Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Biol Trace Elem Res ; 202(3): 1084-1102, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37382810

ABSTRACT

Aluminum contamination is a growing environmental and public health concern, and aluminum testicular toxicity has been reported in male rats; however, the underlying mechanisms of this toxicity are unclear. The objective of this study was to investigate the effects of exposure to aluminum chloride (AlCl3) on alterations in the levels of sex hormones (testosterone [T], luteinizing hormone [LH], and follicle-stimulating hormone [FSH]) and testicular damage. Additionally, the mechanisms of toxicity in the testes of AlCl3-exposed rats were analyzed by proteomics. Three different concentrations of AlCl3 were administered to rats. The results demonstrated a decrease in T, LH, and FSH levels with increasing concentrations of AlCl3 exposure. HE staining results revealed that the spermatogenic cells in the AlCl3-exposed rats were widened, disorganized, or absent, with increased severe tissue destruction at higher concentrations of AlCl3 exposure. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses revealed that differentially expressed proteins (DEPs) after AlCl3 exposure were primarily associated with various metabolic processes, sperm fibrous sheath, calcium-dependent protein binding, oxidative phosphorylation, and ribosomes. Subsequently, DEPs from each group were subjected to protein-protein interaction (PPI) analysis followed by the screening of interactional key DEPs. Western blot experiments validated the proteomics data, revealing the downregulation of sperm-related DEPs (AKAP4, ODF1, and OAZ3) and upregulation of regulatory ribosome-associated protein (UBA52) and mitochondrial ribosomal protein (MRPL32). These findings provide a basis for studying the mechanism of testicular toxicity due to AlCl3 exposure.


Subject(s)
Aluminum , Testis , Rats , Male , Animals , Aluminum/metabolism , Proteomics , Semen , Testosterone/metabolism , Luteinizing Hormone/metabolism , Follicle Stimulating Hormone/metabolism
2.
Gene ; 893: 147961, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-37931853

ABSTRACT

BACKGROUND: Lupus nephritis (LN) is a common immune disease. The microRNA (miR)-181d-5p is a potential target for treating kidney injury. However, the therapeutic role of miR-181d-5p in LN has not been investigated. This study aimed to investigate the role of miR-181d-5p in targeting mitogen-activated protein kinase 8 (MAPK8) and stimulating the MAPK signaling pathway in LN. METHODS: RT-qPCR was performed to identify the variations in miR-181d-5p expression in peripheral blood mononuclear cells (PBMCs) obtained from 42 LN patients, 30 healthy individuals, 6 MRL/lpr mice and 6 C57BL/6 mice. Western blot was used to detect the effect of miR-181d-5p on the MAPK signaling pathway in THP-1 cells and MRL/lpr mice. Enzyme-linked immunosorbent assay (ELISA) was utilized to detect the effect of miR-181d-5p on antinuclear antibodies and inflammatory factors. A dual-luciferase reporter assay was used to verify whether miR-181d-5p directly targets MAPK8. Flow cytometry was performed to evaluate apoptosis rates in transfected THP-1 cells. RESULTS: miR-181d-5p expression was downregulated in PBMCs of LN patients (P < 0.01) and MRL/lpr mice (P < 0.05). A dual luciferase reporter assay demonstrated that miR-181d-5p inhibits MAPK8 (P < 0.01). Overexpression of miR-181d-5p inhibited the phosphorylation of p38 (P < 0.001) and p44/42 (P < 0.01). Moreover, miR-181d-5p decreased the apoptosis rate of THP-1 cells (P < 0.001), and reduced the secretion of IL-6 (P < 0.01) and TNF-α (P < 0.01). Furthermore, overexpression of miR-181d-5p decreased anti-dsDNA antibody (P < 0.05), anti-Sm antibody (P < 0.01), and fibrosis levels in MRL/lpr mice. CONCLUSION: Upregulation of miR-181d-5p showed anti-inflammatory and anti-apoptotic effects on THP-1 cells in vitro and kidney injury in vivo. These effects were achieved by miR-181d-5p targeting MAPK8 to inhibit phosphorylation of p38 and p44/42. These results may offer new insights for improving therapeutic strategies against lupus nephritis.


Subject(s)
Lupus Nephritis , MicroRNAs , Mice , Animals , Humans , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Mitogen-Activated Protein Kinase 8 , MicroRNAs/metabolism , Leukocytes, Mononuclear/metabolism , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Signal Transduction , Mitogen-Activated Protein Kinases/metabolism , Luciferases/metabolism
3.
Funct Integr Genomics ; 23(3): 264, 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37541978

ABSTRACT

Liver cancer is a cunning malignancy with a high incidence and mortality rate among cancers worldwide. The NPC gene family members (NPCs: NPC1, NPC2, and NPC1L1) are closely linked to the development of multiple cancers, but their role in liver cancer remains unclear. As a result, we must investigate their functions in liver hepatocellular carcinoma (LIHC). NPCs were significantly differentially expressed between normal and LIHC tissues, with a high mutation frequency in LIHC. The ROC curve analysis revealed that NPC1/NPC2 had high diagnostic and prognostic values in LIHC. NPC1 expression was also found to be negatively correlated with its methylation level. The differentially expressed genes between high and low NPC1 expression groups in LIHC were mainly related to channel activity, transporter complexes, and plasma membrane adhesion molecules. Additionally, NPC1 expression was significantly associated with multiple immune cells and immunization checkpoints. It was hypothesized that a TUG1/SNHG4-miR-148a-3p-NPC1 regulatory axis is associated with hepatocarcinogenesis. Finally, the protein expression of NPC1 in LIHC tissues and paraneoplastic tissues was detected, and NPC1-knockdown HepG2 cells (NPC1KO) inhibited the proliferation, migration, and invasion. This study helped to identify new prognostic markers and potential immunotherapeutic targets for LIHC and revealed the molecular mechanisms underlying NPC1 regulation in LIHC. The NPCs play a key role in the prognosis and diagnosis of LIHC and may be an important indicator for LIHC prognosis and diagnosis; NPC1 might be a potential therapeutic target in LIHC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Prognosis , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Multiomics
4.
Obesity (Silver Spring) ; 31(1): 123-138, 2023 01.
Article in English | MEDLINE | ID: mdl-36504350

ABSTRACT

OBJECTIVE: Genetic variants in ninjurin-2 (NINJ2; nerve injury-induced protein 2) confer risk of ischemic strokes and coronary artery disease as well as endothelial activation and inflammation. However, little is known about NINJ2's in vivo functions and underlying mechanisms. METHODS: The phenotypes of NINJ2 knockout mice were analyzed, and mechanisms of NINJ2 that regulate body weight, insulin resistance, and glucose homeostasis and lipogenesis were investigated in vivo and in vitro. RESULTS: This study found that mice lacking NINJ2 showed impaired adipogenesis, increased insulin resistance, and abnormal glucose homeostasis, all of which are risk factors for strokes and coronary artery disease. Mechanistically, NINJ2 directly interacts with insulin receptor/insulin-like growth factor 1 receptor (INSR/IGF1R), and NINJ2 knockdown can block insulin-induced mitotic clonal expansion during preadipocyte differentiation by inhibiting protein kinase B/extracellular signal-regulated kinase (AKT/ERK) signaling and by decreasing the expression of key adipocyte transcriptional regulators CCAAT/enhancer-binding protein ß (C/EBP-ß), C/EBP-α, and peroxisome proliferator-activated receptor γ (PPAR-γ). Furthermore, the interaction between NINJ2 and INSR/IGF1R is needed for maintaining insulin sensitivity in adipocytes and muscle via AKT and glucose transporter type 4. Notably, adenovirus-mediated NINJ2 overexpression can ameliorate diet-induced insulin resistance in mice. CONCLUSIONS: In conclusion, these findings reveal NINJ2 as an important new facilitator of insulin receptors, and the authors propose a unique regulatory mechanism between insulin signaling, adipogenesis, and insulin resistance.


Subject(s)
Cell Adhesion Molecules, Neuronal , Insulin Resistance , Animals , Mice , 3T3-L1 Cells , Adipogenesis/genetics , Cell Differentiation/genetics , Coronary Artery Disease , Glucose/metabolism , Insulin , Insulin Resistance/genetics , PPAR gamma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Adhesion Molecules, Neuronal/genetics
5.
Metabolism ; 140: 155380, 2023 03.
Article in English | MEDLINE | ID: mdl-36549436

ABSTRACT

BACKGROUND: Liver fibrogenesis is orchestrated by the paracrine signaling interaction between several resident cell types regulating the activation of hepatic stellate cells (HSCs). However, the molecular mechanisms underlying paracrine regulation are largely unknown. The aim of this study is to elucidate the role of Ninjurin2 in the crosstalk between hepatocytes and HSCs and better understand the implications of Ninjurin2 in liver fibrosis. METHODS: Ninj2 knockout mice (Ninj2-/-) and hepatocyte-specific Ninj2 overexpression mice (Ninj2Hep-tg) were constructed and followed by the induction of liver fibrosis using methionine- and choline-deficient (MCD) diet. The relationship between Ninjurin2 and liver fibrosis phenotype was evaluated in vivo by measurement of fibrotic markers and related genes. We used an in vitro transwell cell co-culture model to examine the impact of Ninjurin2 in hepatocytes on the crosstalk to HSCs. The interaction of Ninjurin2 and IGF1R and the regulation of PI3K-AKT-EGR1 were analyzed in vivo and in vitro. Finally, an inhibitory Ninjurin2 peptide was injected intravenously via the tail vein to investigate whether inhibiting of Ninjurin2 cascade can attenuate MCD diet-induced liver fibrosis in mice. RESULTS: We found that hepatic Ninjurin2 expression was significantly increased in fibrotic human liver and MCD diet-induced liver injury mouse models. In the mouse model, hepatocyte-specific overexpression of Ninj2 exacerbates MCD-induced liver fibrosis, while global Ninj2 knockout reverses the phenotype. To mimic hepatocyte-HSC crosstalk during liver fibrosis, we used co-culture systems containing hepatocytes and HSCs and determined that Ninjurin2 overexpression in hepatocytes directly activates HSCs in vitro. Mechanistically, Ninjurin2 directly interacts with insulin-like growth factor 1 receptor (IGF1R) and increases the hepatocyte secretion of the fibrogenic cytokine, platelet-derived growth factor-BB (PDGF-BB) through IGF1R-PI3K-AKT-EGR1 cascade. Inhibition of PDGFRB signaling in HSCs can abolish the profibrogenic effect of Ninjurin2. In addition, we demonstrated that a specific inhibitory Ninjurin2 peptide containing an N-terminal adhesion motif mitigates liver fibrosis and improves hepatic function in the mouse models by negatively regulating the sensitivity of IGF1R to IGF1 in hepatocytes. CONCLUSION: Hepatic Ninjurin2 plays a key role in liver fibrosis through paracrine regulation of PDGF-BB/PDGFRB signaling in HSCs, and the results suggesting Ninjurin2 may be a potential therapeutic target.


Subject(s)
Cell Adhesion Molecules, Neuronal , Hepatic Stellate Cells , Liver , Signal Transduction , Animals , Humans , Mice , Becaplermin/metabolism , Becaplermin/pharmacology , Becaplermin/therapeutic use , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/pharmacology , Cell Adhesion Molecules, Neuronal/therapeutic use , Disease Models, Animal , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Early Growth Response Protein 1/pharmacology , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Fibrosis
6.
J Biol Chem ; 298(4): 101759, 2022 04.
Article in English | MEDLINE | ID: mdl-35202649

ABSTRACT

Angiogenic factor AGGF1 (AngioGenic factor with G-patch and FHA (Forkhead-Associated) domain 1) blocks neointimal formation (formation of a new or thickened layer of arterial intima) after vascular injury by regulating phenotypic switching of vascular smooth muscle cells (VSMCs). However, the AGGF1 receptor on VSMCs and the underlying molecular mechanisms of its action are unknown. In this study, we used functional analysis of serial AGGF1 deletions to reveal the critical AGGF1 domain involved in VSMC phenotypic switching. This domain was required for VSMC phenotypic switching, proliferation, cell cycle regulation, and migration, as well as the regulation of cell cycle inhibitors cyclin D, p27, and p21. This domain also contains an RDDAPAS motif via which AGGF1 interacts with integrin α7 (ITGA7), but not α8. In addition, we show that AGGF1 enhanced the expression of contractile markers MYH11, α-SMA, and SM22 and inhibited MEK1/2, ERK1/2, and ELK phosphorylation in VSMCs, and that these effects were inhibited by knockdown of ITGA7, but not by knockdown of ITGA8. In vivo, deletion of the VSMC phenotypic switching domain in mice with vascular injury inhibited the functions of AGGF1 in upregulating α-SMA and SM22, inhibiting MEK1/2, ERK1/2, and ELK phosphorylation, in VSMC proliferation, and in blocking neointimal formation. Finally, we show the inhibitory effect of AGGF1 on neointimal formation was blocked by lentivirus-delivered shRNA targeting ITGA7. Our data demonstrate that AGGF1 interacts with its receptor integrin α7 on VSMCs, and this interaction is required for AGGF1 signaling in VSMCs and for attenuation of neointimal formation after vascular injury.


Subject(s)
Muscle, Smooth, Vascular , Vascular System Injuries , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Antigens, CD/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Integrin alpha Chains/metabolism , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/genetics , Neointima/metabolism , Vascular System Injuries/metabolism
7.
Aging (Albany NY) ; 13(23): 25393-25407, 2021 12 12.
Article in English | MEDLINE | ID: mdl-34897030

ABSTRACT

OBJECTIVE: NINJ2 regulates activation of vascular endothelial cells, and genome-wide association studies showed that variants in NINJ2 confer risk to stroke. However, whether variants in NINJ2 are associated with coronary artery disease (CAD) is unknown. METHODS: We genotyped rs34166160 in NINJ2 in two independent Chinese GeneID populations which included 2,794 CAD cases and 4,131 controls, and performed genetics association studies. Functional studies were also performed to reveal the mechanisms. RESULTS: Allele rs34166160 significantly confers risk to CAD in the GeneID Hubei population which contained 1,440 CAD cases and 2,660 CAD-free controls (observed P-obs = 6.39 × 10-3 with an odds ratio (OR) was 3.39, adjusted P-adj = 8.12 × 10-3 with an OR of 3.10). The association was replicated in another population, GeneID Shandong population contained 1,354 CAD cases and 1,471 controls (P-obs = 3.33 × 10-3 with an OR of 3.14, P-adj = 0.01 with an OR of 2.74). After combining the two populations, the association was more significant (P-obs = 1.57 × 10-5 with an OR of 3.58, P-adj = 3.41 × 10-4 with an OR of 2.80). In addition, we found that rs34166160 was associated with the mRNA expression level of NINJ2 and the flanking region of rs34166160 can directly bind with transcriptional factor CCAAT-box/enhancer-binding protein beta, and the risk A allele has more transcription activity than non-risk C allele with or without LPS in HUVEC cells. CONCLUSIONS: Our study demonstrates that the functional rare variant rs34166160 in NINJ2 confers risk to CAD for the first time, and these findings further expand the range of the pathology of CAD and atherosclerosis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Cell Adhesion Molecules, Neuronal/genetics , Coronary Artery Disease/genetics , Genetic Predisposition to Disease/genetics , Alleles , Binding Sites/genetics , Case-Control Studies , Female , Genetic Association Studies , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Real-Time Polymerase Chain Reaction , Risk Factors
8.
Arterioscler Thromb Vasc Biol ; 41(11): 2756-2769, 2021 11.
Article in English | MEDLINE | ID: mdl-34551592

ABSTRACT

Objective: Angiogenic factor AGGF1 (angiogenic factor with G-patch and FHA [Forkhead-associated] domain 1) promotes angiogenesis as potently as VEGFA (vascular endothelial growth factor A) and regulates endothelial cell (EC) proliferation, migration, specification of multipotent hemangioblasts and venous ECs, hematopoiesis, and vascular development and causes vascular disease Klippel-Trenaunay syndrome when mutated. However, the receptor for AGGF1 and the underlying molecular mechanisms remain to be defined. Approach and Results: Using functional blocking studies with neutralizing antibodies, we identified [alpha]5[beta]1 as the receptor for AGGF1 on ECs. AGGF1 interacts with [alpha]5[beta]1 and activates FAK (focal adhesion kinase), Src (proto-oncogene tyrosine-protein kinase), and AKT (protein kinase B). Functional analysis of 12 serial N-terminal deletions and 13 C-terminal deletions by every 50 amino acids mapped the angiogenic domain of AGGF1 to a domain between amino acids 604-613 (FQRDDAPAS). The angiogenic domain is required for EC adhesion and migration, capillary tube formation, and AKT activation. The deletion of the angiogenic domain eliminated the effects of AGGF1 on therapeutic angiogenesis and increased blood flow in a mouse model for peripheral artery disease. A 40-mer or 15-mer peptide containing the angiogenic domain blocks AGGF1 function, however, a 15-mer peptide containing a single amino acid mutation from -RDD- to -RGD- (a classical RGD integrin-binding motif) failed to block AGGF1 function. Conclusions: We have identified integrin [alpha]5[beta]1 as an EC receptor for AGGF1 and a novel AGGF1-mediated signaling pathway of [alpha]5[beta]1-FAK-Src-AKT for angiogenesis. Our results identify an FQRDDAPAS angiogenic domain of AGGF1 crucial for its interaction with [alpha]5[beta]1 and signaling.


Subject(s)
Angiogenic Proteins/metabolism , Endothelial Cells/metabolism , Hindlimb/blood supply , Integrin alpha5beta1/metabolism , Ischemia/metabolism , Neovascularization, Physiologic , 3T3-L1 Cells , Angiogenesis Inducing Agents/pharmacology , Angiogenic Proteins/genetics , Angiogenic Proteins/pharmacology , Animals , Disease Models, Animal , Endothelial Cells/drug effects , Female , Focal Adhesion Kinase 1/metabolism , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Integrin alpha5beta1/genetics , Ischemia/drug therapy , Ischemia/genetics , Ischemia/physiopathology , Ligands , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Phosphorylation , Protein Interaction Domains and Motifs , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , src-Family Kinases/metabolism
9.
Mol Cancer Ther ; 16(9): 1739-1750, 2017 09.
Article in English | MEDLINE | ID: mdl-28572168

ABSTRACT

Ovarian cancer remains a significant cause of gynecologic cancer mortality, and novel therapeutic strategies are urgently needed in clinic as new treatment options. We previously showed that BET bromodomain inhibitors displayed promising efficacy for the treatment of epithelial ovarian cancer by downregulating pivot transcription factors. However, the potential antitumor activities and molecular mechanisms of other epigenetic or transcriptional therapies have not been systematically determined. Here, by performing an unbiased high-throughput drug screen to identify candidate compounds with antineoplastic effects, we identified THZ1, a recently developed covalent CDK7 inhibitor, as a new transcription-targeting compound that exerted broad cytotoxicity against ovarian tumors. Mechanistically, CDK7 represented a previously unappreciated actionable vulnerability in ovarian cancer, and CDK7 inhibition led to a pronounced dysregulation of gene transcription, with a preferential repression of E2F-regulated genes and transcripts associated with super-enhancers. Our findings revealed the molecular underpinnings of THZ1 potency and established pharmaceutically targeting transcriptional addiction as a promising therapeutic strategy in aggressive ovarian cancer. Mol Cancer Ther; 16(9); 1739-50. ©2017 AACR.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Transcription, Genetic , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Screening Assays, Antitumor , E2F Transcription Factors/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Phenylenediamines/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays , Cyclin-Dependent Kinase-Activating Kinase
10.
Cell Signal ; 35: 231-241, 2017 07.
Article in English | MEDLINE | ID: mdl-28431986

ABSTRACT

Previous genetic studies suggested that variants in NINJ2 (encode ninjurin2) confer risk to ischemic stroke or large artery atherosclerotic stroke. However, the underlying mechanisms of NINJ2 in ischemic stroke or atherosclerosis are still unknown. In this study, we hypothesized that NINJ2 may play a role in endothelial inflammation and activation, and regulate the process of atherosclerosis. Here, we demonstrated that NINJ2 can regulate the expression of a panel of genes that are associated with inflammation and atherosclerosis (e.g. IL-1ß, TNF-α, IL-8, IL-6, ICAM-1 and E-selectin) in human vascular endothelial cells (HUVECs). Moreover, we found the expression of ninjurin2 is upregulated in LPS stimulated HUVECs and mouse aorta, and it can regulate LPS-induced endothelial activation and the adhesion of monocytes to endothelial cells. We also found that NINJ2 can regulate NF-κB and c-jun through interacting with TLR4. In conclusion, our study suggests that ninjurin2 is a novel regulator of endothelia inflammation and activation through TLR4 signaling pathways, and these data provided new insights into the mechanisms between NINJ2 and atherosclerosis.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Coronary Artery Disease/genetics , Stroke/genetics , Toll-Like Receptor 4/genetics , Animals , Cell Adhesion/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Coronary Artery Disease/pathology , Endothelium/metabolism , Endothelium/pathology , Gene Expression Regulation/genetics , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/genetics , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Monocytes/metabolism , Monocytes/pathology , NF-kappa B/genetics , Signal Transduction/genetics , Stroke/pathology , Toll-Like Receptor 4/metabolism
11.
Theranostics ; 6(2): 219-30, 2016.
Article in English | MEDLINE | ID: mdl-26877780

ABSTRACT

Ovarian cancer is responsible for the highest mortality among all gynecologic malignancies, and novel therapies are urgently needed to improve patient outcome. Here we performed an integrative genomic analysis and identified the bromodomain and extraterminal domain (BET) protein BRD4 as a potential therapeutic target in ovarian cancer. Suppression of BRD4 using small-molecule BET inhibitors JQ1 and I-BET151, or dual kinase-bromodomain inhibitor volasertib, led to robust and broad antitumor effects across all subclasses of ovarian cancer. In contrast to many other cancers which are susceptible to BET inhibition due to downregulation of super-enhancer-dependent MYC transcript, we discovered that JQ1-sensitive ovarian cancer cells exhibited marked disruption of Forkhead box protein M1 (FoxM1) pathway, a key driver of ovarian carcinoma. These in vitro findings were further supported by in vivo efficacies of JQ1 targeting both cell line-based and patient-derived xenograft models. Our data establish a new treatment strategy against ovarian cancer by employing epigenetic vulnerabilities, and provide a mechanistic rationale for the clinical investigation of BET bromodomain inhibitors in this deadly disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Forkhead Box Protein M1/metabolism , Nuclear Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Azepines/therapeutic use , Cell Line, Tumor , Down-Regulation , Female , Forkhead Box Protein M1/genetics , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/metabolism , Pteridines/pharmacology , Pteridines/therapeutic use , Transcription Factors/metabolism , Triazoles/pharmacology , Triazoles/therapeutic use
12.
Sci Rep ; 5: 16066, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26530441

ABSTRACT

High-grade serous ovarian carcinoma (HGS-OvCa) has the lowest survival rate among all gynecologic cancers and is hallmarked by a high degree of heterogeneity. The Cancer Genome Atlas network has described a gene expression-based molecular classification of HGS-OvCa into Differentiated, Mesenchymal, Immunoreactive and Proliferative subtypes. However, the biological underpinnings and regulatory mechanisms underlying the distinct molecular subtypes are largely unknown. Here we showed that tumor-infiltrating stromal cells significantly contributed to the assignments of Mesenchymal and Immunoreactive clusters. Using reverse engineering and an unbiased interrogation of subtype regulatory networks, we identified the transcriptional modules containing master regulators that drive gene expression of Mesenchymal and Immunoreactive HGS-OvCa. Mesenchymal master regulators were associated with poor prognosis, while Immunoreactive master regulators positively correlated with overall survival. Meta-analysis of 749 HGS-OvCa expression profiles confirmed that master regulators as a prognostic signature were able to predict patient outcome. Our data unraveled master regulatory programs of HGS-OvCa subtypes with prognostic and potentially therapeutic relevance, and suggested that the unique transcriptional and clinical characteristics of ovarian Mesenchymal and Immunoreactive subtypes could be, at least partially, ascribed to tumor microenvironment.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Ovarian Neoplasms/classification , Ovarian Neoplasms/mortality , Stromal Cells/pathology , Algorithms , Biomarkers, Tumor/genetics , Female , Gene Expression Profiling , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis , Survival Rate , Tumor Microenvironment/physiology
13.
Carcinogenesis ; 36(9): 937-45, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26071398

ABSTRACT

It has been increasingly recognized that microRNAs (miRNAs) are often dysregulated in various human malignancies and can function as oncogenes or tumor-suppressors. However, the potential roles of miRNAs and components of the miRNA biogenesis pathway remain poorly defined in melanoma. Here, we systematically profiled miRNA expression in human melanocytes and melanoma cells, and identified a prominent function of miR-125a-5p in suppressing melanoma growth. Mechanistically, we discovered that Lin28B, a well-characterized inhibitor of let-7 miRNA biogenesis, was a direct target of miR-125a-5p in melanoma. We showed that the Lin28B was aberrantly expressed in a large proportion of melanoma patients and was functionally required for melanoma progression. We further demonstrated the involvement of let-7-dependent mechanism downstream of Lin28B, resulting in the activation of transforming growth factor-ß signaling cascade. Collectively, our data implicate Lin28B as a novel oncogene in melanomagenesis by mediating a miRNA regulatory circuit.


Subject(s)
Melanoma/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Proteins/physiology , Skin Neoplasms/pathology , Transforming Growth Factor beta/genetics , Animals , Cell Line, Tumor , Down-Regulation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , RNA-Binding Proteins/genetics , Signal Transduction/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...