Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Science ; 384(6701): eadk5382, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38870290

ABSTRACT

Polycystic ovary syndrome (PCOS), a prevalent reproductive disorder in women of reproductive age, features androgen excess, ovulatory dysfunction, and polycystic ovaries. Despite its high prevalence, specific pharmacologic intervention for PCOS is challenging. In this study, we identified artemisinins as anti-PCOS agents. Our finding demonstrated the efficacy of artemisinin derivatives in alleviating PCOS symptoms in both rodent models and human patients, curbing hyperandrogenemia through suppression of ovarian androgen synthesis. Artemisinins promoted cytochrome P450 family 11 subfamily A member 1 (CYP11A1) protein degradation to block androgen overproduction. Mechanistically, artemisinins directly targeted lon peptidase 1 (LONP1), enhanced LONP1-CYP11A1 interaction, and facilitated LONP1-catalyzed CYP11A1 degradation. Overexpression of LONP1 replicated the androgen-lowering effect of artemisinins. Our data suggest that artemisinin application is a promising approach for treating PCOS and highlight the crucial role of the LONP1-CYP11A1 interaction in controlling hyperandrogenism and PCOS occurrence.


Subject(s)
ATP-Dependent Proteases , Artemisinins , Cholesterol Side-Chain Cleavage Enzyme , Mitochondrial Proteins , Polycystic Ovary Syndrome , Animals , Female , Humans , Mice , Rats , Androgens/metabolism , Artemisinins/therapeutic use , Artemisinins/pharmacology , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Cholesterol Side-Chain Cleavage Enzyme/genetics , Disease Models, Animal , Hyperandrogenism/drug therapy , Hyperandrogenism/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Ovary/drug effects , Ovary/metabolism , Polycystic Ovary Syndrome/drug therapy , Proteolysis , Mice, Inbred C57BL , Young Adult , Adult , Rats, Sprague-Dawley , ATP-Dependent Proteases/genetics , ATP-Dependent Proteases/metabolism
2.
Protein Cell ; 15(7): 512-529, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38167949

ABSTRACT

Polycystic ovary syndrome (PCOS) is the leading cause of anovulatory infertility. Inadequate understanding of the ovulation drivers hinders PCOS intervention. Herein, we report that follicle stimulating hormone (FSH) controls follicular fluid (FF) glutamine levels to determine ovulation. Murine ovulation starts from FF-exposing granulosa cell (GC) apoptosis. FF glutamine, which decreases in pre-ovulation porcine FF, elevates in PCOS patients FF. High-glutamine chow to elevate FF glutamine inhibits mouse GC apoptosis and induces hormonal, metabolic, and morphologic PCOS traits. Mechanistically, follicle-development-driving FSH promotes GC glutamine synthesis to elevate FF glutamine, which maintain follicle wall integrity by inhibiting GC apoptosis through inactivating ASK1-JNK apoptotic pathway. FSH and glutamine inhibit the rapture of cultured murine follicles. Glutamine removal or ASK1-JNK pathway activation with metformin or AT-101 reversed PCOS traits in PCOS models that are induced with either glutamine or EsR1-KO. These suggest that glutamine, FSH, and ASK1-JNK pathway are targetable to alleviate PCOS.


Subject(s)
Follicle Stimulating Hormone , Glutamine , Granulosa Cells , Ovulation , Polycystic Ovary Syndrome , Animals , Female , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Glutamine/metabolism , Mice , Follicle Stimulating Hormone/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Humans , Apoptosis/drug effects , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Kinase Kinase 5/genetics , Swine , Mice, Inbred C57BL
3.
Biochem Biophys Res Commun ; 681: 225-231, 2023 11 12.
Article in English | MEDLINE | ID: mdl-37783121

ABSTRACT

The commitment of mesenchymal stem cells (MSCs) to preadipocytes and the termination of differentiation to adipocytes are critical for maintaining systemic energy homeostasis. However, our knowledge of the molecular mechanisms governing the commitment of MSCs to preadipocytes and the subsequent termination of their differentiation into adipocytes remain limited. Additionally, the role of Sox6 sex-determining region Y (SRY)-box6 (Sox6), a transcription factor that regulates gene transcription, is reportedly involved in various cellular processes, including adipogenesis; however, its function in regulating preadipocyte development and the factors involved in the termination of adipogenic differentiation remain unexplored. Therefore, we investigated the role of Sox6 in regulating the differentiation of adipocytes by monitoring the effects of its overexpression in C3H10T1/2 cells (in vitro) and C57BL/6J mouse (in vivo) models of adipogenesis. We observed lower Sox6 expression in the adipose tissue of obese mice than that in control mice. Sox6 overexpression inhibited the differentiation of MSC by directly binding to the lysyl oxidase (Lox) and preadipocyte factor 1 (Pref1) promoters, which was potentiated by histone deacetylase-1(HDAC1). Our findings suggest that Sox6 is a key regulator of MSC commitment to adipocytes; therefore, targeting the Sox6-mediated regulation of this process could offer potential therapeutic avenues for addressing obesity and related metabolic disorders.


Subject(s)
Adipogenesis , Mesenchymal Stem Cells , Animals , Mice , Adipogenesis/genetics , Cell Differentiation/genetics , Mice, Inbred C57BL , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism
4.
Transl Cancer Res ; 11(8): 2636-2646, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36093536

ABSTRACT

Background: Circulating tumor cells (CTCs) have considered to be promising liquid biopsy in cancer due to the intact information of whole cells and the potential to reflect micrometastasis. However, CTCs research are extremely limited in ovarian cancer, probably due to their rarity. The predictive value of CTCs and circulating tumor microemboli (CTM) in metastasis remains to be elucidated in ovarian cancer. This study tried to identify CTCs/CTM in ovarian cancer with considerably positive rate. To preliminarily identify the invasive capacity of CTCs/CTM, the epithelial-mesenchymal transition (EMT) patterns of CTCs/CTM was evaluated. Moreover, for comprehensive understanding of invasiveness of disseminated cells in ovarian cancer, EMT pattern of exfoliated tumor cells in ascites were also confirmed in this study. Methods: Peripheral blood samples and ascites samples were collected from 22 ovarian cancer patients. The Microfiltration combined with morphological analysis was used to detect CTC single cells or cell clusters. Microfiltration combined with morphological analysis was applied in the CTC isolation and identification. EMT was evaluated by immunofluorescence via markers including vimentin and cytokeratin. Results: Microfiltration combined with morphological analysis was introduced to detect CTCs/CTM with a positivity rate of 40.9% in ovarian cancer patients. The number of CTC varied from 1 to 8, with CTM number from 4 to 30. CTCs/CTM of all samples have experienced EMT process. Vimentin was expressed in all CTC samples and all tumor cells in ascites, while cytokeratin was expressed in 44.4% (4/9) of CTC samples. There were no significant differences of the clinical parameters between the CTC-positive and CTC-negative patients. Conclusions: This study showed that both CTCs/CTM and detached tumor cells in ascites might have undergone complete or partial EMT in ovarian cancer. Moreover, microfiltration combined with cytomorphological analysis showed a considerable CTC detection rate.

5.
Ann Transl Med ; 9(9): 788, 2021 May.
Article in English | MEDLINE | ID: mdl-34268401

ABSTRACT

BACKGROUND: CA125 is the most widely used serum marker for preoperative diagnosis of ovarian cancer. However, CA125 elevation is not specific to ovarian cancer. More than 60% of patients who have elevated CA125 levels do not have ovarian cancer. To overcome the low specificity of CA125, we identified a CA125 glycoform that was specifically elevated in ovarian cancer and that may help in the further triage of patients with elevated serum CA125 levels. METHODS: We used antibody-lectin ELISA to detect various CA125 glycoforms. Among 21 lectins tested, VVA, a plant lectin that preferentially binds Tn antigen, showed significantly stronger binding with ovarian cancer-derived CA125 than benign condition-derived CA125. CA125-Tn levels were tested among patients with elevated CA125 levels (n=328, including 68 ovarian cancer, 15 ovarian borderline tumors, and 245 benign conditions). The efficacy of CA125-Tn in diagnosing ovarian cancer was evaluated using ROC analysis. RESULTS: Medians and 25th to 75th quartiles of CA125-Tn levels were 0.31 (0.18-0.65) in ovarian cancer, 0.07 (0.02-0.12) in ovarian borderline tumor, and 0.07 (0.01-0.12) in benign conditions. AUC of the ROC curve was 0.890 (95% CI: 0.845, 0.935) for CA125-Tn to discriminate ovarian cancer cases from nonmalignant cases (borderline tumors and benign conditions). Its performance was even better among patients older than 45 y (AUC: 0.905, 95% CI: 0.841, 0.969). Specificity was improved from 35.1% for CA125 to 75.7% for CA125-Tn among patients older than 45 y when sensitivity was fixed at 90%. CONCLUSIONS: CA125-Tn ELISA assay can improve specificity of the preoperative diagnosis of ovarian cancer and serve as a further triage strategy for patients with elevated CA125 levels.

6.
Cancer Res Treat ; 52(3): 815-829, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32138466

ABSTRACT

PURPOSE: While numerous epidemiological studies have indicated that omega-3 polyunsaturated fatty acids have anticancer properties in various cancers, the effects and mechanisms of eicosapentaenoic acid (EPA) in ovarian cancer cell growth are poorly understood. MATERIALS AND METHODS: ES2 ovarian clear cell carcinoma cells and SKOV3 adenocarcinoma cells were treated with palmitic acid or EPA, followed by flow cytometry and cell counting to measure apoptosis and proliferation, respectively. A modified protein lipid overlay assay was used to further verify whether EPA was a ligand of G protein-coupled receptor 30 (GPR30) in ES2 cells. The levels of apoptosis-related genes, phosphorylated AKT, and phosphorylated ERK1/2 were detected to explore the underlying mechanism. Finally, inhibitory effect of EPA on tumor growth via GPR30 was determined in vitro and in vivo. RESULTS: EPA suppressed ES2 ovarian clear cell carcinoma cells growth via GPR30, a novel EPA receptor, by inducing apoptosis. As a ligand of GPR30, EPA activated the GPR30-cAMP- protein kinase A signaling pathway. When GPR30 was suppressed by siRNA or its inhibitor G15, the antiproliferative action of EPA was impaired. Furthermore, EPA inhibited tumor growth by blocking the activation of AKT and ERK. In the mouse xenograft model, EPA decreased tumor volume and weight through GPR30 by blocking tumor cell proliferation. CONCLUSION: These results confirm that EPA is a tumor suppressor in human ovarian clear cell carcinoma cells and functions through a novel fatty acid receptor, GPR30, indicating a mechanistic linkage between omega-3 fatty acids and cancers.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Biomarkers, Tumor/metabolism , Eicosapentaenoic Acid/pharmacology , Ovarian Neoplasms/drug therapy , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenocarcinoma, Clear Cell/metabolism , Adenocarcinoma, Clear Cell/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Cycle , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Reprod Sci ; 26(4): 532-542, 2019 04.
Article in English | MEDLINE | ID: mdl-29909746

ABSTRACT

BACKGROUND: Decidual γδ T cells are known to regulate the function of trophoblasts at the maternal-fetal interface; however, little is known about the molecular mechanisms of cross talk between trophoblast cells and decidual γδ T cells. METHODS: Expression of chemokine C-X-C motif ligand 6 (CXCL16) and its receptor CXCR6 was evaluated in first-trimester human villus and decidual tissues by immunohistochemistry. γδ T cells were isolated from first-trimester human deciduae and cocultured with JEG3 trophoblast cells. Cell proliferation and apoptosis-related molecules, together with cytotoxicity factor and cytokine production, were measured by flow cytometry analysis. RESULTS: Expression of CXCL16 and CXCR6 was reduced at the maternal-fetal interface in patients who experienced unexplained recurrent spontaneous abortion as compared to healthy pregnancy women. With the administration of pregnancy-related hormones or coculture with JEG3 cells, CXCR6 expression was upregulated on decidual γδ T cells. CXCL16 derived from JEG3 cells caused a decrease in granzyme B production of decidual γδ T cells. In addition, decidual γδ T cells educated by JEG3-derived CXCL16 upregulated the expression of Bcl-xL in JEG3 cells. CONCLUSION: This study suggested that the CXCL16/CXCR6 axis may contribute to maintaining normal pregnancy by reducing the secretion of cytotoxic factor granzyme B of decidual γδ T cells and promoting the expression of antiapoptotic marker Bcl-xL of trophoblasts.


Subject(s)
Chemokine CXCL16/metabolism , Decidua/metabolism , Granzymes/metabolism , Trophoblasts/metabolism , bcl-X Protein/metabolism , Cell Survival , Cells, Cultured , Chorionic Villi/metabolism , Female , Humans , Pregnancy , Pregnancy Trimester, First , Receptors, CXCR6/metabolism
8.
J Cell Physiol ; 234(5): 6361-6370, 2019 05.
Article in English | MEDLINE | ID: mdl-30229902

ABSTRACT

Patients with ovarian cancer frequently develop acquired drug resistance after the long-term chemotherapy, leading to disease progression. Enhanced epithelial-mesenchymal transition (EMT) has been implicated in chemoresistance of ovarian cancer cells; however, the molecular mechanisms involved are largely undefined. Pyruvate dehydrogenase kinase 1 (PDK1), a key regulatory enzyme in glucose metabolism, has been recognized as a gatekeeper of the Warburg effect, a hallmark of cancer. In this study, the function of PDK1 in cisplatin resistance of ovarian cancer in terms of growth and EMT was investigated. PDK1 was upregulated in cisplatin-resistant ovarian cancer cells. PDK1 knockdown in resistant cells led to increased sensitivity to cisplatin-induced cell death and apoptosis. PDK1 downregulation also reversed the EMT and cell motility in cisplatin-resistant cells. In a mouse xenograft model, tumors derived from PDK1-silenced ovarian cancer cells exhibited decreased tumor growth and EMT compared with control after the cisplatin treatment. Mechanistically, PDK1 overexpression led to increased phosphorylation of EGFR, and blocking EGFR kinase activity by erlotinib reversed cisplatin resistance induced by PDK1 overexpression. Furthermore, in patients with ovarian cancer, higher PDK1 and p-EGFR levels were associated with chemoresistance. These results supported that PDK1 contributes to chemoresistance of ovarian cancer by activating EGFR. Therefore, PDK1 may serve as a promising target to combat chemoresistance of ovarian cancer.


Subject(s)
Drug Resistance, Neoplasm/physiology , Ovarian Neoplasms/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Epithelial-Mesenchymal Transition/physiology , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Drug Deliv ; 25(1): 797-806, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29542355

ABSTRACT

Ovarian cancer is the leading cause of cancer death among gynecological malignancies. The high mortality rate has not been significantly reduced despite advances in surgery and chemotherapy. Gene therapy shows therapeutic potential, but several key issues must be resolved before clinical application. To minimize toxicity in noncancerous tissues, tumor-specific ligands are conjugated to vectors to increase the selectivity of drug delivery. The expression pattern of follicle-stimulating hormone (FSH) receptor in normal and cancer tissues provides an opportunity for highly selective drug delivery in ovarian cancer. Furthermore, tumor-specific promoters can conditionally regulate therapeutic gene expression in tumor or normal tissues. The mucin 16 (MUC16) promoter might be a potential tool to drive ovarian cancer-localized gene expression since MUC16/CA125 is overexpressed in most ovarian carcinomas. Here, we screened the possible MUC16 promoter sequences and constructed MUC16 promoter-driven gro-α shRNA plasmid vectors. The vectors were specifically delivered into ovarian cancer cells via FSH peptide-conjugated nanoparticles. The predicted promoter sequence with TAAA repeats showed high transcriptional activity. The nanoparticle complex containing MUC16 promoter-driven gro-α shRNA and FSH peptides had the ability to decrease gro-α protein secretion in ovarian cancer cells and block tumor growth without obvious toxic effects in a nude mouse model bearing ovarian cancer. Our study provides a novel gene delivery system using a MUC16 promoter trigger and FSH peptide-mediated active targeting in ovarian cancer, and this system may be a promising strategy for specific genetic therapeutic delivery.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/administration & dosage , Membrane Proteins/antagonists & inhibitors , Nanoparticles/chemistry , Ovarian Neoplasms/therapy , Promoter Regions, Genetic , RNA, Small Interfering/administration & dosage , RNAi Therapeutics/methods , Animals , CA-125 Antigen/genetics , CA-125 Antigen/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma/therapy , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chemokine CXCL1/antagonists & inhibitors , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Computational Biology , Female , Follicle Stimulating Hormone, beta Subunit/chemistry , Follicle Stimulating Hormone, beta Subunit/metabolism , Follicle Stimulating Hormone, beta Subunit/therapeutic use , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Neoplasm Invasiveness/prevention & control , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Particle Size , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Surface Properties , Tumor Burden
10.
Drug Deliv ; 25(1): 576-584, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29461120

ABSTRACT

The distinct hormone molecules and receptors, such as follicle-stimulating hormone receptor (FSHR) in ovarian cancer, provide opportunities for more precisely targeted therapy. We previously developed FSHR-mediated nanoparticles and found that FSH peptides on the surface of nanoparticles improved the delivery of short interfering RNA (siRNA) into ovarian cancer cells. However, the high toxicity of the nanoparticles and the transient silencing of the siRNA in vivo limited further study. Here, we developed FSH peptide-conjugated nanoparticles with an increased amount of polyethylene glycol (PEG) grafting and encapsulated short hairpin RNA (shRNA) to silence the target gene, growth-regulated oncogene α (gro-α). The nanoparticle complexes exhibited good stability over three weeks. Expression of the target gene, gro-α, was significantly down-regulated by gro-α shRNA-loaded nanoparticles conjugated with FSH peptides (FSH33-G-NP) in FSHR-positive HEY cells. Cell proliferation, migration, and invasion were also inhibited by FSH33-G-NP. Tumor growth was delayed significantly in the mice treated with FSH33-G-NP. No significant loss of body weight or severe toxic effects were observed in any groups. In conclusion, gro-α shRNA-loaded nanoparticles conjugated with FSH peptides overcame the drawbacks of the in vivo application of RNAi therapeutics and polymer-based nanocarriers and showed safe antitumor efficacy. Our study might contribute to the application of FSHR-based targeted therapy and imaging in cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Chemokine CXCL1/administration & dosage , Follicle Stimulating Hormone/administration & dosage , Nanoparticles/administration & dosage , Ovarian Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , Animals , Cell Line, Tumor , Chemokine CXCL1/biosynthesis , Chemokine CXCL1/genetics , Female , Gene Silencing/drug effects , Gene Silencing/physiology , Gene Transfer Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
11.
Int J Biochem Cell Biol ; 95: 9-16, 2018 02.
Article in English | MEDLINE | ID: mdl-29247711

ABSTRACT

The high mortality rate of ovarian cancer is connected with the development of acquired resistance to multiple cancer drugs, especially cisplatin. Activation of cytoprotective autophagy has been implicated as a contributing mechanism for acquired cisplatin resistance in ovarian cancer cells. Hexokinase 2 (HK2) phosphorylates glucose to generate glucose-6-phosphate, the rate-limiting step in glycolysis. Higher HK2 expression has been associated with chemoresistance in ovarian cancer. However, whether HK2 functionally contributes to cisplatin resistance in ovarian cancer is unclear. In this study, we investigated the role of HK2 in regulating ovarian cancer cisplatin resistance. Increased HK2 levels were detected in drug-resistant human ovarian cancer cells and tissues. Cisplatin downregulated HK2 in cisplatin-sensitive but not in resistant ovarian cancer cells. HK2 knockdown sensitized resistant ovarian cancer cells to cisplatin-induced cell death and apoptosis. Conversely, HK2 overexpression in cisplatin-sensitive cells induced cisplatin resistance. Mechanistically, cisplatin increased ERK1/2 phosphorylation as well as autophagic activity. Blocking autophagy with the autophagy inhibitor 3-MA sensitized resistant ovarian cancer cells to cisplatin. HK2 overexpression enhanced cisplatin-induced ERK1/2 phosphorylation and autophagy while HK2 knockdown showed the opposite effects. Blocking the MEK/ERK pathway using the MEK inhibitor U0126 prevented cisplatin-induced autophagy enhanced by HK2 overexpression. Furthermore, HK2 knockdown sensitized resistance ovarian tumor xenografts to cisplatin in vivo. In conclusion, our data supported that HK2 promotes cisplatin resistance in ovarian cancer by enhancing drug-induced, ERK-mediated autophagy. Therefore, targeting HK2 may be a new therapeutic strategy to combat chemoresistance in ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Hexokinase/metabolism , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Hexokinase/antagonists & inhibitors , Hexokinase/genetics , Humans , MAP Kinase Signaling System/drug effects , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/ultrastructure , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Ovary/ultrastructure , Protein Kinase Inhibitors/pharmacology , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
J Huazhong Univ Sci Technolog Med Sci ; 37(6): 928-932, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29270755

ABSTRACT

To investigate the clinical course and management of congenital vaginal atresia. This retro-spective analysis included patients with congenital vaginal atresia treated from March 2004 to August 2014 at the Obstetrics and Gynecology Hospital of Fudan University. Thirty-nine patients were included in this study. Their average age was 16.87±2.2 years when they came to our hospital. Totally, 51% of the patients had isolated congenital vaginal atresia with a normal cervix, whereas the others had either cervical atresia or imperforate hymen. The primary presenting signs and symptoms included primary amenorrhea (71.8%), periodic abdominalgia (41.0%), abdominal pain (36.0%), dyspareunia (10.3%), menstrual disorders (5.1%), and pelvic mass (5.1%). Ultrasound and magnetic resonance imaging (MRI) were effective inspection methods for the screening of urogenital tract-associated anomalies. Vagi-noplasty mainly included simple vagina reconstruction with insertion of a mold (n=22) and split-thickness skin grafting (n=4). In 64% of surgical patients, normal menstrual bleeding was achieved. Four of the patients subsequently became pregnant and delivered at term. Primary amenorrhea, periodic abdominalgia and abdominal pain are the main reasons for the post pubertal patients to visit doctors. Surgical methods can successfully provide these patients an opportunity for subsequent conservative management, can result in normal menstrual bleeding, resolve cyclic pelvic pain, and provide some po-tential for fertility.


Subject(s)
Abdominal Pain/surgery , Amenorrhea/surgery , Dyspareunia/surgery , Hymen/abnormalities , Menstruation Disturbances/surgery , Plastic Surgery Procedures/methods , Vagina/surgery , Abdominal Pain/physiopathology , Abdominal Pain/rehabilitation , Adolescent , Amenorrhea/physiopathology , Amenorrhea/rehabilitation , Congenital Abnormalities , Dyspareunia/physiopathology , Dyspareunia/rehabilitation , Female , Fertilization/physiology , Humans , Hymen/surgery , Menstruation/physiology , Menstruation Disturbances/rehabilitation , Recovery of Function , Vagina/abnormalities , Young Adult
13.
Oncotarget ; 8(46): 81558-81571, 2017 Oct 06.
Article in English | MEDLINE | ID: mdl-29113414

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) endows epithelial cells with enhanced motility and invasiveness, allowing them to participate in many physiological and pathological processes. Epithelial-to-mesenchymal transition contributes to the generation of circulating tumor cells (CTCs) in epithelial cancers because it increases tumor cell invasiveness, promotes tumor cell intravasation and ensures tumor cell survival in the peripheral system. Although the contribution of epithelial-to-mesenchymal transition to tumor cell invasiveness has been confirmed, the role epithelial-to-mesenchymal transition plays in metastasis remains debated. As a favorable material for a "liquid biopsy", circulating tumor cells have been shown to have promising values in the clinical management of tumors. Furthermore, an increasing number of studies have begun to explore the value of CTC-related biomarkers, and some studies have found that the expression of EMT and stemness markers in circulating tumor cells, in addition to CTC detection, can provide more information on tumor diagnosis, treatment, prognosis and research.

14.
J Biol Chem ; 292(28): 11740-11750, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28572510

ABSTRACT

Polycystic ovary syndrome is a common endocrine disorder and a major cause of anovulatory sterility in women at reproductive age. Most patients with polycystic ovary syndrome have hyperandrogenism, caused by excess androgen synthesis. Bone morphogenetic protein 4 (BMP4) is an essential regulator of embryonic development and organ formation, and recent studies have also shown that BMP4 may be involved in female steroidogenesis process. However, the effect of BMP4 on hyperandrogenism remains unknown. Here, using a female mouse model of hyperandrogenism, we found that ovarian BMP4 levels were significantly decreased in hyperandrogenism. Elevated androgens inhibited BMP4 expression via activation of androgen receptors. Moreover, BMP4 treatment suppressed androgen synthesis in theca cells and promoted estrogen production in granulosa cells by regulating the expression of steroidogenic enzymes, including CYP11A, HSD3B2, CYP17A1, and CYP19A1 Consistently, knockdown of BMP4 augmented androgen levels and inhibited estrogen levels. Mechanistically, Smad signaling rather than the p38 MAPK pathway regulated androgen and estrogen formation, thereby mediating the effect of BMP4. Of note, BMP4-transgenic mice were protected against hyperandrogenism. Our observations clarify a vital role of BMP4 in controlling sex hormone levels and offer new insights into intervention for managing hyperandrogenism by targeting the BMP4-Smad signaling pathway.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Disease Models, Animal , Hyperandrogenism/etiology , Ovary/metabolism , Polycystic Ovary Syndrome/physiopathology , Signal Transduction , Smad4 Protein/metabolism , Androgens/metabolism , Androgens/pharmacology , Animals , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/genetics , Cells, Cultured , Dehydroepiandrosterone , Down-Regulation/drug effects , Estrogens/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Granulosa Cells/pathology , Mice, Inbred C57BL , Mice, Transgenic , Ovary/drug effects , Ovary/pathology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , RNA Interference , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Smad4 Protein/antagonists & inhibitors , Smad4 Protein/genetics , Theca Cells/drug effects , Theca Cells/metabolism , Theca Cells/pathology
15.
Sci Rep ; 7: 44744, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28303973

ABSTRACT

Cervical cancer is an infectious cancer and the most common gynecologic cancer worldwide. E6/E7, the early genes of the high-risk mucosal human papillomavirus type, play key roles in the carcinogenic process of cervical cancer. However, little was known about its roles in modulating tumor microenvironment, particular extracellular matrix (ECM). In this study, we found that E6/E7 could regulate multiple ECM proteins, especially collagen triple helix repeat containing 1 (CTHRC1). CTHRC1 is highly expressed in cervical cancer tissue and serum and closely correlated with clinicopathological parameters. CTHRC1 promotes cervical cancer cell migration and invasion in vitro and metastasis in vivo. E6/E7 regulates the expression of CTHRC1 in cervical cancer by E6/E7-p53-POU2F1 (POU class 2 homeobox 1) axis. Futhermore, CTHRC1 activates Wnt/PCP signaling pathway. Take together, E6/E7-p53-POU2F1-CTHRC1 axis promotes cervical cancer cell invasion and metastasis and may act as a potential therapeutic target for interventions against cervical cancer invasion and metastasis.


Subject(s)
Cell Polarity , Extracellular Matrix Proteins/metabolism , Octamer Transcription Factor-1/metabolism , Oncogene Proteins, Viral/metabolism , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Wnt Signaling Pathway , Cell Movement/genetics , Cell Proliferation/genetics , Extracellular Matrix Proteins/blood , Extracellular Matrix Proteins/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Middle Aged , Neoplasm Metastasis , ROC Curve , Up-Regulation/genetics , Uterine Cervical Neoplasms/blood , Uterine Cervical Neoplasms/genetics
16.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-333402

ABSTRACT

To investigate the clinical course and management of congenital vaginal atresia.This retrospective analysis included patients with congenital vaginal atresia treated from March 2004 to August 2014 at the Obstetrics and Gynecology Hospital of Fudan University.Thirty-nine patients were included in this study.Their average age was 16.87±2.2 years when they came to our hospital.Totally,51% of the patients had isolated congenital vaginal atresia with a normal cervix,whereas the others had either cervical atresia or imperforate hymen.The primary presenting signs and symptoms included primary amenorrhea (71.8%),periodic abdominalgia (41.0%),abdominal pain (36.0%),dyspareunia (10.3%),menstrual disorders (5.1%),and pelvic mass (5.1%).Ultrasound and magnetic resonance imaging (MRI) were effective inspection methods for the screening of urogenital tract-associated anomalies.Vaginoplasty mainly included simple vagina reconstruction with insertion of a mold (n=22) and split-thickness skin grafting (n=4).In 64% of surgical patients,normal menstrual bleeding was achieved.Four of the patients subsequently became pregnant and delivered at term.Primary amenorrhea,periodic abdominalgia and abdominal pain are the main reasons for the post pubertal patients to visit doctors.Surgical methods can successfully provide these patients an opportunity for subsequent conservative management,can result in normal menstrual bleeding,resolve cyclic pelvic pain,and provide some potential for fertility.

17.
Sci Rep ; 6: 34686, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708433

ABSTRACT

To enable rational multi-epitope vaccine and diagnostic antigen design, it is imperative to delineate complete IgG-epitome of the protein. Here, we describe results of IgG-epitome decoding of three proteins from high-risk (HR-) oncogenic human papillomavirus type 58 (HPV58). To reveal their entire epitomes, employing peptide biosynthetic approach, 30 precise linear B-cell epitopes (BCEs) were mapped on E6, E7 and L1 proteins using rabbits antisera to the respective recombinant proteins. Using sequence alignment based on BCE minimal motif, the specificity and conservativeness of each mapped BCE were delineated mainly among known HR-HPVs, including finding 3 broadly antibody cross-reactive BCEs of L1 that each covers almost all HR-HPVs. Western blots revealed that 13 of the 18 BCEs within L1-epitome were recognized by murine antisera to HPV58 virus-like particles, suggesting that these are antibody accessible BCEs. Also, a highly conserved epitope (YGD/XTL) of E6 was found to exist only in known common HR-HPVs, which could be used as the first peptide reference marker for judging HR-HPVs. Altogether, this study provides systemic and exhaustive information on linear BCEs of HR-HPV58 that will facilitate development of novel multi-epitope diagnostic reagents/chips for testing viral antibodies and 'universal' preventive HPV peptide vaccine based on L1 conserved BCEs.


Subject(s)
Capsid Proteins/chemistry , Epitope Mapping/methods , Immunoglobulin G/blood , Oncogene Proteins, Viral/chemistry , Papillomaviridae/metabolism , Papillomavirus E7 Proteins/chemistry , Animals , Binding Sites , Capsid Proteins/immunology , Epitopes, B-Lymphocyte/analysis , Humans , Mice , Models, Molecular , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/immunology , Protein Conformation , Rabbits
18.
EBioMedicine ; 11: 91-100, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27522322

ABSTRACT

Similar to estrogens, bone morphogenetic protein 4 (BMP4) promotes the accumulation of more metabolically active subcutaneous fat and reduction of visceral fat. However, whether there is a cross-talk between BMP4 and estrogen signaling remained unknown. Herein, we found that BMP4 deficiency in white adipose tissue (WAT) increased the estrogen receptor α (ERα) level and its signaling, which prevented adult female mice from developing high fat diet (HFD)-induced obesity and insulin resistance; estrogens depletion up regulated BMP4 expression to overcome overt adiposity and impaired insulin sensitivity with aging, and failure of BMP4 regulation due to genetic knockout led to more fat gain in aged female mice. This mutual regulation between BMP4 and estrogen/ERα signaling may also happen in adipose tissue of women, since the BMP4 level significantly increased after menopause, and was inversely correlated with body mass index (BMI). These findings suggest a counterbalance between BMP4 and estrogen/ERα signaling in the regulation of adiposity and relative metabolism in females.


Subject(s)
Adiposity , Bone Morphogenetic Protein 4/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Glucose/metabolism , Signal Transduction , Adipocytes/metabolism , Adipose Tissue/metabolism , Adiposity/genetics , Age Factors , Animals , Body Mass Index , Bone Morphogenetic Protein 4/genetics , Cell Line , Diet, High-Fat , Estrogens/pharmacology , Female , Gene Expression Regulation , Humans , Insulin Resistance , Metabolic Diseases/metabolism , Mice , Mice, Knockout , Models, Animal , Obesity/metabolism , Protein Binding , Protein Stability
19.
J Mol Cell Biol ; 8(4): 302-12, 2016 08.
Article in English | MEDLINE | ID: mdl-27030507

ABSTRACT

The expansion of subcutaneous (SC) white adipose tissue (WAT) has beneficial effects on metabolic health. Our previous work showed an increased number of bone morphogenetic protein 4 (BMP4)-activated beige adipocytes in SC WAT, indicating a potential role of BMP4 in adipocyte recruitment. It was also demonstrated that BMP4 committed multipotent mesodermal C3H10T1/2 stem cells to the adipocyte lineage ex vivo However, the mechanism by which BMP4 regulates adipogenesis in vivo has not been clarified. In this study, we found that BMP4 stimulated de novo adipogenesis in SC WAT concomitant with enhanced blood vessel formation, thus promoting adipose tissue angiogenesis. Platelet-derived growth factor receptor-ß-positive (PDGFRß(+)) multipotent stem cells within the neoangiogenic vessels were found to be adipocyte progenitors. Moreover, BMP4 downregulated PDGFRß by stimulating the lysosome-dependent degradation, which efficiently initiated adipogenic differentiation. These results suggest how BMP4 regulates adipocyte recruitment in SC WAT, and thus promote its beneficial metabolic effects.


Subject(s)
Adipogenesis , Bone Morphogenetic Protein 4/metabolism , Neovascularization, Physiologic , Subcutaneous Fat/growth & development , Subcutaneous Fat/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Blood Vessels/metabolism , Cell Differentiation , Cytokines/metabolism , Down-Regulation , Inguinal Canal/anatomy & histology , Lysosomes/metabolism , Mice, Knockout , Pericytes/metabolism , Proteolysis , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stem Cells/metabolism , Up-Regulation
20.
Chin Med J (Engl) ; 128(24): 3363-70, 2015 Dec 20.
Article in English | MEDLINE | ID: mdl-26668153

ABSTRACT

OBJECTIVE: This review aimed to update the progress of microRNA (miRNA) in early detection of ovarian cancer. We discussed the current clinical diagnosis methods and biomarkers of ovarian cancer, especially the methods of miRNA in early detection of ovarian cancer. DATA SOURCES: We collected all relevant studies about miRNA and ovarian cancer in PubMed and CNKI from 1995 to 2015. STUDY SELECTION: We included all relevant studies concerning miRNA in early detection of ovarian cancer, and excluded the duplicated articles. RESULTS: miRNAs play a key role in various biological processes of ovarian cancer, such as development, proliferation, differentiation, apoptosis and metastasis, and these phenomena appear in the early-stage. Therefore, miRNA can be used as a new biomarker for early diagnosis of ovarian cancer, intervention on miRNA expression of known target genes, and potential target genes can achieve the effect of early prevention. With the development of nanoscience and technology, analysis methods of miRNA are also quickly developed, which may provide better characterization of early detection of ovarian cancer. CONCLUSIONS: In the near future, miRNA therapy could be a powerful tool for ovarian cancer prevention and treatment, and combining with the new analysis technology and new nanomaterials, point-of-care tests for miRNA with high throughput, high sensitivity, and strong specificity are developed to achieve the application of diagnostic kits in screening of early ovarian cancer.


Subject(s)
Early Detection of Cancer/methods , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...