Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Sci ; 136(12)2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37232206

RESUMO

Mitochondrial dynamics regulate the quality and morphology of mitochondria. Calcium (Ca2+) plays an important role in regulating mitochondrial function. Here, we investigated the effects of optogenetically engineered Ca2+ signaling on mitochondrial dynamics. More specifically, customized illumination conditions could trigger unique Ca2+ oscillation waves to trigger specific signaling pathways. In this study, we found that modulating Ca2+ oscillations by increasing the light frequency, intensity and exposure time could drive mitochondria toward the fission state, mitochondrial dysfunction, autophagy and cell death. Moreover, illumination triggered phosphorylation at the Ser616 residue but not the Ser637 residue of the mitochondrial fission protein, dynamin-related protein 1 (DRP1, encoded by DNM1L), via the activation of Ca2+-dependent kinases CaMKII, ERK and CDK1. However, optogenetically engineered Ca2+ signaling did not activate calcineurin phosphatase to dephosphorylate DRP1 at Ser637. In addition, light illumination had no effect on the expression levels of the mitochondrial fusion proteins mitofusin 1 (MFN1) and 2 (MFN2). Overall, this study provides an effective and innovative approach to altering Ca2+ signaling for controlling mitochondrial fission with a more precise resolution than pharmacological approaches in the temporal dimension.


Assuntos
Cálcio , Dinâmica Mitocondrial , Dinâmica Mitocondrial/fisiologia , Cálcio/metabolismo , Dinaminas/genética , Dinaminas/metabolismo , Mitocôndrias/metabolismo , Fosforilação , Morte Celular , Proteínas Mitocondriais/metabolismo
2.
Reprod Med Biol ; 23(1): e12588, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38854774

RESUMO

Background: Endometriosis is a common gynecological disease affecting women of reproductive age. Patients with endometriosis frequently experience severe chronic pain and have higher chances to experience infertility. Progesterone resistance is a major problem that develops during the medical treatment of endometriosis, which often leads to treatment failure of hormonal therapies. Previous studies indicated that the dysregulation of progesterone receptors (PR) is the primary factor leading to progesterone resistance in endometriosis. Methods: This review article systematically reviewed and summarized findings extracted from previously published papers available on PubMed, encompassing both experimental studies and clinical trials. Main findings: Various determinants influencing PR expression in endometriosis have been identified, including the environmental toxins, microRNAs, cell signaling pathways, genetic mutations, and the pro-inflammatory cytokines. The selective estrogen/progesterone receptor modulators have emerged as novel therapeutic approaches for treating endometriosis, offering potential improvements in overcoming progesterone resistance. Conclusion: Concerns and limitations persist despite the newly developed drugs. Therefore, studies on unraveling new therapeutic targets based on the molecular mechanisms of progesterone resistance is warranted for the development potential alternatives to overcome hormonal treatment failure in endometriosis.

3.
Hum Reprod ; 38(6): 1124-1134, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37071897

RESUMO

STUDY QUESTION: Does YAP1 inhibition alleviate progesterone resistance in endometriosis? SUMMARY ANSWER: YAP1 inhibition reduces progesterone resistance in vitro and in vivo. WHAT IS KNOWN ALREADY: Progesterone resistance not only causes treatment failure for endometriosis but also inhibits eutopic endometrial cell proliferation, dysregulates decidualization, and reduces the success rates of pregnancy. Hippo/yes-associated protein 1 (YAP1) signaling pathway plays an important role in the pathogenesis of endometriosis. STUDY DESIGN, SIZE, DURATION: Paraffin-embedded tissues containing paired endometriotic and endometrial specimens (n = 42) and serum samples isolated from normal controls (n = 15) or endometriotic patients with (n = 25) or without (n = 21) prior dienogest treatment were analyzed. A mouse model of endometriosis was also used to evaluate the effects of YAP1 inhibition on progesterone resistance. PARTICIPANTS/MATERIALS, SETTING, METHODS: Primary endometriotic and endometrial stromal cells treated with YAP1 inhibitor or miR-21 mimic/inhibitor were used for the in vitro studies including decidualization induction, chromatin immunoprecipitation (ChIP), and RNA immunoprecipitation. Tissue specimens and serum from human and mouse were used for immunohistochemistry staining, exosome isolation, and microRNA (miRNA) quantification, respectively. MAIN RESULTS AND THE ROLE OF CHANCE: Herein, we report, by using ChIP-PCR and RNA-IP, that YAP1 inhibits progesterone receptor (PGR) expression through upregulation of miR-21-5p. Upregulation of miR-21-5p not only reduces PGR expression but also inhibits endometrial stromal cell decidualization. Indeed, levels of YAP1 and miR-21-5p are inversely correlated with the level of PGR in human endometrial samples. In contrast, knockdown of YAP1 or treatment with verteporfin (VP), a YAP1 inhibitor, reduces miR-21-5p expression, thus leading to an increase in PGR expression in ectopic endometriotic stromal cells. In the mouse model of endometriosis, treatment with VP increases PGR expression and enhances decidualization. More importantly, VP synergistically increases the treatment effect of progestin in causing the regression of endometriotic lesions and improves the decidualization capability of the endometrium. Interestingly, treatment with dienogest, a synthetic progestin, reduces YAP1 and miR-21-5p expression in human cells and in the mouse model of endometriosis. Patients who received dienogest treatment for 6 months show a significant decrease in serum extracellular vesicle-associated miR-21-5p level. LARGE SCALE DATA: A public dataset (GSE51981) containing a large cohort of endometriotic tissues is available from the Gene Expression Omnibus (GEO). LIMITATIONS, REASONS FOR CAUTION: A large cohort of clinical samples is needed to verify the current diagnostic value of miR-21-5p in future studies. WIDER IMPLICATIONS OF THE FINDINGS: The reciprocal regulation of YAP1 and PGR suggests that combined YAP1 inhibitor and progestin may be a better therapeutic approach for treating endometriosis. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by the Ministry of Science and Technology, Taiwan (MOST-111-2636-B-006-012, MOST-111-2314-B-006-075-MY3, and MOST-106-2320-B-006-072-MY3). The authors have no conflict of interest to disclose.


Assuntos
Endometriose , MicroRNAs , Gravidez , Feminino , Humanos , Animais , Camundongos , Endometriose/patologia , Progestinas/uso terapêutico , Endométrio/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Progesterona/metabolismo , Fatores de Transcrição/metabolismo , Células Estromais/metabolismo
4.
Br J Dermatol ; 189(6): 719-729, 2023 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-37540988

RESUMO

BACKGROUND: Hidradenitis suppurativa (HS) significantly diminishes the quality of life for patients. Delayed diagnosis represents a significant challenge in effectively managing HS. OBJECTIVES: To identify and characterize the key mediator in HS. METHODS: Bioinformatic transcriptomic analysis was applied to identify potential candidates contributing to the disease process of HS. Skin samples from 40 patients with HS, four with psoriasis and 29 with normal skin were included. The expression of interleukin (IL)-17A was evaluated and compared among samples of normal skin, psoriatic skin and skin from different stages of HS by immunohistochemistry or dual-colour immunofluorescence. In vitro experiments and RNA sequencing analysis were also conducted to validate the expression of IL-17A and its pathogenic effect in HS. RESULTS: Transcriptomic database analyses identified IL-17 signalling as a potential contributor to HS. In HS, the predominant IL-17A+ cell population was identified as mast cells. IL-17A+ mast-cell density was significantly elevated in HS, especially in samples with advanced Hurley stages, compared with normal skin and psoriasis samples. The close contact between IL-17A+ mast cells and IL-17 receptor A (IL-17RA)-expressing keratinocytes was demonstrated, along with the significant effects of IL-17A on keratinocyte cell proliferation and HS pathogenic gene expression. Treatment with biologics (brodalumab or adalimumab) reduced the severity of the disease and the number of IL-17A+ mast cells in affected tissues. CONCLUSIONS: The presence of high-density IL-17A+ mast cells may serve as a valuable pathological marker for diagnosing HS. Moreover, developing therapeutic drugs targeting IL-17A+ mast cells may provide a new approach to treating HS.


Assuntos
Hidradenite Supurativa , Psoríase , Humanos , Hidradenite Supurativa/tratamento farmacológico , Interleucina-17/metabolismo , Mastócitos/metabolismo , Psoríase/patologia , Qualidade de Vida , Pele/patologia
5.
Proc Natl Acad Sci U S A ; 117(41): 25859-25868, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33004630

RESUMO

Endometriosis is a highly prevalent gynecological disease with severe negative impacts on life quality and financial burden. Unfortunately, there is no cure for this disease, which highlights the need for further investigation about the pathophysiology of this disease to provide clues for developing novel therapeutic regimens. Herein, we identified that vascular endothelial growth factor (VEGF)-C, a potent lymphangiogenic factor, is up-regulated in endometriotic cells and contributes to increased lymphangiogenesis. Bioinformatic analysis and molecular biological characterization revealed that VEGF-C is negatively regulated by an orphan nuclear receptor, chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII). Further studies demonstrated that proinflammatory cytokines, via suppression of COUP-TFII level, induce VEGF-C overexpression. More importantly, we show that functional VEGF-C is transported by extracellular vesicles (EVs) to enhance the lymphangiogenic ability of lymphatic endothelial cells. Autotransplanted mouse model of endometriosis showed lenvatinib treatment abrogated the increased lymphatic vessels development in the endometriotic lesion, enlarged retroperitoneal lymph nodes, and immune cells infiltration, indicating that blocking VEGF-C signaling can reduce local chronic inflammation and concomitantly endometriosis development. Evaluation of EV-transmitted VEGF-C from patients' sera demonstrates it is a reliable noninvasive way for clinical diagnosis. Taken together, we identify the vicious cycle of inflammation, COUP-TFII, VEGF-C, and lymphangiogenesis in the endometriotic microenvironment, which opens up new horizons in understanding the pathophysiology of endometriosis. VEGF-C not only can serve as a diagnostic biomarker but also a molecular target for developing therapeutic regimens.


Assuntos
Endometriose/imunologia , Vesículas Extracelulares/imunologia , Sistema Imunitário/imunologia , Linfangiogênese , Fator C de Crescimento do Endotélio Vascular/imunologia , Animais , Fator II de Transcrição COUP/genética , Fator II de Transcrição COUP/imunologia , Citocinas/genética , Citocinas/imunologia , Endometriose/genética , Endometriose/fisiopatologia , Células Endoteliais/imunologia , Vesículas Extracelulares/genética , Feminino , Humanos , Vasos Linfáticos/imunologia , Camundongos , Fator C de Crescimento do Endotélio Vascular/genética
7.
J Cell Physiol ; 237(12): 4487-4503, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36251015

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers because of its late diagnosis and chemoresistance. Primary cilia, the cellular antennae, are observed in most human cells to maintain development and differentiation. Primary cilia are gradually lost during the progression of pancreatic cancer and are eventually absent in PDAC. Here, we showed that cisplatin-resistant PDAC regrew primary cilia. Additionally, genetic or pharmacological disruption of primary cilia sensitized PDAC to cisplatin treatment. Mechanistically, ataxia telangiectasia mutated (ATM) and ATM and RAD3-related (ATR), tumor suppressors that initiate DNA damage responses, promoted the excessive formation of centriolar satellites (EFoCS) and autophagy activation. Disruption of EFoCS and autophagy inhibited primary ciliogenesis, sensitizing PDAC cells to cisplatin treatment. Collectively, our findings revealed an unexpected interplay among the DNA damage response, primary cilia, and chemoresistance in PDAC and deciphered the molecular mechanism by which ATM/ATR-mediated EFoCS and autophagy cooperatively regulate primary ciliogenesis.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia , Carcinoma Ductal Pancreático , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pancreáticas , Humanos , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Dano ao DNA , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Cílios , Neoplasias Pancreáticas
8.
J Cell Physiol ; 236(6): 4681-4693, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33244795

RESUMO

The ability of a single Ca2+ ion to play an important role in cell biology is highlighted by the need for cells to form Ca2+ signals in the dimensions of space, time, and amplitude. Thus, spatial and temporal changes in intracellular Ca2+ concentration are important for determining cell fate. Optogenetic technology has been developed to provide more precise and targeted stimulation of cells. Here, U2OS cells overexpressing Ca2+ translocating channelrhodopsin (CatCh) were used to mediate Ca2+ influx through blue light illumination with various parameters, such as intensity, frequency, duty cycle, and duration. We identified that several Ca2+ -dependent transcription factors and certain kinases can be activated by specific Ca2+ waves. Using a wound-healing assay, we found that low-frequency Ca2+ oscillations increased cell migration through the activation of NF-κB. This study explores the regulation of cell migration by Ca2+ signals. Thus, we can choose optical parameters to modulate Ca2+ waves and achieve activation of specific signaling pathways. This novel methodology can be applied to clarify related cell-signaling mechanisms in the future.


Assuntos
Neoplasias Ósseas/metabolismo , Sinalização do Cálcio , Cálcio/metabolismo , Movimento Celular , Optogenética , Osteossarcoma/metabolismo , Análise de Célula Única , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Humanos , Microscopia de Fluorescência , NF-kappa B/metabolismo , Osteossarcoma/genética , Osteossarcoma/patologia , Fatores de Tempo , Imagem com Lapso de Tempo
9.
Reproduction ; 161(1): 11-19, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33112285

RESUMO

Endometriosis is a common gynecological disease in reproductive-age women. Although the hormone-dependent therapy is the first line treatment for endometriosis, it is not a curative regimen and associated with severe side-effects, which significantly decrease the life quality of affected individuals. To seek a target for treatment of endometriosis, we focused on plasma membrane proteins that are elevated in ectopic cells and exert beneficial effects in cell growth and survival. We performed bioinformatics analysis and identified the neurotrophic receptor tyrosine kinase 2 (NTRK2) as a potential candidate for treatment. The expression levels of NTRK2 were markedly upregulated in the lesions of clinical specimen as well as in the mouse endometriotic-like lesion. Mechanistic investigation demonstrated that upregulation of NTRK2 is induced by hypoxia in a hypoxia-inducible factor 1 alpha-dependent manner. Knockdown of NTRK2 or administration of ANA-12, a selective antagonist of NTRK2, significantly induced endometriotic stromal cells death, suggesting it may be a potential therapeutic agent. In vivo study using surgery-induced endometriosis mice model showed ANA-12 (1.5 mg/kg body weight) treatment induced apoptosis of endometriotic cells and caused the regression of ectopic lesions. Taken together, our findings suggest a possible mechanism responsible for the aberrant expression of NTRK2 in endometriotic lesions and this may be involved in the pathogenesis of endometriosis.


Assuntos
Endometriose/tratamento farmacológico , Glicoproteínas de Membrana/metabolismo , RNA Interferente Pequeno/uso terapêutico , Receptor trkB/metabolismo , Animais , Coristoma/metabolismo , Avaliação Pré-Clínica de Medicamentos , Endometriose/metabolismo , Feminino , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Cultura Primária de Células , RNA Interferente Pequeno/farmacologia , Receptor trkB/antagonistas & inibidores , Células Estromais/metabolismo
10.
Reproduction ; 161(1): F19-F31, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33112784

RESUMO

Endometriosis is a benign gynecological disease that affects about 10% of women of reproductive age. Patients with endometriosis suffer from long-term coexistence with dysmenorrhea, dyspareunia, and even infertility, which severely reduces quality of life. So far, surgical removal and hormonal medication are the major treatment options; however, high recurrence and severe adverse effects hamper the therapeutic efficacy. Hypoxia is an inevitable cellular stress in many diseases that regulates the expression of a significant subset of genes involved in pathophysiological processes. A growing body of evidence demonstrates that hypoxia plays critical role in controlling the disease phenotypes of endometriosis, such as increasing adhesion ability, causing dysregulation of estrogen biosynthesis, aberrant production of proinflammatory cytokines, increasing angiogenic ability, and suppression of immune functions. In this review, we summarize the findings of the most recent studies in exploring the underlying mechanisms of hypoxia involved in endometriosis. Potential therapeutic options for targeting HIF and downstream effectors will also be discussed.


Assuntos
Endometriose/etiologia , Hipóxia/metabolismo , Endometriose/metabolismo , Epigênese Genética , Feminino , Redes Reguladoras de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo
11.
Chin J Physiol ; 64(6): 257-265, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975118

RESUMO

Hidradenitis suppurativa is a cutaneous chronic inflammatory disease that is estimated to affect about 1% of the population and caused pain, malodorous discharge, disfigurement, and poor quality of life with psychosocial problems. The typical features are recurrent painful nodules, abscesses, and sinus tracts on the axillae, groins, gluteal areas, and anogenital regions since postpuberty. Smoking and obesity are two major triggering factors of hidradenitis suppurativa. Women are prone to have hidradenitis suppurativa than men in Western countries, but the male-to-female ratio is reversed in oriental countries. The disease severity can be affected by menstruation, pregnancy, and menopause. Furthermore, the phenotypes are different among men and women with hidradenitis suppurativa. Men are prone to have buttock involvement while women are prone to have axillary, groins, and submammary lesions. This review introduces the skin appendages and pathophysiology of hidradenitis suppurativa and then focuses on the sex difference and the effects of sex hormones on hidradenitis suppurativa and current hormone-associated treatments.


Assuntos
Hidradenite Supurativa , Feminino , Hormônios Esteroides Gonadais , Hidradenite Supurativa/epidemiologia , Hidradenite Supurativa/etiologia , Humanos , Inflamação , Masculino , Qualidade de Vida , Pele
12.
Biol Reprod ; 103(6): 1300-1313, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32886743

RESUMO

Fibroblast growth factor 9 (FGF9) is an autocrine/paracrine growth factor that plays critical roles in embryonic and organ developments and is involved in diverse physiological events. Loss of function of FGF9 exhibits male-to-female sex reversal in the transgenic mouse model and gain of FGF9 copy number was found in human 46, XX sex reversal patient with disorders of sex development. These results suggested that FGF9 plays a vital role in male sex development. Nevertheless, how FGF9/Fgf9 expression is regulated during testis determination remains unclear. In this study, we demonstrated that human and mouse SRY bind to -833 to -821 of human FGF9 and -1010 to -998 of mouse Fgf9, respectively, and control FGF9/Fgf9 mRNA expression. Interestingly, we showed that mouse SRY cooperates with SF1 to regulate Fgf9 expression, whereas human SRY-mediated FGF9 expression is SF1 independent. Furthermore, using an ex vivo gonadal culture system, we showed that FGF9 expression is sufficient to switch cell fate from female to male sex development in 12-16 tail somite XX mouse gonads. Taken together, our findings provide evidence to support the SRY-dependent, fate-determining role of FGF9 in male sex development.


Assuntos
Transtornos do Desenvolvimento Sexual/genética , Fator 9 de Crescimento de Fibroblastos/metabolismo , Gônadas/fisiologia , Processos de Determinação Sexual/fisiologia , Proteína da Região Y Determinante do Sexo/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Fator 9 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína da Região Y Determinante do Sexo/genética , Técnicas de Cultura de Tecidos , Regulação para Cima
13.
J Biomed Sci ; 27(1): 63, 2020 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-32389123

RESUMO

Oxygen is essentially required by most eukaryotic organisms as a scavenger to remove harmful electron and hydrogen ions or as a critical substrate to ensure the proper execution of enzymatic reactions. All nucleated cells can sense oxygen concentration and respond to reduced oxygen availability (hypoxia). When oxygen delivery is disrupted or reduced, the organisms will develop numerous adaptive mechanisms to facilitate cells survived in the hypoxic condition. Normally, such hypoxic response will cease when oxygen level is restored. However, the situation becomes complicated if hypoxic stress persists (chronic hypoxia) or cyclic normoxia-hypoxia phenomenon occurs (intermittent hypoxia). A series of chain reaction-like gene expression cascade, termed hypoxia-mediated gene regulatory network, will be initiated under such prolonged or intermittent hypoxic conditions and subsequently leads to alteration of cellular function and/or behaviors. As a result, irreversible processes occur that may cause physiological disorder or even pathological consequences. A growing body of evidence implicates that hypoxia plays critical roles in the pathogenesis of major causes of mortality including cancer, myocardial ischemia, metabolic diseases, and chronic heart and kidney diseases, and in reproductive diseases such as preeclampsia and endometriosis. This review article will summarize current understandings regarding the molecular mechanism of hypoxia in these common and important diseases.


Assuntos
Endometriose/fisiopatologia , Cardiopatias/fisiopatologia , Hipóxia/fisiopatologia , Nefropatias/fisiopatologia , Doenças Metabólicas/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Neoplasias/fisiopatologia , Pré-Eclâmpsia/fisiopatologia , Doença Crônica , Endometriose/etiologia , Feminino , Cardiopatias/etiologia , Humanos , Hipóxia/complicações , Nefropatias/etiologia , Masculino , Doenças Metabólicas/etiologia , Isquemia Miocárdica/etiologia , Neoplasias/etiologia , Pré-Eclâmpsia/etiologia , Gravidez
14.
Nature ; 493(7431): 236-40, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23201680

RESUMO

Mutations in phosphatase and tensin homologue (PTEN) or genomic alterations in the phosphatidylinositol-3-OH kinase-signalling pathway are the most common genetic alterations reported in human prostate cancer. However, the precise mechanism underlying how indolent tumours with PTEN alterations acquire metastatic potential remains poorly understood. Recent studies suggest that upregulation of transforming growth factor (TGF)-ß signalling triggered by PTEN loss will form a growth barrier as a defence mechanism to constrain prostate cancer progression, underscoring that TGF-ß signalling might represent a pre-invasive checkpoint to prevent PTEN-mediated prostate tumorigenesis. Here we show that COUP transcription factor II (COUP-TFII, also known as NR2F2), a member of the nuclear receptor superfamily, serves as a key regulator to inhibit SMAD4-dependent transcription, and consequently overrides the TGF-ß-dependent checkpoint for PTEN-null indolent tumours. Overexpression of COUP-TFII in the mouse prostate epithelium cooperates with PTEN deletion to augment malignant progression and produce an aggressive metastasis-prone tumour. The functional counteraction between COUP-TFII and SMAD4 is reinforced by genetically engineered mouse models in which conditional loss of SMAD4 diminishes the inhibitory effects elicited by COUP-TFII ablation. The biological significance of COUP-TFII in prostate carcinogenesis is substantiated by patient sample analysis, in which COUP-TFII expression or activity is tightly correlated with tumour recurrence and disease progression, whereas it is inversely associated with TGF-ß signalling. These findings reveal that the destruction of the TGF-ß-dependent barrier by COUP-TFII is crucial for the progression of PTEN-mutant prostate cancer into a life-threatening disease, and supports COUP-TFII as a potential drug target for the intervention of metastatic human prostate cancer.


Assuntos
Fator II de Transcrição COUP/metabolismo , Transformação Celular Neoplásica , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Fator II de Transcrição COUP/deficiência , Fator II de Transcrição COUP/genética , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Deleção de Genes , Humanos , Masculino , Camundongos , Metástase Neoplásica , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Modelos de Riscos Proporcionais , Próstata/metabolismo , Próstata/patologia , Proteína Smad4/deficiência , Proteína Smad4/genética , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/metabolismo
15.
J Obstet Gynaecol Res ; 45(3): 532-541, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30618168

RESUMO

AIM: Summarize recent findings of how hypoxia regulates numerous important processes to facilitate the implantation, proliferation and progression of ectopic endometriotic lesions. METHODS: Most up-to-date evidences about how hypoxia contributes to the disease pathogenesis of endometriosis and potential therapeutic approaches were collected by conducting a comprehensive search of medical literature electronic databases. Quality of data was analyzed by experienced experts including gynecologist and basic scientists. RESULTS: Uterus is a highly vascularized organ, which makes endometrial cells constantly expose to high concentration of oxygen. When endometrial tissues shed off from the eutopic uterus and retrograde to the peritoneal cavity, they face severe hypoxic stress. Even with successful implantation to ovaries or peritoneum, the hypoxic stress remains as a critical issue because endometrial cells are used to live in the well-oxygenated environment. Under the hypoxia condition, cells undergo epigenetic modulation and evolve several survival processes including steroidogenesis, angiogenesis, inflammation and metabolic switch. The complex gene regulatory network driven by hypoxia ensures endometriotic cells can survive under the hostile peritoneal microenvironment. CONCLUSION: Hypoxia plays critical roles in promoting pathological processes to facilitate the development of endometriosis. Targeting hypoxia-mediated gene network represents an alternative approach for the treatment of endometriosis.


Assuntos
Endometriose/etiologia , Hipóxia/complicações , Neovascularização Patológica/etiologia , Endometriose/patologia , Endométrio/patologia , Feminino , Humanos , Hipóxia/patologia , Neovascularização Patológica/patologia
16.
Chin J Physiol ; 62(1): 2-10, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30942193

RESUMO

Osteoarthritis (OA) is a degenerative joint disorder and is the leading cause of disability of people, which negatively impact people's physical and mental health. Although OA causes great socioeconomic burden and individual suffering, no effective treatment options are provided so far. This is partially resulted from poor regenerative activity of articular cartilage and our incomplete understanding of the underlying mechanism of OA. Traditional drug therapies such as acetaminophen and opioids are effective in relieving pain but do not reverse cartilage damage and are often associated with adverse events. Therefore, it is necessary to find effective OA drugs. In recent years, novel regenerative therapies have received much attention because they can effectively promote tissue repair and regeneration. The fibroblast growth factor (FGF) signaling has been suggested to involve in cartilage homeostasis for decades. The current research shows that sprifermin/recombinant FGF18 significantly reduces the loss of cartilage thickness and volume without serious side effects, thus warrants a continued research for potential new medications of OA. This review mainly highlights the current research progress on FGFs and FGF receptors as a potential therapeutic target for OA.


Assuntos
Cartilagem Articular , Osteoartrite , Condrócitos , Fatores de Crescimento de Fibroblastos , Humanos , Transdução de Sinais
17.
Cell Physiol Biochem ; 48(2): 605-617, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30021209

RESUMO

BACKGROUND/AIMS: Huntington's disease (HD) is a heritable neurodegenerative disorder, and there is no cure for HD to date. A type of fibroblast growth factor (FGF), FGF9, has been reported to play prosurvival roles in other neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. However, the effects of FGF9 on HD is still unknown. With many similarities in the cellular and pathological mechanisms that eventually cause cell death in neurodegenerative diseases, we hypothesize that FGF9 might provide neuroprotective functions in HD. METHODS: In this study, STHdhQ7/Q7 (WT) and STHdhQ111/Q111 (HD) striatal knock-in cell lines were used to evaluate the neuroprotective effects of FGF9. Cell proliferation, cell death and neuroprotective markers were determined via the MTT assay, propidium iodide staining and Western blotting, respectively. The signaling pathways regulated by FGF9 were demonstrated using Western blotting. Additionally, HD transgenic mouse models were used to further confirm the neuroprotective effects of FGF9 via ELISA, Western blotting and immunostaining. RESULTS: Results show that FGF9 not only enhances cell proliferation, but also alleviates cell death as cells under starvation stress. In addition, FGF9 significantly upregulates glial cell line-derived neurotrophic factor (GDNF) and an anti-apoptotic marker, Bcl-xL, and decreases the expression level of an apoptotic marker, cleaved caspase 3. Furthermore, FGF9 functions through ERK, AKT and JNK pathways. Especially, ERK pathway plays a critical role to influence the effects of FGF9 toward cell survival and GDNF production. CONCLUSIONS: These results not only show the neuroprotective effects of FGF9, but also clarify the critical mechanisms in HD cells, further providing an insight for the therapeutic potential of FGF9 in HD.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 9 de Crescimento de Fibroblastos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Caspase 3/metabolismo , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Transgênicos , Nitrilas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Córtex Visual/citologia , Córtex Visual/efeitos dos fármacos , Córtex Visual/metabolismo , Proteína bcl-X/metabolismo
18.
Hum Reprod ; 33(8): 1517-1527, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29982401

RESUMO

STUDY QUESTION: How does hypoxia-mediated downregulation of chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) promote angiogenesis in endometriosis? SUMMARY ANSWER: Suppression of COUP-TFII by hypoxia stimulates angiogenesis through induction of angiogenin (ANG). WHAT IS KNOWN ALREADY: The level of COUP-TFII is downregulated in endometriotic tissues, and downregulation of COUP-TFII contributes to the development of endometriosis. STUDY DESIGN, SIZE, DURATION: Twenty-seven patients of reproductive age with endometriosis were recruited in this study. Eutopic endometrial and ectopic endometriotic stromal cells were isolated, cultured and subjected to various treatments. PARTICIPANTS/MATERIALS, SETTING, METHODS: Microarray hybridization, quantitative RT-PCR, and Western blot were used to detect gene expression in normal and endometriotic samples. A luciferase reporter assay and chromatin immunoprecipitation in normoxia- or hypoxia-treated primary cultures of human endometrial stromal cells were performed. Tube formation analysis was performed using primary human umbilical vein endothelial cells (HUVECs). MAIN RESULTS AND THE ROLE OF CHANCE: Protein level of COUP-TFII was downregulated by hypoxia (P < 0.05, normoxia versus hypoxia). Loss of COUP-TFII increased the angiogenic capacity of endometrial stromal cells (P < 0.05, COUP-TFII knockdown versus knockdown control). A novel COUP-TFII target gene, ANG, was identified through microarray analysis. Chromatin immunoprecipitation and promoter activity assays demonstrated that the ANG promoter was bound and suppressed by COUP-TFII (P < 0.05, COUP-TFII overexpression versus empty vector). The levels of ANG mRNA and protein were elevated in ectopic endometriotic stromal cells and negatively correlated with COUP-TFII (P < 0.05, endometrial versus endometriotic tissues/stromal cells). Both knockdown and forced-expression of COUP-TFII further demonstrated that ANG expression and ANG-mediated angiogenic activity were negatively regulated by COUP-TFII (P < 0.05, COUP-TFII knockdown versus knockdown control, and COUP-TFII overexpression versus empty vector). LIMITATIONS, REASONS FOR CAUTION: This study was conducted in primary human endometrial stromal cell cultures and HUVECs, therefore, may not fully reflect the situation in vivo. LARGE SCALE DATA: The raw data were submitted to Gene Expression Omnibus (GSE107469). WIDER IMPLICATIONS OF THE FINDINGS: This is the first study to highlight that the aberrant expression of ANG in endometriotic lesions is mediated by hypoxia-suppressed COUP-TFII expression, which reveals an as yet unidentified molecular pathogenesis of endometriosis. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by research grants (MOST 105-2314-B-006-059-MY3 to M.H.W. and MOST 104-2320-B-006-036-MY3 to S.J.T.) from the Ministry of Science and Technology, Taiwan. The authors declare that there is no conflict of interest.


Assuntos
Fator II de Transcrição COUP/metabolismo , Endometriose/metabolismo , Endométrio/irrigação sanguínea , Endométrio/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Patológica , Comunicação Parácrina , Ribonuclease Pancreático/metabolismo , Células Estromais/metabolismo , Fator II de Transcrição COUP/genética , Estudos de Casos e Controles , Hipóxia Celular , Células Cultivadas , Microambiente Celular , Endometriose/genética , Endometriose/patologia , Endométrio/patologia , Feminino , Humanos , Cultura Primária de Células , Ribonuclease Pancreático/genética , Transdução de Sinais , Células Estromais/patologia
19.
J Pathol ; 241(5): 638-648, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28026024

RESUMO

Dual-specificity phosphatase 2 (DUSP2) is a negative regulator of mitogen-activated protein kinases. Our previous study showed that DUSP2 expression is down-regulated in many human cancers and loss of DUSP2 promotes cancer progression; however, the underlying mechanism remains largely uncharacterized. Herein, we found that loss of DUSP2 induces angiogenesis, while forced expression of DUSP2 inhibits microvessel formation in xenografted mouse tumours. Genome-wide screening of expression profiles, and meta-analysis of clinical data, identified that the level of interleukin-8 (IL-8) correlated negatively with that of DUSP2, suggesting that it may be a downstream target of DUSP2. Molecular characterization revealed that DUSP2 inversely regulates IL-8 expression, mediated by ERK1/2 and C/EBPα-dependent transcriptional regulation. Further study showed that hypoxia-induced IL-8 expression in cancer cells is also mediated via down-regulation of DUSP2. Treatment with the IL-8 receptor inhibitor reparixin or knockdown of IL-8 in cancer cells abolished angiogenesis induced by loss of DUSP2. Functionally, knockdown of DUSP2 enhanced tumour growth and metastasis, which were abolished by treatment with reparixin or knockdown of IL-8 in an orthotopic mouse model. Taken together, our results demonstrate that hypoxia inhibits DUSP2 expression in colon cancer, leading to up-regulation of IL-8, which facilitates angiogenesis and tumour metastasis. Our findings suggest that blocking hypoxia-DUSP2-IL-8 signalling may be a plausible approach for therapeutic intervention in cancer. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neoplasias do Colo/enzimologia , Fosfatase 2 de Especificidade Dupla/genética , Interleucina-8/genética , Neovascularização Patológica/genética , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Colo/enzimologia , Colo/patologia , Neoplasias do Colo/irrigação sanguínea , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação para Baixo , Fosfatase 2 de Especificidade Dupla/metabolismo , Xenoenxertos , Humanos , Interleucina-8/metabolismo , Masculino , Camundongos , Camundongos SCID , Microvasos/enzimologia , Microvasos/patologia , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais , Regulação para Cima
20.
J Pathol ; 242(4): 476-487, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28608501

RESUMO

Endometriosis is a highly prevalent gynaecological disease that severely reduces women's health and quality of life. Ectopic endometriotic lesions have evolved mechanisms to survive in the hypoxic peritoneal microenvironment by regulating the expression of a significant subset of genes. However, the master regulator controlling these genes remains to be characterized. Herein, by using bioinformatics analysis and experimental verification, we identified yes-associated protein 1 (YAP1) as a master regulator of endometriosis. Nuclear localization and transcriptional activity of YAP1 were up-regulated by hypoxia via down-regulation of LATS1, a kinase that inactivates YAP1. Disruption of hypoxia-induced YAP1 signalling by siRNA knockdown or inhibitor treatment abolished critical biological processes involved in endometriosis development such as steroidogenesis, angiogenesis, inflammation, migration, innervation, and cell proliferation. Treatment with a YAP1 inhibitor caused the regression of endometriotic lesions without affecting maternal fertility or the growth rate of offspring in the mouse model of endometriosis. Taken together, we identify hypoxia/LATS1/YAP1 as a novel pathway for the pathogenesis of endometriosis and demonstrate that targeting YAP1 might be an alternative approach to treat endometriosis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Endometriose/tratamento farmacológico , Fertilidade/efeitos dos fármacos , Fosfoproteínas/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Proteínas de Ciclo Celular , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Biologia Computacional/métodos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Endometriose/etiologia , Endometriose/genética , Endometriose/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Estudo de Associação Genômica Ampla , Humanos , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular/métodos , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Porfirinas/farmacologia , Porfirinas/uso terapêutico , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Células Estromais/metabolismo , Fatores de Transcrição , Transcrição Gênica , Verteporfina , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA