Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Breast Cancer Res Treat ; 201(2): 317-328, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37378696

RESUMO

PURPOSE: HER2-positive breast cancer has a high chance of achieving pathological complete response when HSD17B4, responsible for peroxisomal ß-oxidation of very long-chain fatty acids (VLCFA) and estradiol, is methylation-silenced. Here, we aimed to identify the underlying molecular mechanism. METHODS: Using a HER2-positive breast cancer cell line, BT-474, control and knock-out (KO) clones were obtained. Metabolic characteristics were analyzed using a Seahorse Flux analyzer. RESULTS: HSD17B4 KO suppressed cellular proliferation, and enhanced sensitivity to lapatinib approximately tenfold. The KO led to accumulation of VLCFA and a decrease of polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and arachidonic acid. HSD17B4 KO increased Akt phosphorylation, possibly via decreased DHA, and genes involved in oxidative phosphorylation (OxPhos) and electron transport chain (ETC) were upregulated. Increased mitochondrial ATP production in the KO cells was confirmed by extracellular flux analyzer. Increased OxPhos led to severe dependence of the KO cells on pyruvate from glycolysis. Suppression of glycolysis by lapatinib led to severe delayed suppression of OxPhos in KO cells. CONCLUSION: HSD17B4 KO in BT-474 cells caused a decrease of PUFAs, increased Akt phosphorylation, enhanced glucose dependence of OxPhos, and increased sensitivity to inhibition of HER2, upstream of Akt. This mechanism may be applicable to other HER2-positive glucose-dependent breast cancer cells with HSD17B4 silencing.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Lapatinib/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Metilação , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Glucose , Linhagem Celular Tumoral , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Proteína Multifuncional do Peroxissomo-2/genética , Proteína Multifuncional do Peroxissomo-2/metabolismo
2.
Am J Med Genet A ; 188(1): 357-363, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34623748

RESUMO

D-bifunctional protein (DBP) deficiency is a rare, autosomal recessive peroxisomal enzyme deficiency resulting in a high burden of morbidity and early mortality. Patients with DBP deficiency resemble those with a severe Zellweger phenotype, with neonatal hypotonia, seizures, craniofacial dysmorphisms, psychomotor delay, deafness, blindness, and death typically within the first 2 years of life, although patients with residual enzyme function can survive longer. The clinical severity of the disease depends on the degree of enzyme deficiency. Loss-of-function variants typically result in no residual enzyme activity; however, splice variants may result in protein with residual function. We describe a full-term newborn presenting with hypotonia, seizures, and unexplained hypoglycemia, who was later found to have rickets at follow up. Rapid whole genome sequencing identified two HSD17B4 variants in trans; one likely pathogenic variant and one variant of uncertain significance (VUS) located in the polypyrimidine tract of intron 13. To determine the functional consequence of the VUS, we analyzed RNA from the patient's father with RNA-seq which showed skipping of Exon 14, resulting in a frameshift mutation three amino acids from the new reading frame. This RNA-seq analysis was correlated with virtually absent enzyme activity, elevated very-long-chain fatty acids in fibroblasts, and a clinically severe phenotype. Both variants are reclassified as pathogenic. Due to the clinical spectrum of DBP deficiency, this provides important prognostic information, including early mortality. Furthermore, we add persistent hypoglycemia to the clinical spectrum of the disease, and advocate for the early management of fat-soluble vitamin deficiencies to reduce complications.


Assuntos
Perda Auditiva Neurossensorial , Hipoglicemia , Deficiência de Proteína , Éxons , Perda Auditiva Neurossensorial/genética , Humanos , Hipoglicemia/genética , Recém-Nascido , Proteína Multifuncional do Peroxissomo-2/genética , Deficiência de Proteína/genética
3.
Int J Mol Sci ; 22(6)2021 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-33805726

RESUMO

An electromagnetic field (EMF) may affect the functions of uterine tissues. This study hypothesized that EMF changes the estrogenic activity of pig myometrium during the peri-implantation period. Tissue was collected on days 15-16 of the gestation and incubated in the presence of EMF (50 and 120 Hz, 2 and 4 h). The cytochrome P450 aromatase type 3 (CYP19A3) and hydroxysteroid 17ß dehydrogenase type 4 (HSD17B4) mRNA transcript abundance, cytochrome P450arom (aromatase), and 17ß hydroxysteroid dehydrogenase 17ßHSD) protein abundance and estrone (E1) and estradiol-17ß (E2) release were examined using Real-Time PCR, Western blot and radioimmunoassay. Selected myometrial slices were treated with progesterone (P4) to determine whether it functions as a protector against EMF. CYP19A3 mRNA transcript abundance in slices treated with EMF was less at 50 Hz (2 h) and greater at 120 Hz (2 and 4 h). HSD17B4 mRNA transcript was greater in slices treated with EMF at 120 Hz (2 h). Progesterone diminished EMF-related effects on CYP19A3 and HSD17B4. When P4 was added, EMF had suppressive (50 and 120 Hz, 2 h) or enhancing (50 Hz, 4 h) effects on aromatase abundance. The E1 release was lower after 4 h of EMF treatment at 50 Hz and P4 did not protect myometrial E1 release. In conclusion, EMF alters the synthesis and release of E1 and did not affect E2 release in the myometrium during the peri-implantation period.


Assuntos
Campos Eletromagnéticos/efeitos adversos , Implantação do Embrião/efeitos da radiação , Estradiol/metabolismo , Estrona/metabolismo , Regulação da Expressão Gênica/efeitos da radiação , Miométrio/efeitos da radiação , Animais , Aromatase/genética , Aromatase/metabolismo , Radiação Eletromagnética , Feminino , Miométrio/metabolismo , Proteína Multifuncional do Peroxissomo-2/genética , Proteína Multifuncional do Peroxissomo-2/metabolismo , Progesterona/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos , Técnicas de Cultura de Tecidos
4.
Cancer Res ; 81(13): 3593-3606, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33762355

RESUMO

Molecular mechanisms underlying intratumoral androgenesis and aberrant androgen receptor (AR) activation in prostate cancer remain poorly understood. Here we demonstrate that ectopic expression of the E3 ubiquitin ligase adaptor speckle-type poxvirus and zinc finger domain protein (SPOP) stabilizes 17ßHSD4. SPOP bound a functional substrate-binding consensus (SBC) motif 315RATST319 in 17ßHSD4 and promoted nondegradable K27- and K29-linked polyubiquitination of 17ßHSD4. The effect of SPOP was antagonized by serum- and glucocorticoid kinase-3 (SGK3)-mediated phosphorylation of serine 318 (S318) in the SBC and S318 phosphorylation-dependent binding of SKP2 E3 ligase and subsequent K48-linked polyubiquitination and proteasomal degradation of 17ßHSD4. Prostate cancer-associated SPOP mutations impaired the SPOP-17ßHSD4 interaction, caused 17ßHSD4 protein destruction in prostate cancer cells in culture and patient specimens, and increased testosterone production and prostate cancer cell growth in vitro and in mouse models. Thus, we have identified SPOP and SKP2 as two essential E3 ubiquitin ligases that exert opposite effects on 17ßHSD4 protein degradation and intratumoral androgenesis in prostate cancer cells. We further demonstrate that SPOP mutations or SKP2 overexpression contribute to prostate cancer progression by decreasing 17ßHSD4 expression and increasing intratumoral androgen synthesis. SIGNIFICANCE: This study reveals a novel mechanism of aberrant AR activation in SPOP-mutated prostate cancer and uncovers putative biomarkers for effective treatment by AR-targeted therapies.


Assuntos
Androgênios/metabolismo , Regulação Neoplásica da Expressão Gênica , Mutação , Proteínas Nucleares/metabolismo , Proteína Multifuncional do Peroxissomo-2/metabolismo , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Humanos , Masculino , Camundongos , Camundongos SCID , Proteínas Nucleares/genética , Proteína Multifuncional do Peroxissomo-2/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteólise , Receptores Androgênicos/genética , Proteínas Repressoras/genética , Células Tumorais Cultivadas , Ubiquitinação , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Sci Rep ; 10(1): 15530, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32968149

RESUMO

HER2-positive breast cancers that achieve pathological complete response (pCR) after HER2-directed therapy consistently have good survival. We previously identified HSD17B4 methylation as a marker for pCR by methylation screening. Here, we aimed to identify a new marker by conducting a multi-omics analysis of materials prepared by laser capture microdissection, and adding 71 new samples. In the screening set (n = 36), mutations, methylation, and expression were analyzed by targeted sequencing, Infinium 450 K, and expression microarray, respectively, and 15 genes were identified as differentially expressed and eight genomic regions as differentially methylated between cancer samples with and without pCR. In a validation set (n = 47), one gene showed differential expression, and one region had differential methylation. Further, in the re-validation set (n = 55), all new samples, only HSD17B4 methylation was significantly different. The HSD17B4 methylation was at the transcriptional start site of its major variant, and was associated with its silencing. HSD17B4 was highly expressed in the vast majority of human cancers, and its methylation was present only in breast cancers and one lymphoblastic leukemia cell line. A combination of estrogen receptor-negative status and HSD17B4 methylation showed a positive predictive value of 80.0%. During HER2-directed neoadjuvant therapy, HSD17B4 methylation was the most reliable marker to monitor response to the therapy. These results showed that HSD17B4 methylation is a candidate predictive and response marker of HER2-positive breast cancer to HER2-directed therapy.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais , Neoplasias da Mama/genética , Proteína Multifuncional do Peroxissomo-2/genética , Receptor ErbB-2/genética , Biomarcadores Tumorais/genética , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Metilação de DNA , DNA de Neoplasias/genética , Feminino , Inativação Gênica , Humanos , Pessoa de Meia-Idade , Receptor ErbB-2/antagonistas & inibidores , Transcriptoma , Resultado do Tratamento
6.
Aging (Albany NY) ; 12(14): 14699-14717, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32678070

RESUMO

Steroidogenic enzymes are crucial in prostate cancer (PCa) progression. 17ß-Hydroxysteroid dehydrogenase type 4 (HSD17B4), encoded by HSD17B4, lacks catalytic capacity in androgen metabolism. Now the detailed role and molecular mechanism of PCa development are largely unknown. Here we showed that the expression of HSD17B4 was increased in PCa tissues compared to paired paratumor tissues. HSD17B4 knockdown in PCa cells significantly suppressed its proliferation, migration and invasion, while overexpressing HSD17B4 had opposite effects. Mechanistically, we found that the protein level of HSD17B4 was regulated by its acetylation at lysine 669(K669). Dihydroxytestosterone (DHT) treatment increased HSD17B4 acetylation and then promoted its degradation via chaperone-mediated autophagy (CMA). SIRT3 directly interacted with HSD17B4 to inhibit its acetylation and enhance its stability. In addition, we identified CREBBP as a regulator of the K669 acetylation and degradation of HSD17B4, affecting PC cell proliferation, migration and invasion. Notably, in PCa tissues and paired paratumor tissues, the level of HSD17B4 was negatively correlated with its K669 acetylation. Taken together, this study identified a novel role of HSD17B4 in PCa progression and suggested that HSD17B4 and its upstream regulators may be potential therapeutic targets for PCa intervention.


Assuntos
Proteína Multifuncional do Peroxissomo-2/genética , Proteína Multifuncional do Peroxissomo-2/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Acetilação , Animais , Proteína de Ligação a CREB/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Lisina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Invasividade Neoplásica/genética , Sirtuína 3/genética , Testosterona/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncol Rep ; 41(3): 2009-2019, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30747222

RESUMO

Accumulating evidence has implicated that the activation of signal transducer and activator of transcription 3 (STAT3) contributes to the progression of liver cancer by affecting the expression of proliferation­associated genes. A previous study reported that elevated levels of 17ß­hydroxysteroid dehydrogenase 4 (HSD17B4) are observed in patients with liver cancer. The current study investigated how upregulated HSD17B4 expression promoted the expression of proliferation­associated genes in rats with liver cancer. HSD17B4 expression in rats with liver cancer was significantly increased compared with the control group as determined by reverse transcription­quantitative polymerase chain reaction and western blot assays. Immunohistochemical results revealed that STAT3 activation was positively correlated with increased HSD17B4 expression in tumor tissues from patients with liver cancer. Western blot results further suggested that HSD17B4 overexpression increased STAT3 activation via the protein kinase B and the mitogen­activated protein kinase/extracellular­signal­regulated kinase signaling pathways in HepG2 cells. The present study suggested that overexpression may promote HepG2 proliferation by enhancing expression of various downstream targets of STAT3. Targeted inhibition of HSD17B4 may describe a novel approach in the prevention and treatment of liver cancer.


Assuntos
Proliferação de Células/genética , Neoplasias Hepáticas/genética , Proteína Multifuncional do Peroxissomo-2/genética , Fator de Transcrição STAT3/genética , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Células Hep G2 , Humanos , Fígado/patologia , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Ratos Wistar , Transdução de Sinais/genética , Ativação Transcricional/genética , Regulação para Cima/genética
9.
Mol Cell Neurosci ; 80: 123-133, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28286294

RESUMO

An important hallmark of various neurodegenerative disorders is the proliferation and activation of microglial cells, the resident immune cells of the central nervous system (CNS). Mice that lack multifunctional protein-2 (MFP2), the key enzyme in peroxisomal ß-oxidation, develop excessive microgliosis that positively correlates with behavioral deficits whereas no neuronal loss occurs. However, the precise contribution of neuroinflammation to the fatal neuropathology of MFP2 deficiency remains largely unknown. Here, we first attempted to suppress the inflammatory response by administering various anti-inflammatory drugs but they failed to reduce microgliosis. Subsequently, Mfp2-/- mice were treated with the selective colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 as microglial proliferation and survival is dependent on CSF1R signaling. This resulted in the elimination of >95% of microglia from control mice but only 70% of the expanded microglial population from Mfp2-/- mice. Despite microglial diminution in Mfp2-/- brain, inflammatory markers remained unaltered and residual microglia persisted in a reactive state. CSF1R inhibition did not prevent neuronal dysfunction, cognitive decline and clinical deterioration of Mfp2-/- mice. Collectively, the unaltered inflammatory profile despite suppressed microgliosis concurrent with persevering clinical decline strengthens our hypothesis that neuroinflammation importantly contributes to the Mfp2-/- phenotype.


Assuntos
Anti-Inflamatórios/uso terapêutico , Encefalite , Gliose/etiologia , Proteína Multifuncional do Peroxissomo-2/deficiência , Estimulação Acústica , Análise de Variância , Animais , Anti-Inflamatórios/farmacologia , Antígenos de Diferenciação/metabolismo , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Encefalite/complicações , Encefalite/genética , Encefalite/patologia , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Potenciais Evocados Auditivos do Tronco Encefálico/genética , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Força Muscular/efeitos dos fármacos , Força Muscular/genética , Proteína Multifuncional do Peroxissomo-2/genética , Índice de Gravidade de Doença
10.
Autophagy ; 13(3): 538-553, 2017 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-28296597

RESUMO

Dysregulation of hormone metabolism is implicated in human breast cancer. 17ß-hydroxysteroid dehydrogenase type 4 (HSD17B4) catalyzes the conversion of estradiol (E2) to estrone (E1), and is associated with the pathogenesis and development of various cancers. Here we show that E1 upregulates HSD17B4 acetylation at lysine 669 (K669) and thereby promotes HSD17B4 degradation via chaperone-mediated autophagy (CMA), while a single mutation at K669 reverses the degradation and confers migratory and invasive properties to MCF7 cells upon E1 treatment. CREBBP and SIRT3 dynamically control K669 acetylation level of HSD17B4 in response to E1. More importantly, K669 acetylation is inversely correlated with HSD17B4 in human breast cancer tissues. Our study reveals a crosstalk between acetylation and CMA degradation in HSD17B4 regulation, and a critical role of the regulation in the malignant progression of breast cancer.


Assuntos
Autofagia/efeitos dos fármacos , Estrona/farmacologia , Proteína Multifuncional do Peroxissomo-2/metabolismo , Proteólise/efeitos dos fármacos , Acetilação , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Lisina/metabolismo , Modelos Biológicos , Mutação/genética , Invasividade Neoplásica , Proteína Multifuncional do Peroxissomo-2/genética , Sirtuína 3/metabolismo , Especificidade por Substrato/efeitos dos fármacos
11.
J Immunol ; 198(6): 2414-2425, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28179495

RESUMO

Peroxisomes are proposed to play an important role in the regulation of systemic inflammation; however, the functional role of these organelles in inflammatory responses of myeloid immune cells is largely unknown. In this article, we demonstrate that the nonclassical peroxisome proliferator 4-phenyl butyric acid is an efficient inducer of peroxisomes in various models of murine macrophages, such as primary alveolar and peritoneal macrophages and the macrophage cell line RAW264.7, but not in primary bone marrow-derived macrophages. Further, proliferation of peroxisomes blocked the TLR4 ligand LPS-induced proinflammatory response, as detected by the reduced induction of the proinflammatory protein cyclooxygenase (COX)-2 and the proinflammatory cytokines TNF-α, IL-6, and IL-12. In contrast, disturbing peroxisome function by knockdown of peroxisomal gene Pex14 or Mfp2 markedly increased the LPS-dependent upregulation of the proinflammatory proteins COX-2 and TNF-α. Specifically, induction of peroxisomes did not affect the upregulation of COX-2 at the mRNA level, but it reduced the half-life of COX-2 protein, which was restored by COX-2 enzyme inhibitors but not by proteasomal and lysosomal inhibitors. Liquid chromatography-tandem mass spectrometry analysis revealed that various anti-inflammatory lipid mediators (e.g., docosahexaenoic acid) were increased in the conditioned medium from peroxisome-induced macrophages, which blocked LPS-induced COX-2 upregulation in naive RAW264.7 cells and human primary peripheral blood-derived macrophages. Importantly, LPS itself induced peroxisomes that correlated with the regulation of COX-2 during the late phase of LPS activation in macrophages. In conclusion, our findings identify a previously unidentified role for peroxisomes in macrophage inflammatory responses and suggest that peroxisomes are involved in the physiological cessation of macrophage activation.


Assuntos
Ativação de Macrófagos , Macrófagos/imunologia , Peroxissomos/imunologia , Fenilbutiratos/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína Multifuncional do Peroxissomo-2/genética , Cultura Primária de Células , Células RAW 264.7 , Proteínas Repressoras/genética
12.
Oncotarget ; 8(12): 19039-19048, 2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28186977

RESUMO

Human epidermal growth factor (HER) 2-directed therapy is the standard treatment for HER2-positive breast cancer. Patients who achieved a pathological complete response (pCR) to the therapy are associated with excellent disease-free survival. However, few molecular markers are available to predict pCR. Here, we aimed to establish a DNA methylation marker to predict the response to trastuzumab and chemotherapy. A total of 67 patients were divided into screening (n = 21) and validation (n = 46) sets. Genome-wide DNA methylation analysis of the screening set identified eight genomic regions specifically methylated in patients with pCR. Among these, HSD17B4 encoding type 4 17ß-hydroxysteroid dehydrogenase was most significantly differentially methylated. The differential methylation was confirmed by pyrosequencing (P = 0.03), and a cutoff value was determined. This association was successfully validated in the validation set (P < 0.001), and patients with pCR were predicted with a high specificity (79%). Multivariate analysis, including tumor stage and hormone receptor status, showed that HSD17B4 methylation was an independent predictive factor (odds ratio: 10.0, 95% confidence interval 2.54-39.50, P = 0.001). Combination with ER status and HSD17B4 methylation improved the specificity up to 91%. Identification of HER2-positive breast cancer patients who would achieve pCR only by trastuzumab and chemotherapy may lead to surgery-free treatment for this group of breast cancer patients.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Metilação de DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Proteína Multifuncional do Peroxissomo-2/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Ilhas de CpG/genética , Feminino , Humanos , Regiões Promotoras Genéticas/genética , Receptor ErbB-2/genética , Trastuzumab/uso terapêutico , Resultado do Tratamento
13.
Sci Rep ; 5: 15136, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26456697

RESUMO

SQAP is a novel and promising anticancer agent that was obtained by structural modifications from a natural compound. SQAP inhibits angiogenesis in vivo resulting in increased hypoxia and reduced tumor volume. In this study, the mechanism by which SQAP modifies the tumor microenvironment was revealed through the application of a T7 phage display screening. This approach identified five SQAP-binding proteins including sterol carrier protein 2, multifunctional enzyme type 2, proteasomal ubiquitin receptor, UV excision repair protein and focal adhesion kinase (FAK). All the interactions were confirmed by surface plasmon resonance analysis. Since FAK plays an important role in cell turnover and angiogenesis, the influence of SQAP on FAK was the principal goal of this study. SQAP decreased FAK phosphorylation and cell migration in human umbilical vein endothelial cells and A549 cancer cells. These findings suggest that inhibition of FAK phosphorylation works as the mechanism for the anti-angiogenesis activity of SQAP.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Pequenas/tratamento farmacológico , Quinase 1 de Adesão Focal/antagonistas & inibidores , Glicolipídeos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Sequência de Aminoácidos , Animais , Antineoplásicos/síntese química , Sítios de Ligação , Carcinoma de Células Pequenas/enzimologia , Carcinoma de Células Pequenas/genética , Carcinoma de Células Pequenas/patologia , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Enzimas Reparadoras do DNA/química , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Quinase 1 de Adesão Focal/química , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Glicolipídeos/síntese química , Células Endoteliais da Veia Umbilical Humana , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Biblioteca de Peptídeos , Proteína Multifuncional do Peroxissomo-2/química , Proteína Multifuncional do Peroxissomo-2/genética , Proteína Multifuncional do Peroxissomo-2/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
14.
PLoS One ; 10(6): e0128749, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26046837

RESUMO

The discrete effects of obesity on infertility in females remain undefined to date. To investigate obesity-induced ovarian dysfunction, we characterized metabolic parameters, steroidogenesis, and folliculogenesis in obese and lean female Ossabaw mini-pigs. Nineteen nulliparous, sexually mature female Ossabaw pigs were fed a high fat/cholesterol/fructose diet (n=10) or a control diet (n=9) for eight months. After a three-month diet-induction period, pigs remained on their respective diets and had ovarian ultrasound and blood collection conducted during a five-month study period after which ovaries were collected for histology, cell culture, and gene transcript level analysis. Blood was assayed for steroid and protein hormones. Obese pigs developed abdominal obesity and metabolic syndrome, including hyperglycemia, hypertension, insulin resistance and dyslipidemia. Obese pigs had elongated estrous cycles and hyperandrogenemia with decreased LH, increased FSH and luteal phase progesterone, and increased numbers of medium, ovulatory, and cystic follicles. Theca cells of obese, compared to control, pigs displayed androstenedione hypersecretion in response to in vitro treatment with LH, and up-regulated 3-beta-hydroxysteroid dehydrogenase 1 and 17-beta-hydroxysteroid dehydrogenase 4 transcript levels in response to in vitro treatment with LH or LH + insulin. Granulosa cells of obese pigs had increased 3-beta-hydroxysteroid dehydrogenase 1 transcript levels. In summary, obese Ossabaw pigs have increased transcript levels and function of ovarian enzymes in the delta 4 steroidogenic pathway. Alterations in LH, FSH, and progesterone, coupled with theca cell dysfunction, contribute to the hyperandrogenemia and disrupted folliculogenesis patterns observed in obese pigs. The obese Ossabaw mini-pig is a useful animal model in which to study the effects of obesity and metabolic syndrome on ovarian function and steroidogenesis. Ultimately, this animal model may be useful toward the development of therapies to improve fertility in obese and/or hyperandrogenemic females or in which to examine the effects of obesity on the maternal-fetal environment and offspring health.


Assuntos
Androstenodiona/sangue , Hormônio Foliculoestimulante/sangue , Infertilidade Feminina/metabolismo , Hormônio Luteinizante/sangue , Síndrome Metabólica/metabolismo , Obesidade/metabolismo , Progesterona/sangue , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Ciclo Estral , Feminino , Hormônio Foliculoestimulante/farmacologia , Expressão Gênica , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Humanos , Infertilidade Feminina/etiologia , Infertilidade Feminina/genética , Infertilidade Feminina/patologia , Insulina/farmacologia , Hormônio Luteinizante/farmacologia , Síndrome Metabólica/etiologia , Síndrome Metabólica/genética , Síndrome Metabólica/patologia , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia , Proteína Multifuncional do Peroxissomo-2/genética , Proteína Multifuncional do Peroxissomo-2/metabolismo , Cultura Primária de Células , RNA Mensageiro/agonistas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos , Porco Miniatura , Células Tecais/efeitos dos fármacos , Células Tecais/metabolismo , Células Tecais/patologia
15.
Mol Cell Endocrinol ; 401: 1-11, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25448063

RESUMO

Hepatocellular carcinoma (HCC) arises in a setting of chronic inflammation induced by inflammatory cytokines, such as nuclear factor-kappaB (NF-κB). HCC is a male-predominant cancer that can be attenuated by estradiol (E2) in vitro and in vivo. Although 17ß-hydroxysteroid dehydrogenase 4 (HSD17B4) has been implicated as an estradiol-inactivating enzyme, and its promoter sequence contains two putative NF-κB elements: it is currently unknown whether HSD17B4 is the link between inflammation, estradiol and proliferation in hepatoma cells. In this study, HepG2 cells were used to investigate the role of HSD17B4 in the proliferation of liver cancer cells treated with the NF-κB activator, tumor necrosis factor-alpha (TNF-α), with the inhibitor of NF-κB activation, pyrrolidinedithiocarbamate (PDTC), or with a related specific siRNA. We demonstrated that the human HSD17B4 gene is a target for NF-κB activation in inflammation-stimulated HepG2 cells. HSD17B4 is up-regulated via the binding of activated NF-κB to the distal NF-κB-responsive element via TNF-α stimulation, which then promotes cell proliferation by decreasing the levels of E2 and enhancing the expression of interleukin 6 (IL-6), cyclin D1 and proliferating cell nuclear antigen (PCAN). These results from HepG2 cells are consistent with the observation that HSD17B4 is highly expressed and activated NF-κB is highly co-localized with the NF-κB-responsive element of HSD17B4 in liver tumor tissues from HCC patients. Our findings indicate for the first time that HSD17B4 plays an important role in aggravated HCC progression and provides a novel therapeutic target for HCC.


Assuntos
Carcinoma Hepatocelular/patologia , Estradiol/metabolismo , Neoplasias Hepáticas/patologia , NF-kappa B/metabolismo , Proteína Multifuncional do Peroxissomo-2/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Células Hep G2 , Humanos , Interleucina-6/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Proteína Multifuncional do Peroxissomo-2/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Prolina/análogos & derivados , Prolina/farmacologia , Regiões Promotoras Genéticas , Tiocarbamatos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima
16.
BMC Med Genet ; 15: 30, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24602372

RESUMO

BACKGROUND: D-bifunctional protein deficiency, caused by recessive mutations in HSD17B4, is a severe, infantile-onset disorder of peroxisomal fatty acid oxidation. Few affected patients survive past two years of age. Compound heterozygous mutations in HSD17B4 have also been reported in two sisters diagnosed with Perrault syndrome (MIM # 233400), who presented in adolescence with ovarian dysgenesis, hearing loss, and ataxia. CASE PRESENTATION: An adult male presented with cerebellar ataxia, peripheral neuropathy, hearing loss, and azoospermia. The clinical presentation, in combination with biochemical findings in serum, urine, and muscle biopsy, suggested a mitochondrial disorder. Commercial genetic testing of 18 ataxia and mitochondrial disease genes was negative. Targeted exome sequencing followed by analysis of single nucleotide variants and small insertions/deletions failed to reveal a genetic basis of disease. Application of a computational algorithm to infer copy number variants (CNVs) from exome data revealed a heterozygous 12 kb deletion of exons 10-13 of HSD17B4 that was compounded with a rare missense variant (p.A196V) at a highly conserved residue. Retrospective review of patient records revealed mildly elevated ratios of pristanic:phytanic acid and arachidonic:docosahexaenoic acid, consistent with dysfunctional peroxisomal fatty acid oxidation. CONCLUSION: Our case expands the phenotypic spectrum of HSD17B4-deficiency, representing the first male case reported with infertility. Furthermore, it points to crosstalk between mitochondria and peroxisomes in HSD17B4-deficiency and Perrault syndrome.


Assuntos
Anormalidades Múltiplas/diagnóstico , Ataxia/diagnóstico , Perda Auditiva Neurossensorial/diagnóstico , Doenças Mitocondriais/diagnóstico , Proteína Multifuncional do Peroxissomo-2/deficiência , Anormalidades Múltiplas/enzimologia , Anormalidades Múltiplas/genética , Adulto , Ataxia/enzimologia , Ataxia/genética , Azoospermia/diagnóstico , Azoospermia/enzimologia , Azoospermia/genética , Sequência de Bases , Variações do Número de Cópias de DNA , Dosagem de Genes , Perda Auditiva Neurossensorial/enzimologia , Perda Auditiva Neurossensorial/genética , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Doenças Mitocondriais/enzimologia , Doenças Mitocondriais/genética , Técnicas de Diagnóstico Molecular , Dados de Sequência Molecular , Proteína Multifuncional do Peroxissomo-2/genética , Fenótipo , Análise de Sequência de DNA , Deleção de Sequência
17.
Neurobiol Dis ; 58: 258-69, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23777740

RESUMO

Although peroxisome biogenesis and ß-oxidation disorders are well known for their neurodevelopmental defects, patients with these disorders are increasingly diagnosed with neurodegenerative pathologies. In order to investigate the cellular mechanisms of neurodegeneration in these patients, we developed a mouse model lacking multifunctional protein 2 (MFP2, also called D-bifunctional protein), a central enzyme of peroxisomal ß-oxidation, in all neural cells (Nestin-Mfp2(-/-)) or in oligodendrocytes (Cnp-Mfp2(-/-)) and compared these models with an already established general Mfp2 knockout. Nestin-Mfp2 but not Cnp-Mfp2 knockout mice develop motor disabilities and ataxia, similar to the general mutant. Deterioration of motor performance correlates with the demise of Purkinje cell axons in the cerebellum, which precedes loss of Purkinje cells and cerebellar atrophy. This closely mimics spinocerebellar ataxias of patients affected with mild peroxisome ß-oxidation disorders. However, general knockouts have a much shorter life span than Nestin-Mfp2 knockouts which is paralleled by a disparity in activation of the innate immune system. Whereas in general mutants a strong and chronic proinflammatory reaction proceeds throughout the brain, elimination of MFP2 from neural cells results in minor neuroinflammation. Neither the extent of the inflammatory reaction nor the cerebellar degeneration could be correlated with levels of very long chain fatty acids, substrates of peroxisomal ß-oxidation. In conclusion, MFP2 has multiple tasks in the adult brain, including the maintenance of Purkinje cells and the prevention of neuroinflammation but this is not mediated by its activity in oligodendrocytes nor by its role in very long chain fatty acid degradation.


Assuntos
Deficiências Nutricionais/complicações , Encefalite/etiologia , Ácidos Graxos/metabolismo , Degeneração Neural/etiologia , Proteína Multifuncional do Peroxissomo-2/deficiência , Células de Purkinje/patologia , 2',3'-Nucleotídeo Cíclico 3'-Fosfodiesterase/genética , Fatores Etários , Animais , Antígenos de Diferenciação/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Regulação da Expressão Gênica/genética , Locomoção/fisiologia , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Proteína Básica da Mielina/metabolismo , Nestina/genética , Proteína Multifuncional do Peroxissomo-2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA