mechanism. CONCLUSION: Imatinib has clinical activity against both localized and metastatic DFSP with t(17;22). However, fibrosarcomatous variants of DFSP lacking t(17;22) may not respond to imatinib.


Assuntos
Antineoplásicos/uso terapêutico , Dermatofibrossarcoma/tratamento farmacológico , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Adulto , Idoso , Benzamidas , Aberrações Cromossômicas , Dermatofibrossarcoma/genética , Dermatofibrossarcoma/mortalidade , Intervalo Livre de Doença , Feminino , Rearranjo Gênico , Genes sis , Humanos , Mesilato de Imatinib , Masculino , Pessoa de Meia-Idade , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/mortalidade
19.
Oncogene ; 23(28): 4885-93, 2004 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-15077165

RESUMO

We showed previously that exogenous overexpression of C3G, a guanine nucleotide releasing factor (GEF) for Rap1 and R-Ras proteins, blocks the focus-forming activity of cotransfected, activated, sis, ras and v-raf oncogenes in NIH 3T3 cells. In this report, we show that C3G also interferes with dbl and R-Ras focus-forming activity and demonstrate that the transformation suppressor ability of C3G maps to its Crk-binding region (SH3-b domain). Using full-length C3G and C3GDeltaCat mutant, lacking catalytic domain, we showed here that overexpression of cotransfected C3G or C3GDeltaCat inhibited oncogenic Hraslys12-mediated phosphorylation of ERK, without altering Ras and Raf-1 kinase activation. We also showed that, overexpressed C3G and C3GdeltaCat inhibited the viability of oncogenic Ras-induced colonies in soft agar, indicating that C3G interferes with the anchorage-independent growth of Ras-transformed cells in a Rap1-independent manner. Consistent with both observations, overexpression of exogenous C3G and C3GDeltaCat also caused downregulation of Ras-induced cyclin A expression. Altogether, our results indicate that C3G interferes with at least two separate aspects of oncogenic transformation - cell cycle progression and loss of contact inhibition - and that these inhibitory effects probably account for its transformation suppressor activity.


Assuntos
Ciclina A/genética , Fator 2 de Liberação do Nucleotídeo Guanina/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Oncogênicas v-raf/genética , Células 3T3 , Animais , Sequência de Bases , Adesão Celular , Divisão Celular , Clonagem Molecular , Primers do DNA , Fibroblastos/fisiologia , Genes ras , Genes sis , Humanos , Camundongos , Reação em Cadeia da Polimerase , Mapeamento por Restrição , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA