Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Aging Cell ; 20(5): e13343, 2021 05.
Article in English | MEDLINE | ID: mdl-33721419

ABSTRACT

Although important factors governing the meiosis have been reported in the embryonic ovary, meiosis in postnatal testis remains poorly understood. Herein, we first report that SRY-box 30 (Sox30) is an age-related and essential regulator of meiosis in the postnatal testis. Sox30-null mice exhibited uniquely impaired testis, presenting the abnormal arrest of germ-cell differentiation and irregular Leydig cell proliferation. In aged Sox30-null mice, the observed testicular impairments were more severe. Furthermore, the germ-cell arrest occurred at the stage of meiotic zygotene spermatocytes, which is strongly associated with critical regulators of meiosis (such as Cyp26b1, Stra8 and Rec8) and sex differentiation (such as Rspo1, Foxl2, Sox9, Wnt4 and Ctnnb1). Mechanistically, Sox30 can activate Stra8 and Rec8, and inhibit Cyp26b1 and Ctnnb1 by direct binding to their promoters. A different Sox30 domain required for regulating the activity of these gene promoters, providing a "fail-safe" mechanism for Sox30 to facilitate germ-cell differentiation. Indeed, retinoic acid levels were reduced owing to increased degradation following the elevation of Cyp26b1 in Sox30-null testes. Re-expression of Sox30 in Sox30-null mice successfully restored germ-cell meiosis, differentiation and Leydig cell proliferation. Moreover, the restoration of actual fertility appeared to improve over time. Consistently, Rec8 and Stra8 were reactivated, and Cyp26b1 and Ctnnb1 were reinhibited in the restored testes. In summary, Sox30 is necessary, sufficient and age-associated for germ-cell meiosis and differentiation in testes by direct regulating critical regulators. This study advances our understanding of the regulation of germ-cell meiosis and differentiation in the postnatal testis.


Subject(s)
SOX Transcription Factors/physiology , Spermatozoa/cytology , Testis/cytology , Aging , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Gene Expression Regulation , Male , Meiosis , Meiotic Prophase I , Mice , Promoter Regions, Genetic , Protein Domains , SOX Transcription Factors/chemistry , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Sex Differentiation , Testis/metabolism , Tretinoin/metabolism
2.
Endocr Relat Cancer ; 27(2): X1-X2, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31995343

ABSTRACT

The authors and journal apologize for errors in the above paper, which appeared in volume 26 part 3, pages 303­319. The errors relate to the legend of Fig. 1A on page 307 and the artwork of Fig. 8D on page 316:

3.
Cell Death Dis ; 10(4): 316, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30962417

ABSTRACT

Depression is a condition with a complex etiological pattern, whose effective treatments are highly limited. MicroRNAs (miRNAs) have been investigated in intensive studies owing to their involvement in pathophysiology of mood disorders. The current study aimed to elucidate the role of miR-301b in hippocampus in mouse models of depressive-like behavior. Microarray-based prediction identified the differentially expressed gene neuronal pentraxin II (NPTX2) related to mental depression. Next, the putative miR-301b binding sites on the 3'UTR of NPTX2 were verified. Then the effect of miR-301b on cognitive function of mice with depressive-like behavior was analyzed using the Morris water maze test. In addition, the regulation of miR-301b to NPTX2 and activation of NF-κB signaling pathway was assessed. Following that, the microglia activation and inflammation in hippocampus were evaluated, with the expressions of inflammatory factors being examined. At last, microglia were flow cytometrically sorted and the inflammatory reaction was also assessed in vitro. The obtained findings revealed that miR-301b targeted and negatively regulated NPTX2. Moreover, overexpressed miR-301b activated the NF-κB signaling pathway, as reflected by increasing protein expressions of p-NF-κB. Upregulated miR-301b accelerated cognitive impairment in mice with depressive-like behavior. In addition, overexpression of miR-301b activated microglia and stimulated inflammation in hippocampus, accompanied by enhanced release of tumor necrosis factor-α (TNF-α), interleukin-Iß (IL-Iß) and cyclooxygenase-2(COX-2). Taken together, the evidence provided by the current study indicated that overexpression of miR-301b augmented hippocampal microglia activation, thus exacerbating cognitive impairment and inflammation in mice with depressive-like behavior by activating the NF-κB signaling pathway.


Subject(s)
Cognitive Dysfunction/metabolism , Depressive Disorder/metabolism , Hippocampus/metabolism , MicroRNAs/metabolism , Microglia/metabolism , NF-kappa B/metabolism , Animals , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Cognitive Dysfunction/genetics , Cyclooxygenase 2/metabolism , Depressive Disorder/genetics , Female , Hippocampus/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred ICR , MicroRNAs/genetics , Microglia/immunology , NF-kappa B/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
4.
Endocr Relat Cancer ; 26(3): 303-319, 2019 03.
Article in English | MEDLINE | ID: mdl-30608899

ABSTRACT

New potential biomarkers and therapeutic targets for ovarian cancer should be identified. The amplification in chromosomal region 5q31-5q35.3 exhibits the strongest correlation with overall survival (OS) of ovarian cancer. SOX30 coincidentally located at this chromosomal region has been determined as a new important tumor suppressor. However, the prognostic value, role and mechanism of SOX30 in ovarian cancer are unexplored. Here, we reveal that SOX30 is frequently overexpressed in ovarian cancer tissues and is associated with clinical stage and metastasis of ovarian cancer patients. High SOX30 expression predicts better OS and acts as an independent prognostic factor in advanced-stage patients, but is not associated with OS in early-stage patients. Based on the survival analyses, the advanced-stage patients with high SOX30 expression can receive platin- and/or taxol-based chemotherapy, whereas they should not receive chemotherapy containing gemcitabine or topotecan. Functionally, SOX30 strongly inhibits tumor cell migration and invasion in intro and suppresses tumor metastasis in vivo. SOX30 regulates some markers (E-CADHERIN, FIBRONECTIN, N-CADHERIN and VIMENTIN) and prevents the characteristics of epithelial-mesenchymal transition (EMT). SOX30 transcriptionally regulates the expression of E-CADHERIN, FIBRONECTIN and N-CADHERIN by binding to their promoters. Restoration of E-CADHERIN and/or N-CADHERIN when overexpressing SOX30 significantly reduces the anti-metastatic role of SOX30. Indeed, chemotherapy treatment containing platin or gemcitabine combined with SOX30 expression influences tumor cell metastasis and the survival of nude mice differently, which is closely associated with EMT. In conclusion, SOX30 antagonizes tumor metastasis by preventing EMT process that can be used to predict survival and incorporated into chemotherapeutics of advanced-stage ovarian cancer patients.


Subject(s)
Ovarian Neoplasms/diagnosis , SOX Transcription Factors/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Mice, Nude , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Prognosis , Survival Analysis
5.
FASEB J ; 33(1): 606-618, 2019 01.
Article in English | MEDLINE | ID: mdl-30118321

ABSTRACT

Depression represents a condition characterized by cognitive deficits and neural dysfunction and has recently been correlated with microRNAs (miRs) and their respective target genes. The present study was conducted with the goal of investigating the expression of miR-192-5p and its target gene fibulin (Fbln)-2 in an attempt to evaluate their roles in the occurrence and progression of cognitive impairment and neural function in mice with chronic unpredictable mild stress (CUMS)-induced depression through regulation of the TGF-ß1 signal transduction pathway. Verification of the targeting relationship between miR-192-5p and Fbln2 was provided in the form of initial bioinformatics prediction, followed by a further verification in the form of a dual-luciferase reporter gene assay. Normal mice and models induced by CUMS were assigned into various groups, whereas mimics, inhibitors, and small interfering RNA were introduced to validate the regulatory mechanism by which miR-192-5p regulates Fbln2 depression. Novel object recognition, tail suspension testing, and Morris water maze were all employed 28 d after transfection. Hippocampal electrophysiological recordings, Golgi staining, HPLC mass spectrometry, and fluorescence immunohistochemistry were performed to further evaluate cognitive function and neuron regeneration. CUMS-induced depression was determined to represent a predisposing factor for cognitive impairment and damage to neural function in mice, highlighted by novel object recognition, learning and memory abilities, population spike amplitude, synaptic transmission, cAMP levels, neuronal regeneration, and increased behavioral changes that resemble depression. Furthermore, increased Fbln2 expression, an activated TGF-ß1 signaling pathway, and decreased expression of miR-192-5p, synaptophysin, brain-derived neurotrophic factor, N-methyl-d-aspartate receptor subunit 2B, and calmodulin-dependent protein kinase II were noted. Up-regulated miR-192-5p targeting Fbln2 acts to alleviate CUMS-induced depression by inhibiting the TGF-ß1 signaling pathway, resulting in the enhanced cognitive function in novel object recognition, learning and memory ability, population spike amplitude, synaptic transmission, neuron regeneration, and alleviation of behavioral symptoms. The central findings of the present study indicate that up-regulated levels of miR-192-5p expression act to suppress activation of the TGF-ß1 signaling pathway by means of binding to Fbln2, thereby ameliorating cognitive impairment and strengthening neural function in a mouse model of depression.-Tang, C.-Z., Yang, J.-T., Liu, Q.-H., Wang, Y.-R., Wang, W.-S. Up-regulated miR-192-5p expression rescues cognitive impairment and restores neural function in mice with depression via the Fbln2-mediated TGF-ß1 signaling pathway.


Subject(s)
Calcium-Binding Proteins/metabolism , Cognitive Dysfunction/prevention & control , Depression/complications , Disease Models, Animal , Extracellular Matrix Proteins/metabolism , MicroRNAs/genetics , Transforming Growth Factor beta1/metabolism , Animals , Behavior, Animal , Calcium-Binding Proteins/genetics , Cell Proliferation , Cognitive Dysfunction/etiology , Depression/physiopathology , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Signal Transduction , Transforming Growth Factor beta1/genetics , Up-Regulation
6.
EBioMedicine ; 31: 253-266, 2018 May.
Article in English | MEDLINE | ID: mdl-29739711

ABSTRACT

Although high mortality of lung cancer is greatly due to distant metastasis, the mechanism of this metastasis remains unclear. Here, we investigate in lung cancer that SOX30 is sharply under-expressed in metastatic tumors compared with non-metastatic tumors, and suppresses plenty of metastasis related processes or pathways. SOX30 strongly inhibits tumor cell metastasis in vitro and in vivo. Sox30 deficiency promotes lung metastasis in Sox30-/- mice and this uncontrollable lung-metastasis is re-inhibited upon Sox30 re-expression. Mechanistically, SOX30 diminishes Wnt-signaling via directly transcriptional repressing ß-catenin or interacting with ß-catenin to compete with TCF for binding to ß-catenin. The carboxyl-terminus of SOX30 is required for attenuating ß-catenin transcriptional activity, whereas the amino-terminus of SOX30 is required for its interaction with ß-catenin protein. Enhance of ß-catenin attenuates the anti-metastatic role of SOX30. Moreover, Sox30 deficiency promotes tumor metastasis and reduces survival of mice. In addition, nuclear SOX30 expression is closely associated with metastasis and represents a favorable independent prognostic biomarker of lung cancer patients. Altogether, these results highlight an important role and mechanism of SOX30 in lung cancer metastasis, providing a potential therapeutic target for anti-metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , SOX Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , A549 Cells , Adult , Aged , Aged, 80 and over , Animals , Female , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Knockout , Middle Aged , Neoplasm Metastasis , SOX Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , beta Catenin/genetics
7.
Oncotarget ; 8(45): 78713-78725, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-29108259

ABSTRACT

The vital copy number variation (CNV) plays a crucial role in clear cell renal cell carcinoma (ccRCC). MPDZ inhibit cell polarity associate with osmotic pressure response and cancer-related biological processes. In order to clarify the role of the CNV of MPDZ in the progression of ccRCC, we analyzed the CNV and expression of MPDZ and prognosis in ccRCC patients from The Cancer Genome Atlas data portal. Notably, we found that the deletion of MPDZ was the common CNV, which was present in 28.65% of ccRCC patients. With the development of tumors, the percentage of MPDZ deletion increased significantly (19.38% in stage I; 20.00% in stage II; 40.94% in stage III; and 45.00% in stage IV). The deletion of MPDZ significantly increased ccRCC risk (P=0.0025). Low MPDZ expression associated with its deletion was significantly associated with adverse outcomes in ccRCC patients (P=0.0342). Furthermore, immunohistochemical analysis by tissue microarray showed that MPDZ was expressed at lower levels in tumor tissues compared with adjacent tissues (P<0.01). Kaplan-Meier survival curves showed that ccRCC patients with low MPDZ expression had significantly shorter survival than those with high MPDZ expression (P=0.002). These results indicated that low MPDZ expression associated with CNV is a potential biomarker for the prognosis of ccRCC patients.

SELECTION OF CITATIONS
SEARCH DETAIL
...