1
|
Zhu L, Liu C, Wang Y, Zhu X, Wu L, Chen L, Zhou J, Wang F. METTL3/IGF2BP2/IκBα axis participates in neuroinflammation in Alzheimer's disease by regulating M1/M2 polarization of microglia. Neurochem Int 2025; 186:105964. [PMID: 40107503 DOI: 10.1016/j.neuint.2025.105964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Microglia-mediated neuroinflammation is closely related to the development of Alzheimer's disease (AD). This study further elucidated the regulatory mechanism of microglia polarization in AD. METHOD Microglia polarization was assessed using RT-qPCR, ELISA, and immunofluorescence (IF). Western blot (WB) analyzed inflammation-related, p-tau, and apoptosis-related proteins. Neuronal damage was evaluated by immunofluorescence, and neuronal apoptosis by flow cytometry and TUNEL assay. METTL3 and IκBα expression were detected using RT-qPCR and WB. N6-methyladenosine (m6A) levels were quantified with a colorimetric assay. RNA pull-down assay examined METTL3, IGF2BP2, and IκBα mRNA binding. IGF2BP expression was assessed by RT-qPCR. Learning and memory abilities were evaluated using morris water maze (MWM) test and novel object recognition (NOR) test. Inflammation-related proteins were detected using IF. RESULTS Stimulation with Aβ1-42 led to microglia M1 polarization, upregulation of inflammation-related proteins, and exacerbation of neuronal injury and apoptosis, along with increased p-tau expression in neurons. METTL3/IGF2BP2 modulated IκBα m6A modification through binding to IκBα mRNA, enhancing its expression. Enhanced METTL3 or IGF2BP2 expression suppressed M1 polarization, inflammation, and neuronal apoptosis in microglia, reversed by knockdown of IκBα. AD model mice exhibited cognitive impairments, neuroinflammation, and elevated M1 polarization. METTL3 or IGF2BP2 overexpression improved cognitive function, reduced neuroinflammation, and inhibited M1 polarization, and this effect was similarly reversed by knockdown of IκBα. CONCLUSION Our study demonstrates that the METTL3/IGF2BP2/IκBα axis is involved in neuroinflammation in AD by modulating microglia M1/M2 polarization, which sheds light on the treatment of AD.
Collapse
Affiliation(s)
- Ling Zhu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Congyan Liu
- Department of pharmacy, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Yang Wang
- Department of radiology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Xuanang Zhu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Lei Wu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Lvan Chen
- Department of neurosurgery, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Jing Zhou
- College of Medical, Jingchu University of Technology, Jingmen, 448000, China.
| | - Fan Wang
- Department of neurosurgery, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China; College of Medical, Jingchu University of Technology, Jingmen, 448000, China.
| |
Collapse
|
2
|
Yu P, Zhao X, Zhou D, Wang S, Hu Z, Lian K, Zhang N, Duan P. The microRNA-mediated apoptotic signaling axis in male reproduction: a possible and targetable culprit in male infertility. Cell Biol Toxicol 2025; 41:54. [PMID: 40038116 DOI: 10.1007/s10565-025-10006-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
Recently, infertility has emerged as a significant and prevalent public health concern warranting considerable attention. Apoptosis, recognized as programmed cell death, constitutes a crucial process essential for the maintenance of normal spermatogenesis. Multiple investigations have illustrated that the dysregulated apoptosis of reproductive cells, encompassing spermatogonial stem cells, Sertoli cells, and Leydig cells, serves as a causative factor in male infertility. MicroRNAs represent a class of small RNA molecules that exert negative regulatory control over gene expression using direct interaction with messenger RNA transcripts. Previous studies have established that aberrant expression of miRNAs induces apoptosis in reproductive tissues, correlating with reproductive dysfunctions and infertility. In this review, we offer a comprehensive overview of miRNAs and their respective target genes implicated in the apoptotic process. As well, miRNAs are involved in multiple apoptotic signaling pathways, namely the PI3K/AKT, NOTCH, Wnt/β-catenin, and mTOR signaling cascades, exerting both negative and positive effects. We additionally elucidate the significant functions played by lncRNAs and circular RNAs as competing endogenous RNAs in the process of apoptosis within reproductive cells. We further illustrate that external factors, including silica nanoparticles, Cyclosporine A, and smoking, induce dysregulation of miRNAs, resulting in apoptosis within reproductive cells and subsequent male reproductive toxicity. Further, we discuss the implication of heat stress, hypoxia, and diabetes in reproductive cell apoptosis induced by miRNA dysregulation in male infertility. Finally, we demonstrate that the modulation of miRNAs via traditional and novel medicine could protect reproductive cells from apoptosis and be implemented as a therapeutic approach in male infertility.
Collapse
Affiliation(s)
- Pengxia Yu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Xue Zhao
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
- Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Dan Zhou
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Songtao Wang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Zihuan Hu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Kai Lian
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Nanhui Zhang
- Department of Nephrology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| | - Peng Duan
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| |
Collapse
|
3
|
Garabed LR, Flannigan R. The changing landscape of nonobstructive azoospermia. Curr Opin Urol 2025; 35:127-134. [PMID: 39604250 DOI: 10.1097/mou.0000000000001252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
PURPOSE OF REVIEW This article aims to describe new developments in the field of nonobstructive azoospermia biology, diagnostics, biomarkers, and therapeutic strategies. RECENT FINDINGS Recent studies have investigated the molecular underpinnings of cellular dysfunction that is contributing to spermatogenic dysfunction and findings suggest abnormalities across both somatic and germ cells. Biomarkers to predict the chances of sperm retrieval are being explored utilizing cell free (cf) DNA and RNA from various body fluids, in addition to a full range of transcripts and epigenetics within seminal fluid. Various approaches are being explored to optimize sperm identification from surgical specimens including microfluidic and machine learning approaches. Finally, approaches to regenerating sperm production from males with nonobstructive azoospermia are evolving to include various 3-dimensional culture techniques with integration of computational modeling. SUMMARY The landscape of nonobstructive azoospermia biomarkers, molecular underpinnings, technological approaches to more reliably identify sperm and novel regenerative therapeutic strategies are likely to transform the field of male reproduction in years to come.
Collapse
Affiliation(s)
- Laurianne Rita Garabed
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
4
|
He CM, Zhang D, He Z. Gene regulation and signaling transduction in mediating the self-renewal, differentiation, and apoptosis of spermatogonial stem cells. Asian J Androl 2025; 27:4-12. [PMID: 39162186 PMCID: PMC11784953 DOI: 10.4103/aja202464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/04/2024] [Indexed: 08/21/2024] Open
Abstract
ABSTRACT Infertility has become one of the most serious diseases worldwide, and 50% of this disease can be attributed to male-related factors. Spermatogenesis, by definition, is a complex process by which spermatogonial stem cells (SSCs) self-renew to maintain stem cell population within the testes and differentiate into mature spermatids. It is of great significance to uncover gene regulation and signaling pathways that are involved in the fate determinations of SSCs with aims to better understand molecular mechanisms underlying human spermatogenesis and identify novel targets for gene therapy of male infertility. Significant achievement has recently been made in demonstrating the signaling molecules and pathways mediating the fate decisions of mammalian SSCs. In this review, we address key gene regulation and crucial signaling transduction pathways in controlling the self-renewal, differentiation, and apoptosis of SSCs, and we illustrate the networks of genes and signaling pathways in SSC fate determinations. We also highlight perspectives and future directions in SSC regulation by genes and their signaling pathways. This review could provide novel insights into the genetic regulation of normal and abnormal spermatogenesis and offer molecular targets to develop new approaches for gene therapy of male infertility.
Collapse
Affiliation(s)
- Cai-Mei He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Changsha 410013, China
- Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
- Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Dong Zhang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Changsha 410013, China
- Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
- Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Changsha 410013, China
- Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha 410013, China
- Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| |
Collapse
|
5
|
Li C, Chen W, Cui Y, Zhang D, Yuan Q, Yu X, He Z. Essential Regulation of YAP1 in Fate Determinations of Spermatogonial Stem Cells and Male Fertility by Interacting with RAD21 and Targeting NEDD4 in Humans and Mice. RESEARCH (WASHINGTON, D.C.) 2024; 7:0544. [PMID: 39659446 PMCID: PMC11628678 DOI: 10.34133/research.0544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
Spermatogenesis is a sophisticated biological process by which spermatogonial stem cells (SSCs) undergo self-renewal and differentiation into spermatozoa. Molecular mechanisms underlying fate determinations of human SSCs by key genes and signaling pathways remain elusive. Here, we report for the first time that Yes1-associated transcriptional regulator (YAP1) is required for fate determinations of SSCs and male fertility by interacting with RAD21 and targeting NEDD4 in humans and mice. YAP1 was mainly located at cell nuclei of human SSCs. YAP1 silencing resulted in the decreases in proliferation and DNA synthesis as well as an enhancement in apoptosis of human SSCs both in vivo and in vitro. RNA sequencing and real-time polymerase chain reaction assays identified NEDD4 as a target of YAP1, and NEDD4 knockdown inhibited the proliferation of human SSCs and increased their apoptosis. Furthermore, YAP1 interacted with RAD21 to regulate NEDD4 transcription in human SSCs. Importantly, YAP1 abnormalities were found to be associated with non-obstructive azoospermia (NOA) as manifested as lower expression level of YAP1 in testicular tissues of NOA patients and YAP1 single-nucleotide variants (SNVs) in 777 NOA patients. Finally, Yap1 germline conditional knockout (cKO) mice assumed mitotic arrest, low sperm count, and motility. Collectively, these results highlight a critical role of YAP1 in determining the fate determinations of human SSCs and male infertility through the YAP1/RAD21/NEDD4 pathway. This study provides new insights into the genetic regulatory mechanisms underlying human spermatogenesis and the pathogenesis of NOA, and it offers new targets for gene therapy of male infertility.
Collapse
Affiliation(s)
- Chunyun Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Wei Chen
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Dong Zhang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Qingqing Yuan
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200135, China
| | - Xing Yu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province;
Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha 410013, China
| |
Collapse
|
6
|
Kong R, Ma Y, Li W, Xu Z, Gong S, Liu A, Cheng C, Zhang X, Qin J, Li S, Feng J, Jiang J. Zinc finger protein 367 exerts a cancer-promoting role in small cell lung cancer by influencing the CIT/LATS2/YAP signaling cascade. Toxicol Appl Pharmacol 2024; 489:117005. [PMID: 38880190 DOI: 10.1016/j.taap.2024.117005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
A remarkable cancer-related role of zinc finger protein 367 (ZNF367) has been demonstrated in multiple malignancies. However, whether ZNF367 has a role in small-cell lung cancer (SCLC) remains unexplored. The purpose of this work was to explore the potential role and mechanism of ZNF367 in SCLC. In silico analysis using the Gene Expression Omnibus (GEO) dataset revealed high levels of the ZNF367 transcript in SCLC. Examination of clinical tissues confirmed the significant abundance of ZNF367 in SCLC tissues compared with adjacent non-malignant tissues. The genetic depletion of ZNF367 in SCLC cells led to remarkable alterations in cell proliferation, the cell cycle, colony formation and chemosensitivity. Mechanistically, ZNF367 was shown to regulate the activation of yes-associated protein (YAP) associated with the up-regulation of phosphorylated large tumour suppressor kinase 2 (LATS2). Further investigation revealed that ZNF367 affected the LATS2-YAP cascade by regulating the expression of citron kinase (CIT). Re-expression of constitutively active YAP diminished the tumour-inhibiting function of ZNF367 depletion. Xenograft experiments confirmed the tumour-inhibiting effect of ZNF367 depletion in vivo. In summary, our results demonstrate that the inhibition of ZNF367 displays anticancer effects in SCLC by inhibiting YAP activation, suggesting it as a potential druggable oncogenic target.
Collapse
Affiliation(s)
- Ranran Kong
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Thoracic Surgery, Luoyang Hospital, the Second Affiliated Hospital of Xi'an Jiaotong University, Luoyang, Henan 471003, China
| | - Yuefeng Ma
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Wendeng Li
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Zhengshui Xu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Songyu Gong
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Aoran Liu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Chuantao Cheng
- Department of Dermatology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xinwu Zhang
- Department of General Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jie Qin
- Department of Orthopedics, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Shaomin Li
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jie Feng
- Department of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Jiantao Jiang
- Department of Thoracic Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
7
|
Bai M, Liu Y, Liu H, Jia Y, Tian X, Sun C. Production of recombinant human epidermal growth factor fused with HaloTag protein and characterisation of its biological functions. PeerJ 2024; 12:e17806. [PMID: 39035165 PMCID: PMC11259126 DOI: 10.7717/peerj.17806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
Epidermal growth factor (EGF) protein is a crucial biomolecule involved in regulating cell growth, proliferation, migration and differentiation, which is used in various therapeutic applications, such as wound healing and tissue regeneration. The production of recombinant EGF is essential for studying its biological function and for its clinical translation. However, EGF protein expressed in prokaryotic cells often occurs in inclusion bodies, and co-expression with soluble tag protein is an effective method to prepare recombinant EGF. In this study, we expressed recombinant human EGF (rhEGF) fused to a HaloTag (Halo-rhEGF) and a large portion of Halo-rhEGF was found in the soluble fraction. Cell growth assay showed that the purified Halo-rhEGF protein could promote the proliferation of fibroblasts (NIH 3T3) and epithelial cells (HaCaT), and significantly increased their viability. Phosphorylation of the intracellular signaling proteins, ERK1/2 and c-Jun, was stimulated by treatment with Halo-rhEGF and the expression levels of proteins regulating cell proliferation were significantly increased. RNA sequencing analysis revealed that rhEGF could increase the transcription of genes enriched in ribosome generation and cell proliferation. Moreover, Halo-rhEGF can be labelled by HaloTag ligand for fluorescence imaging and can be slowly released in tissue repair by binding to anion biomaterials. In conclusion, HaloTag is an efficient fusion tag for rhEGF protein expression, purification and controlled release, and Halo-rhEGF can promote the proliferation and viability of epithelial and fibroblast cells.
Collapse
Affiliation(s)
- Mengru Bai
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yezhuo Liu
- Favorsun Medical Group, Suzhou, China
- Shanghai Hongdu New Material Science and Technology, Shanghai, China
| | - Hongyin Liu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
- Shanghai Hongdu New Material Science and Technology, Shanghai, China
| |
Collapse
|
8
|
Piechka A, Sparanese S, Witherspoon L, Hach F, Flannigan R. Molecular mechanisms of cellular dysfunction in testes from men with non-obstructive azoospermia. Nat Rev Urol 2024; 21:67-90. [PMID: 38110528 DOI: 10.1038/s41585-023-00837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 12/20/2023]
Abstract
Male factor infertility affects 50% of infertile couples worldwide; the most severe form, non-obstructive azoospermia (NOA), affects 10-15% of infertile males. Treatment for individuals with NOA is limited to microsurgical sperm extraction paired with in vitro fertilization intracytoplasmic sperm injection. Unfortunately, spermatozoa are only retrieved in ~50% of patients, resulting in live birth rates of 21-46%. Regenerative therapies could provide a solution; however, understanding the cell-type-specific mechanisms of cellular dysfunction is a fundamental necessity to develop precision medicine strategies that could overcome these abnormalities and promote regeneration of spermatogenesis. A number of mechanisms of cellular dysfunction have been elucidated in NOA testicular cells. These mechanisms include abnormalities in both somatic cells and germ cells in NOA testes, such as somatic cell immaturity, aberrant growth factor signalling, increased inflammation, increased apoptosis and abnormal extracellular matrix regulation. Future cell-type-specific investigations in identifying modulators of cellular transcription and translation will be key to understanding upstream dysregulation, and these studies will require development of in vitro models to functionally interrogate spermatogenic niche dysfunction in both somatic and germ cells.
Collapse
Affiliation(s)
- Arina Piechka
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Sydney Sparanese
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Luke Witherspoon
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Urology, Department of Surgery, University of Ottawa, Ontario, Canada
| | - Faraz Hach
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.
- Department of Urology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Zhang J, Chen X, Chen G, Wang H, Jia L, Hao Y, Yao D. Identification of a novel PAK1/HDAC6 dual inhibitor ZMF-23 that triggers tubulin-stathmin regulated cell death in triple negative breast cancer. Int J Biol Macromol 2023; 251:126348. [PMID: 37586623 DOI: 10.1016/j.ijbiomac.2023.126348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/04/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most poorly treated subtype of breast cancer, and targeting the heterogeneity of TNBC has emerged as a fascinating therapeutic strategy. In this study, we propose for the first time that dual-targeting PAK1 and HDAC6 is a promising novel strategy for TNBC treatment due to their essential roles in the regulation of energy metabolism and epigenetic modification. We discovered a novel dual-targeting PAK1/HDAC6 inhibitor, 6 - (2-(cyclopropylamino) - 6 - (2,4-dichlorophenyl) - 7 - oxopyrido [2,3-d] pyrimidin - 8 (7H) -yl) - N-hydroxyhexanamide (ZMF-23), which presented profound inhibitory activity against PAK1 and HDAC6 and robust antiproliferative potency in MDA-MB-231 cells. In addition, SPR and CETSA assay demonstrated the targeted binding of ZMF-23 with PAK1/HDAC6. Mechanically, ZMF-23 strongly inhibited the cellular PAK1 and HDAC6 activity, impeded PAK1 and HDAC6 regulated aerobic glycolysis and migration. By RNA-seq analysis, ZMF-23 was found to induce TNF-α-regulated necroptosis, which further enhanced apoptosis. Additionally, ZMF-23 triggered PAK1-tubulin/HDAC6-Stathmin regulated microtubule structure changes, which further induced the G2/M cycle arrest. Moreover, prominent anti-proliferative effect of ZMF-23 was confirmed in the TNBC xenograft zebrafish and mouse model via PAK1 and HDAC6 inhibition. Collectively, ZMF-23 is a novel dual PAK1/HDAC6 inhibitor with TNBC treatment potential.
Collapse
Affiliation(s)
- Jin Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xiya Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Gang Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Hailing Wang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Lin Jia
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Yue Hao
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China.
| | - Dahong Yao
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| |
Collapse
|
10
|
Blanco FC, Bigi MM, García EA, Elola MT, Vázquez CL, Bigi F. A Transcriptional Analysis of Cattle Immune Cells Reveals a Central Role of Type 1 Interferon in the In Vitro Innate Immune Response against Mycobacterium bovis. Pathogens 2023; 12:1159. [PMID: 37764968 PMCID: PMC10536033 DOI: 10.3390/pathogens12091159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Bovine tuberculosis is a chronic infectious disease primarily caused by Mycobacterium bovis, a bacterium that affects cattle and other mammals, including humans. Despite the availability of vast research about the immune response mechanisms of human tuberculosis caused by Mycobacterium tuberculosis, the knowledge of bovine tuberculosis's immunology, particularly regarding the innate immune response, still remains scarce. In this study, we compared the transcriptome of cell cultures containing lymphocytes and M. bovis infected-macrophages with two strains of variable virulence, the virulent Mb04-303 strain and the attenuated Mb534. To that end, we infected bovine macrophages at a multiplicity of infection of one, and co-cultured the infections with autologous lymphocytes. RNA obtained from the co-cultures was sequenced to identify differentially expressed gene pathways by using the database Reactome. The RNA-seq analysis showed that the Mb04-303 infection upregulated the type 1 interferon signalling pathway, while it downregulated the KEAP1-NFE2L2 pathway. According to the literature, this last pathway is involved in the activation of antioxidant genes and inflammasome. In addition, the macrophages infected with Mb04-303 recruited more Galectin 8 than those infected with Mb534. This result indicates that Mb04-303 induced higher phagosome membrane damage, with the possible concomitant release of bacterial compounds into the cytoplasm that activates the type I signalling pathway. Altogether, Mb04-303 repressed the antioxidant and anti-inflammatory responses, likely impairing interleukin-1β activation, and trigged the canonical type 1 interferon signalling. Although these responses led to the control of bacterial replication during early infection, the virulent strain eventually managed to establish a successful infection.
Collapse
Affiliation(s)
- Federico Carlos Blanco
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina; (F.C.B.); (E.A.G.)
- Instituto de Biotecnología, CICVyA, Instituto Nacional de Tecnología Agropecuaria, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina
| | - María Mercedes Bigi
- Instituto de Investigaciones Biomédicas (UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires 1417, Argentina;
| | - Elizabeth Andrea García
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina; (F.C.B.); (E.A.G.)
- Instituto de Biotecnología, CICVyA, Instituto Nacional de Tecnología Agropecuaria, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina
| | - María Teresa Elola
- Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro Paladini (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113, Argentina
| | - Cristina Lourdes Vázquez
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina; (F.C.B.); (E.A.G.)
- Instituto de Biotecnología, CICVyA, Instituto Nacional de Tecnología Agropecuaria, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina
| | - Fabiana Bigi
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), INTA-CONICET, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina; (F.C.B.); (E.A.G.)
- Instituto de Biotecnología, CICVyA, Instituto Nacional de Tecnología Agropecuaria, N. Repetto and De los Reseros, Buenos Aires 1686, Argentina
| |
Collapse
|
11
|
Ahmed SBM, Radwan N, Amer S, Saheb Sharif-Askari N, Mahdami A, Samara KA, Halwani R, Jelinek HF. Assessing the Link between Diabetic Metabolic Dysregulation and Breast Cancer Progression. Int J Mol Sci 2023; 24:11816. [PMID: 37511575 PMCID: PMC10380477 DOI: 10.3390/ijms241411816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetes mellitus is a burdensome disease that affects various cellular functions through altered glucose metabolism. Several reports have linked diabetes to cancer development; however, the exact molecular mechanism of how diabetes-related traits contribute to cancer progression is not fully understood. The current study aimed to explore the molecular mechanism underlying the potential effect of hyperglycemia combined with hyperinsulinemia on the progression of breast cancer cells. To this end, gene dysregulation induced by the exposure of MCF7 breast cancer cells to hyperglycemia (HG), or a combination of hyperglycemia and hyperinsulinemia (HGI), was analyzed using a microarray gene expression assay. Hyperglycemia combined with hyperinsulinemia induced differential expression of 45 genes (greater than or equal to two-fold), which were not shared by other treatments. On the other hand, in silico analysis performed using a publicly available dataset (GEO: GSE150586) revealed differential upregulation of 15 genes in the breast tumor tissues of diabetic patients with breast cancer when compared with breast cancer patients with no diabetes. SLC26A11, ALDH1A3, MED20, PABPC4 and SCP2 were among the top upregulated genes in both microarray data and the in silico analysis. In conclusion, hyperglycemia combined with hyperinsulinemia caused a likely unique signature that contributes to acquiring more carcinogenic traits. Indeed, these findings might potentially add emphasis on how monitoring diabetes-related metabolic alteration as an adjunct to diabetes therapy is important in improving breast cancer outcomes. However, further detailed studies are required to decipher the role of the highlighted genes, in this study, in the pathogenesis of breast cancer in patients with a different glycemic index.
Collapse
Affiliation(s)
- Samrein B M Ahmed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Health, Wellbeing and Life Sciences, Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield S1 1WB, UK
| | - Nada Radwan
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sara Amer
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amena Mahdami
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Kamel A Samara
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rabih Halwani
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Herbert F Jelinek
- Department of Biomedical Engineering and Health Engineering Innovation Center, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
12
|
Wu S, Cheng Z, Peng Y, Cao Y, He Z. GPx3 knockdown inhibits the proliferation and DNA synthesis and enhances the early apoptosis of human spermatogonial stem cells via mediating CXCL10 and cyclin B1. Front Cell Dev Biol 2023; 11:1213684. [PMID: 37484915 PMCID: PMC10361659 DOI: 10.3389/fcell.2023.1213684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Spermatogenesis is regulated by genetic and epigenetic factors. However, the genes and signaling pathways mediating human spermatogenesis remain largely unknown. Here, we have for the first time explored the expression, function, and mechanism of glutathione peroxidase 3 (GPx3) in controlling the proliferation and apoptosis of human spermatogonial stem cells (SSCs). We found that GPx3 was expressed in human SSCs. Notably, we revealed that GPx3 knockdown resulted in the decrease in the proliferation, DNA synthesis, and cyclin B1 level in human SSC lines, which possessed the phenotypic features of human primary SSCs. Flow cytometry and TUNEL assays showed that GPx3 silencing led to enhancement of early apoptosis of human SSC line. RNA sequencing was utilized to identify CXCL10 as a target of GPx3 in human SSCs, and notably, both double immunostaining and co-immunoprecipitation (co-IP) demonstrated that there was an association between GPx3 and CXCL10 in these cells. CXCL10-shRNA resulted in the reduction in the proliferation and DNA synthesis of human SSC line and an increase in apoptosis of these cells. Taken together, these results implicate that GPx3 regulates the proliferation, DNA synthesis, and early apoptosis of human SSC line via mediating CXCL10 and cyclin B1. This study, thus, offers a novel insight into the molecular mechanism regulating the fate determinations of human SSCs and human spermatogenesis.
Collapse
Affiliation(s)
- Si Wu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Zixin Cheng
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Ye Peng
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Ying Cao
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, China
- The Research Center of Reproduction and Translational Medicine of Hunan Province, The Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
13
|
Ribeiro J, Crossan GP. GCNA is a histone binding protein required for spermatogonial stem cell maintenance. Nucleic Acids Res 2023; 51:4791-4813. [PMID: 36919611 PMCID: PMC10250205 DOI: 10.1093/nar/gkad168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/01/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Recycling and de-novo deposition of histones during DNA replication is a critical challenge faced by eukaryotic cells and is coordinated by histone chaperones. Spermatogenesis is highly regulated sophisticated process necessitating not only histone modification but loading of testis specific histone variants. Here, we show that Germ Cell Nuclear Acidic protein (GCNA), a germ cell specific protein in adult mice, can bind histones and purified GCNA exhibits histone chaperone activity. GCNA associates with the DNA replication machinery and supports progression through S-phase in murine undifferentiated spermatogonia (USGs). Whilst GCNA is dispensable for embryonic germ cell development, it is required for the maintenance of the USG pool and for long-term production of sperm. Our work describes the role of a germ cell specific histone chaperone in USGs maintenance in mice. These findings provide a mechanistic basis for the male infertility observed in patients carrying GCNA mutations.
Collapse
|
14
|
Cui Y, Chen W, Du L, He Z. OIP5 Interacts with NCK2 to Mediate Human Spermatogonial Stem Cell Self-Renewal and Apoptosis through Cell Cyclins and Cycle Progression and Its Abnormality Is Correlated with Male Infertility. RESEARCH (WASHINGTON, D.C.) 2023; 6:0162. [PMID: 37292517 PMCID: PMC10246317 DOI: 10.34133/research.0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/14/2023] [Indexed: 06/10/2023]
Abstract
Spermatogonial stem cells (SSCs) have important applications in both reproduction and regenerative medicine. Nevertheless, specific genes and signaling transduction pathways in mediating fate decisions of human SSCs remain elusive. Here, we have demonstrated for the first time that OIP5 (Opa interacting protein 5) controlled the self-renewal and apoptosis of human SSCs. RNA sequencing identified that NCK2 was a target for OIP5 in human SSCs, and interestingly, OIP5 could interact with NCK2 as shown by Co-IP (co-immunoprecipitation), IP-MS (mass spectrometry), and GST pulldown assays. NCK2 silencing decreased human SSC proliferation and DNA synthesis but enhanced their apoptosis. Notably, NCK2 knockdown reversed the influence of OIP5 overexpression on human SSCs. Moreover, OIP5 inhibition decreased the numbers of human SSCs at S and G2/M phases, while the levels of numerous cell cycle proteins, including cyclins A2, B1, D1, E1 and H, especially cyclin D1, were remarkably reduced. Significantly, whole-exome sequencing of 777 patients with nonobstructive azoospermia (NOA) revealed 54 single-nucleotide polymorphism mutations of the OIP5 gene (6.95%), while the level of OIP5 protein was obviously lower in testes of NOA patients compared to fertile men. Collectively, these results implicate that OIP5 interacts with NCK2 to modulate human SSC self-renewal and apoptosis via cell cyclins and cell cycle progression and that its mutation and/or lower expression is correlated with azoospermia. As such, this study offers novel insights into molecular mechanisms underlying the fate determinations of human SSCs and the pathogenesis of NOA, and it provides new targets for treating male infertility.
Collapse
|
15
|
Li N, Zhou Q, Yi Z, Zhang H, Zhou D. Ubiquitin protein E3 ligase ASB9 suppresses proliferation and promotes apoptosis in human spermatogonial stem cell line by inducing HIF1AN degradation. Biol Res 2023; 56:4. [PMID: 36683111 PMCID: PMC9869568 DOI: 10.1186/s40659-023-00413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Spermatogonial stem cells (SSCs) are critical for sustaining spermatogenesis. Even though several regulators of SSC have been identified in rodents, the regulatory mechanism of SSC in humans has yet to be discovered. METHODS To explore the regulatory mechanisms of human SSCs, we analyzed publicly available human testicular single-cell sequencing data and found that Ankyrin repeat and SOCS box protein 9 (ASB9) is highly expressed in SSCs. We examined the expression localization of ASB9 using immunohistochemistry and overexpressed ASB9 in human SSC lines to explore its role in SSC proliferation and apoptosis. Meanwhile, we used immunoprecipitation to find the target protein of ASB9 and verified its functions. In addition, we examined the changes in the distribution of ASB9 in non-obstructive azoospermia (NOA) patients using Western blot and immunofluorescence. RESULTS The results of uniform manifold approximation and projection (UMAP) clustering and pseudotime analysis showed that ASB9 was highly expressed in SSCs, and its expression gradually increased during development. The immunohistochemical and dual-color immunofluorescence results displayed that ASB9 was mainly expressed in nonproliferating SSCs. Overexpression of ASB9 in the SSC line revealed significant inhibition of cell proliferation and increased apoptosis. We predicted the target proteins of ASB9 and verified that hypoxia-inducible factor 1-alpha inhibitor (HIF1AN), but not creatine kinase B-type (CKB), has a direct interaction with ASB9 in human SSC line using protein immunoprecipitation experiments. Subsequently, we re-expressed HIF1AN in ASB9 overexpressing cells and found that HIF1AN reversed the proliferative and apoptotic changes induced by ASB9 overexpression. In addition, we found that ABS9 was significantly downregulated in some NOA patients, implying a correlation between ASB9 dysregulation and impaired spermatogenesis. CONCLUSION ASB9 is predominantly expressed in human SSCs, it affects the proliferation and apoptotic process of the SSC line through HIF1AN, and its abnormal expression may be associated with NOA.
Collapse
Affiliation(s)
- Ning Li
- grid.216417.70000 0001 0379 7164Operating Department of Xiangya Hospital, Central South University, Changsha, 410008 Hunan China ,grid.216417.70000 0001 0379 7164Xiangya Nursing School, Central South University, Changsha, 410013 Hunan China
| | - Qianyin Zhou
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China
| | - Zhang Yi
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China
| | - Huan Zhang
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China
| | - Dai Zhou
- grid.477823.d0000 0004 1756 593XReproductive & Genetic Hospital of CITIC-Xiangya, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science, Central South University, Changsha, 410013 Hunan China ,grid.411427.50000 0001 0089 3695College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan China ,Clinical Research Center for Reproduction and Genetics in Hunan Province, Changsha, 410021 Hunan China ,grid.216417.70000 0001 0379 7164NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, 410013 Hunan China
| |
Collapse
|
16
|
Rahbar M, Asadpour R, Azami M, Mazaheri Z, Hamali H. Improving the process of spermatogenesis in azoospermic mice using spermatogonial stem cells co-cultured with epididymosomes in three-dimensional culture system. Life Sci 2022; 310:121057. [DOI: 10.1016/j.lfs.2022.121057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/09/2022]
|
17
|
Li F, Tan B, Chen Z, Zhao Q, Li S, Ding P, Liu C, Wang X, Li X, Li Y. Long non-coding RNA CNALPTC1 promotes gastric cancer progression by regulating the miR-6788-5p/PAK1 pathway. J Gastrointest Oncol 2022; 13:2809-2822. [PMID: 36636079 PMCID: PMC9830357 DOI: 10.21037/jgo-22-1069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Background Gastric cancer (GC) is a globally prevalent gastrointestinal tumor. Long non-coding RNAs (lncRNAs) are a new type of transcript which has become a hotspot of current research; however, the function of most lncRNAs in the advancement of GC is still not clear. The focus of this research was to elucidate the role and expression of lncRNA CNALPTC1 in GC. Methods In GC cells and tissues, the detection of CNALPTC1 expression was carried out using quantitative real-time polymerase chain reaction (qRT-PCR), and the link between its expression and clinicopathological features was investigated. The impacts of inhibition and upregulation of CNALPTC1 on the physiological behavior of GC cells were observed. Furthermore, through bioinformatics analysis and prediction of microRNA (miRNA) targeted to CNALPTC1 and target genes interacting with miRNA, the effects on invasion, proliferation, and migration of GC cells were investigated. Results The elevated expression level of CNALPTC1 was observed in GC tissues and cell lines. The in vitro analysis indicated that gene silencing of CNALPTC1 resulted in inhibition, whereas upregulation of CNALPTC1 resulted in the promotion of invasion, proliferation, and migration of GC cells, respectively. In addition, we observed that CNALPTC1 functions as a molecular sponge for miR-6788-5p, and the level of expression of CNALPTC1 exhibited a negative correlation with miR-6788-5p. Moreover, it was revealed that the miR-6788-5p's direct target was PAK1, which could reverse the inhibitory function of miR-6788-5p. Conclusions Our research revealed that the CNALPTC1 promotes GC development by negatively regulating the miR-6788-5p/PAK1 pathway. GC therapy may be improved by conducting targeted studies of the CNALPTC1/miR-6788-5p/PAK1 axis.
Collapse
Affiliation(s)
- Fang Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bibo Tan
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zihao Chen
- Department of Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shi Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pingan Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chang Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoxiao Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoya Li
- Department of Scientific Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
18
|
MAP4K4/JNK Signaling Pathway Stimulates Proliferation and Suppresses Apoptosis of Human Spermatogonial Stem Cells and Lower Level of MAP4K4 Is Associated with Male Infertility. Cells 2022; 11:cells11233807. [PMID: 36497065 PMCID: PMC9739186 DOI: 10.3390/cells11233807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Spermatogonial stem cells (SSCs) serve as a foundation for spermatogenesis and they are essential for male fertility. The fate of SSC is determined by genetic and epigenetic regulatory networks. Many molecules that regulate SSC fate determinations have been identified in mice. However, the molecules and signaling pathways underlying human SSCs remain largely unclear. In this study, we have demonstrated that MAP4K4 was predominantly expressed in human UCHL1-positive spermatogonia by double immunocytochemical staining. MAP4K4 knockdown inhibited proliferation of human SSCs and induced their apoptosis. Moreover, MAP4K4 silencing led to inhibition of JNK phosphorylation and MAP4K4 phosphorylation at Ser801. RNA sequencing indicated that MAP4K4 affected the transcription of SPARC, ADAM19, GPX7, GNG2, and COLA1. Interestingly, the phenotype of inhibiting JNK phosphorylation by SP600125 was similar to MAP4K4 knockdown. Notably, MAP4K4 protein was lower in the testes of patients with non-obstructive azoospermia than those with normal spermatogenesis as shown by Western blots and immunohistochemistry. Considered together, our data implicate that MAP4K4/JNK signaling pathway mediates proliferation and apoptosis of human SSCs, which provides a novel insight into molecular mechanisms governing human spermatogenesis and might offer new targets for gene therapy of male infertility.
Collapse
|
19
|
Luo C, Wang Z, Wang J, Yun F, Lu F, Fu J, Liu Q, Shi D. Individual variation in buffalo somatic cell cloning efficiency is related to glycolytic metabolism. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2076-2092. [PMID: 35366153 DOI: 10.1007/s11427-021-2039-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Mammalian individuals differ in their somatic cell cloning efficiency, but the mechanisms leading to this variation is poorly understood. Here we found that high cloning efficiency buffalo fetal fibroblasts (BFFs) displayed robust energy metabolism, looser chromatin structure, high H3K9 acetylation and low heterochromatin protein 1α (HP1α) expression. High cloning efficiency BFFs had more H3K9ac regions near to the upstream of glycolysis genes by ChIP-seq, and involved more openness loci related to glycolysis genes through ATAC-seq. The expression of these glycolysis genes was also found to be higher in high cloning efficiency BFFs by qRT-PCR. Two key enzymes of glycolysis, PDKs and LDH, were confirmed to be associated with histone acetylation and chromatin openness of BFFs. Treatment of low cloning efficiency BFFs with PS48 (activator of PDK1) resulted in an increase in the intracellular lactate production and H3K9 acetylation, decrease in histone deacetylase activity and HP1α expression, less condensed chromatin structure and more cloning embryos developing to blastocysts. These results indicate that the cloning efficiency of buffalo somatic cells is associated with their glycolytic metabolism and chromatin structure, and can be improved by increasing glycolytic metabolism.
Collapse
Affiliation(s)
- Chan Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Zhiqiang Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
| | - Jinling Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Feng Yun
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Fenghua Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Jiayuan Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Qingyou Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, 530005, China.
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, China.
| |
Collapse
|
20
|
Estrada-Meza C, Torres-Copado A, Loreti González-Melgoza L, Ruiz-Manriquez LM, De Donato M, Sharma A, Pathak S, Banerjee A, Paul S. Recent insights into the microRNA and long non-coding RNA-mediated regulation of stem cell populations. 3 Biotech 2022; 12:270. [PMID: 36101546 PMCID: PMC9464284 DOI: 10.1007/s13205-022-03343-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/29/2022] [Indexed: 12/19/2022] Open
Abstract
Stem cells are undifferentiated cells that have multi-lineage differentiation. The transition from self-renewal to differentiation requires rapid and extensive gene expression alterations. Since different stem cells exhibit diverse non-coding RNAs (ncRNAs) expression profiles, the critical roles of ncRNAs in stem cell reprogramming, pluripotency maintenance, and differentiation have been widely investigated over the past few years. Hence, in this current review, the two main categories of ncRNAs, microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are discussed. While the primary way by which miRNAs restrict mRNA transcription is through miRNA-mRNA interaction, lncRNAs have a wide range of effects on mRNA functioning, including interactions with miRNAs. Both of these ncRNAs participate in the post-transcriptional regulation of crucial biological mechanisms, such as cell cycle regulation, apoptosis, aging, and cell fate decisions. These findings shed light on a previously unknown aspect of gene regulation in stem cell fate determination and behavior. Overall, we summarized the key roles of miRNAs (including exosomal miRNAs) and lncRNAs in the regulation of stem cell populations, such as cardiac, hematopoietic, mesenchymal, neural, and spermatogonial, as well ncRNAs' influence on malignancy through modulating cancer stem cells, which might significantly contribute to clinical stem cell therapy and in regenerative medicine.
Collapse
Affiliation(s)
- Carolina Estrada-Meza
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Andrea Torres-Copado
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Luisa Loreti González-Melgoza
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Luis M. Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Marcos De Donato
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Ashutosh Sharma
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, CP 76130 Queretaro, Mexico
| |
Collapse
|
21
|
Bashiri Z, Gholipourmalekabadi M, Falak R, Amiri I, Asgari H, Chauhan NPS, Koruji M. In vitro production of mouse morphological sperm in artificial testis bioengineered by 3D printing of extracellular matrix. Int J Biol Macromol 2022; 217:824-841. [PMID: 35905760 DOI: 10.1016/j.ijbiomac.2022.07.127] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 11/30/2022]
Abstract
Since autologous stem cell transplantation is prone to cancer recurrence, in vitro sperm production is regarded a safer approach to fertility preservation. In this study, the spermatogenesis process on testicular tissue extracellular matrix (T-ECM)-derived printing structure was evaluated. Ram testicular tissue was decellularized using a hypertonic solution containing triton and the extracted ECM was used as a bio-ink to print an artificial testis. Following cell adhesion and viability examination, pre-meiotic and post-meiotic cells in the study groups (as testicular suspension and co-culture with Sertoli cells) were confirmed by real-time PCR, flow-cytometry and immunocytochemistry methods. Morphology of differentiated cells was evaluated using transmission electron microscopy (TEM), toluidine blue, Giemsa, and hematoxylin and eosin (H&E) staining. The functionality of Leydig and Sertoli cells was determined by their ability for hormone secretion. The decellularization of testicular tissue fragments was successful and had efficiently removed the cellular debris and preserved the ECM compounds. High cell viability, colonization, and increased expression of pre-meiotic markers in cultured testicular cells on T-ECM-enriched scaffolds confirmed their proliferation. Furthermore, the inoculation of neonatal mouse testicular cells onto T-ECM-enriched scaffolds resulted in the generation of sperm. Morphology evaluation showed that the structure of these cells was quite similar to mature sperm with a specialized tail structure. The hormonal analysis also confirmed production and secretion of testosterone and inhibin B by Leydig and Sertoli cells. T-ECM printed artificial testis is a future milestone that promises for enhancing germ cell maintenance and differentiation, toxicology studies, and fertility restoration to pave the way for new human infertility treatments in the future.
Collapse
Affiliation(s)
- Zahra Bashiri
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center (IRC), Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Iraj Amiri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Endometrium and Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamidreza Asgari
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Morteza Koruji
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Elcombe CS, Monteiro A, Elcombe MR, Ghasemzadeh-Hasankolaei M, Sinclair KD, Lea R, Padmanabhan V, Evans NP, Bellingham M. Developmental exposure to real-life environmental chemical mixture programs a testicular dysgenesis syndrome-like phenotype in prepubertal lambs. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 94:103913. [PMID: 35738462 PMCID: PMC9554787 DOI: 10.1016/j.etap.2022.103913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 05/30/2023]
Abstract
Current declines in male reproductive health may, in part, be driven by anthropogenic environmental chemical (EC) exposure. Using a biosolids treated pasture (BTP) sheep model, this study examined the effects of gestational exposure to a translationally relevant EC mixture. Testes of 8-week-old ram lambs from mothers exposed to BTP during pregnancy contained fewer germ cells and had a greater proportion of Sertoli-cell-only seminiferous tubules. This concurs with previous published data from fetuses and neonatal lambs from mothers exposed to BTP. Comparison between the testicular transcriptome of biosolids lambs and human testicular dysgenesis syndrome (TDS) patients indicated common changes in genes involved in apoptotic and mTOR signalling. Gene expression data and immunohistochemistry indicated increased HIF1α activation and nuclear localisation in Leydig cells of BTP exposed animals. As HIF1α is reported to disrupt testosterone synthesis, these results provide a potential mechanism for the pathogenesis of this testicular phenotype, and TDS in humans.
Collapse
Affiliation(s)
- Chris S Elcombe
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK; School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK.
| | - Ana Monteiro
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Matthew R Elcombe
- MicroMatrices Associates Ltd, Dundee Technopole, James Lindsay Place, Dundee, UK
| | - Mohammad Ghasemzadeh-Hasankolaei
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Kevin D Sinclair
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | - Richard Lea
- University of Nottingham, Sutton Bonington Campus, Loughborough, UK
| | | | - Neil P Evans
- Institute of Biodiversity Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Michelle Bellingham
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK.
| |
Collapse
|
23
|
Du L, Chen W, Li C, Cui Y, He Z. RNF144B stimulates the proliferation and inhibits the apoptosis of human spermatogonial stem cells via the FCER2/NOTCH2/HES1 pathway and its abnormality is associated with azoospermia. J Cell Physiol 2022; 237:3565-3577. [PMID: 35699595 DOI: 10.1002/jcp.30813] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/05/2022] [Accepted: 05/30/2022] [Indexed: 11/08/2022]
Abstract
Studies on gene regulation and signaling transduction pathways of human spermatogonial stem cells (SSCs) are of the utmost significance for unveiling molecular mechanisms underlying human spermatogenesis and gene therapy of male infertility. We have demonstrated, for the first time, that RNF144B stimulated cell proliferation and inhibited the apoptosis of human SSCs. The target of RNF144B was identified as FCER2 by RNA sequencing. We revealed that RNF144B interacted with FCER2 by immunoprecipitation. Consistently, overexpression of FCER2 reversed the phenotype of proliferation and apoptosis of human SSCs caused by RNF144B knockdown. Interestingly, FCER2 pulled down N2ICD (NOTCH2 intracellular domain), while N2ICD could bind to FCER2 in human SSCs. The levels of NOTCH2, FCER2, HES1, and HEY1 were reduced by RNF144B siRNA in human SSCs. Significantly, RNF144B was expressed at a lower level in nonobstructive azoospermia (NOA) patients than in the obstructive azoospermia (OA) patients with normal spermatogenesis, and 52 patients with heterozygous mutations of RNF144B were detected in 1,000 NOA patients. These results implicate that RNF144B promotes the proliferation of human SSCs and suppresses their apoptosis via the FCER2/NOTCH2/HES1 pathway and that the abnormality of RNF144B is associated with spermatogenesis failure. This study thus provides novel molecular mechanisms regulating the fate determinations of human SSCs, and it offers new biomarkers for the diagnosis and treatment of male infertility.
Collapse
Affiliation(s)
- Li Du
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University; The Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Wei Chen
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University; The Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Chunyun Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University; The Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Yinghong Cui
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University; The Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University; The Manufacture-Based Learning and Research Demonstration Center for Human Reproductive Health New Technology of Hunan Normal University, Changsha, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Shah K, Kazi JU. Phosphorylation-Dependent Regulation of WNT/Beta-Catenin Signaling. Front Oncol 2022; 12:858782. [PMID: 35359365 PMCID: PMC8964056 DOI: 10.3389/fonc.2022.858782] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/16/2022] [Indexed: 01/11/2023] Open
Abstract
WNT/β-catenin signaling is a highly complex pathway that plays diverse roles in various cellular processes. While WNT ligands usually signal through their dedicated Frizzled receptors, the decision to signal in a β-catenin-dependent or -independent manner rests upon the type of co-receptors used. Canonical WNT signaling is β-catenin-dependent, whereas non-canonical WNT signaling is β-catenin-independent according to the classical definition. This still holds true, albeit with some added complexity, as both the pathways seem to cross-talk with intertwined networks that involve the use of different ligands, receptors, and co-receptors. β-catenin can be directly phosphorylated by various kinases governing its participation in either canonical or non-canonical pathways. Moreover, the co-activators that associate with β-catenin determine the output of the pathway in terms of induction of genes promoting proliferation or differentiation. In this review, we provide an overview of how protein phosphorylation controls WNT/β-catenin signaling, particularly in human cancer.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Julhash U. Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
- *Correspondence: Julhash U. Kazi,
| |
Collapse
|
25
|
Salazar-Anzures T, Pastén-Hidalgo K, Sicilia-Argumedo G, Riverón-Negrete L, Hernández-Vázquez ADJ, Fernanadez-Mejia C. Dietary biotin supplementation increases proliferation pathways in mice testes without affecting serum follicle-stimulating hormone levels and stem cell factor expression. Toxicol Appl Pharmacol 2021; 433:115774. [PMID: 34699867 DOI: 10.1016/j.taap.2021.115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 11/18/2022]
Abstract
Supplements containing pharmacological concentrations of biotin are commercially available. The mechanisms by which biotin at pharmacological concentrations exerts its action have been the subject of multiple investigations, particularly for biotin's medicinal potential and wide use for cosmetic purposes. Several studies have reported that biotin supplementation increases cell proliferation; however, the mechanisms involved in this effect have not yet been characterized. In a previous study, we found that a biotin-supplemented diet increased spermatogonia proliferation. The present study was focused on investigating the molecular mechanisms involved in biotin-induced testis cell proliferation. Male BALB/cAnNHsd mice were fed a control or a biotin-supplemented diet (1.76 or 97.7 mg biotin/kg diet) for eight weeks. Compared with the control group, the biotin-supplemented mice presented augmented protein abundance of the c-kit-receptor and pERK1/2Tyr204 and pAKTSer473, the active forms of ERK/AKT proliferation signaling pathways. No changes were observed in the testis expression of the stem cell factor and in the serum levels of the follicle-stimulating hormone. Analysis of mRNA abundance found an increase in cyclins Ccnd3, Ccne1, Ccna2; Kinases Cdk4, Cdk2; and E2F; and Sp1 & Sp3 transcription factors. Decreased expression of cyclin-dependent kinase inhibitor 1a (p21) was observed but not of Cdkn2a inhibitor (p16). The results of the present study identifies, for the first time, the mechanisms associated with biotin supplementation-induced cell proliferation, which raises concerns about the effects of biotin on male reproductive health because of its capacity to cause hyperplasia, especially because this vitamin is available in large amounts without regulation.
Collapse
Affiliation(s)
- Tonatiuh Salazar-Anzures
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Avenida del Iman#1, 4th floor, Mexico City 04500, Mexico
| | - Karina Pastén-Hidalgo
- Cátedra CONACYT, Instituto Nacional de Pediatría, Avenida del Iman#1, 4th floor, Mexico City 04500, Mexico
| | - Gloria Sicilia-Argumedo
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Avenida del Iman#1, 4th floor, Mexico City 04500, Mexico
| | - Leticia Riverón-Negrete
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Avenida del Iman#1, 4th floor, Mexico City 04500, Mexico
| | - Alain de Jesús Hernández-Vázquez
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Avenida del Iman#1, 4th floor, Mexico City 04500, Mexico
| | - Cristina Fernanadez-Mejia
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Avenida del Iman#1, 4th floor, Mexico City 04500, Mexico.
| |
Collapse
|
26
|
Huang ZH, Huang C, Ji XR, Zhou WJ, Luo XF, Liu Q, Tang YL, Gong F, Zhu WB. MKK7-mediated phosphorylation of JNKs regulates the proliferation and apoptosis of human spermatogonial stem cells. World J Stem Cells 2021; 13:1797-1812. [PMID: 34909124 PMCID: PMC8641020 DOI: 10.4252/wjsc.v13.i11.1797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/28/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human spermatogonial stem cells (SSCs) are the basis of spermatogenesis. However, little is known about the developmental regulatory mechanisms of SSC due to sample origin and species differences.
AIM To investigates the mechanisms involved in the proliferation of human SSC.
METHODS The expression of mitogen-activated protein kinase kinase 7 (MKK7) in human testis was identified using immunohistochemistry and western blotting (WB). MKK7 was knocked down using small interfering RNA, and cell proliferation and apoptosis were detected by WB, EdU, cell counting kit-8 and fluorescence-activated cell sorting. After bioinformatic analysis, the interaction of MKK7 with c-Jun N-terminal kinases ( JNKs ) was verified by protein co-immunoprecipitation and WB. The phosphorylation of JNKs was inhibited by SP600125, and the phenotypic changes were detected by WB, cell counting kit-8 and fluorescence-activated cell sorting.
RESULTS MKK7 is mainly expressed in human SSCs, and MKK7 knockdown inhibits SSC proliferation and promotes their apoptosis. MKK7 mediated the phosphorylation of JNKs, and after inhibiting the phosphorylation of JNKs, the phenotypic changes of the cells were similar to those after MKK7 downregulation. The expression of MKK7 was significantly downregulated in patients with abnormal spermatogenesis, suggesting that abnormal MKK7 may be associated with spermatogenesis impairment.
CONCLUSION MKK7 regulates the proliferation and apoptosis of human SSC by mediating the phosphorylation of JNKs.
Collapse
Affiliation(s)
- Zeng-Hui Huang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
- Department of Reproductive Center, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Chuan Huang
- Department of Sperm Bank, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha 410008, Hunan Province, China
| | - Xi-Ren Ji
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Jun Zhou
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Xue-Feng Luo
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Qian Liu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Yu-Lin Tang
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Fei Gong
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| | - Wen-Bing Zhu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
27
|
Lei T, Gao Y, Duan Y, Cui C, Zhang L, Si M. Inhibition of zinc finger protein 367 exerts a tumor suppressive role in colorectal cancer by affecting the activation of oncogenic YAP1 signaling. ENVIRONMENTAL TOXICOLOGY 2021; 36:2278-2290. [PMID: 34351699 DOI: 10.1002/tox.23341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/12/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
Zinc finger protein 367 (ZNF367) has been documented as a new cancer-related protein that exerts a pivotal role in the carcinogenesis of multiple cancers. However, whether ZNF367 plays a role in colorectal cancer has not been fully understood. Our data showed that ZNF367 expression was higher in colorectal cancer. Depletion of ZNF367 weakened the proliferative and invasive capabilities of colorectal cancer cells. Up-regulation of ZNF367 enhanced the in vitro malignant features of colorectal cancer cells. Knockdown of ZNF367 impeded the activation of Yes-associated protein (YAP1). Reactivation of YAP1 reversed the ZNF367-knockdown-mediated anticancer effects. Suppression of YAP1 significantly abolished ZNF367-overexpression-induced tumor-promotion effects. Depletion of ZNF367 repressed the tumorigenicity of colorectal cancer cells in vivo. These findings demonstrate that ZNF367 is overexpressed in colorectal cancer and acts as a potential tumor-promoter that contributes to the proliferation and invasion of colorectal cancer by enhancing the activation of YAP1 signaling.
Collapse
Affiliation(s)
- Ting Lei
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of General Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ya Gao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuhong Duan
- Endocrinology Department, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chunli Cui
- Department of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Li Zhang
- Institutional Pharmacy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Mingming Si
- Department of General Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
28
|
Binsila B, Selvaraju S, Ranjithkumaran R, Archana SS, Krishnappa B, Ghosh SK, Kumar H, Subbarao RB, Arangasamy A, Bhatta R. Current scenario and challenges ahead in application of spermatogonial stem cell technology in livestock. J Assist Reprod Genet 2021; 38:3155-3173. [PMID: 34661801 DOI: 10.1007/s10815-021-02334-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022] Open
Abstract
PURPOSE Spermatogonial stem cells (SSCs) are the source for the mature male gamete. SSC technology in humans is mainly focusing on preserving fertility in cancer patients. Whereas in livestock, it is used for mining the factors associated with male fertility. The review discusses the present status of SSC biology, methodologies developed for in vitro culture, and challenges ahead in establishing SSC technology for the propagation of superior germplasm with special reference to livestock. METHOD Published literatures from PubMed and Google Scholar on topics of SSCs isolation, purification, characterization, short and long-term culture of SSCs, stemness maintenance, epigenetic modifications of SSCs, growth factors, and SSC cryopreservation and transplantation were used for the study. RESULT The fine-tuning of SSC isolation and culture conditions with special reference to feeder cells, growth factors, and additives need to be refined for livestock. An insight into the molecular mechanisms involved in maintaining stemness and proliferation of SSCs could facilitate the dissemination of superior germplasm through transplantation and transgenesis. The epigenetic influence on the composition and expression of the biomolecules during in vitro differentiation of cultured cells is essential for sustaining fertility. The development of surrogate males through gene-editing will be historic achievement for the foothold of the SSCs technology. CONCLUSION Detailed studies on the species-specific factors regulating the stemness and differentiation of the SSCs are required for the development of a long-term culture system and in vitro spermatogenesis in livestock. Epigenetic changes in the SSCs during in vitro culture have to be elucidated for the successful application of SSCs for improving the productivity of the animals.
Collapse
Affiliation(s)
- Balakrishnan Binsila
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India.
| | - Sellappan Selvaraju
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Rajan Ranjithkumaran
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Santhanahalli Siddalingappa Archana
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Balaganur Krishnappa
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Subrata Kumar Ghosh
- Animal Reproduction Division, Indian Council of Agricultural Research-Indian Veterinary Research Institute, Izatnagar, 243 122, India
| | - Harendra Kumar
- Animal Reproduction Division, Indian Council of Agricultural Research-Indian Veterinary Research Institute, Izatnagar, 243 122, India
| | - Raghavendra B Subbarao
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Arunachalam Arangasamy
- Reproductive Physiology Laboratory, Animal Physiology Division, Indian Council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| | - Raghavendra Bhatta
- Indian council of Agricultural Research-National Institute of Animal Nutrition and Physiology, Bengaluru, 560 030, India
| |
Collapse
|
29
|
Chen W, Cui Y, Ning M, Zhang H, Yin C, He Z. The mechanisms and functions of microRNAs in mediating the fate determinations of human spermatogonial stem cells and Sertoli cells. Semin Cell Dev Biol 2021; 121:32-39. [PMID: 34034987 DOI: 10.1016/j.semcdb.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 01/12/2023]
Abstract
Human spermatogonial stem cells (SSCs) and Sertoli cells might have the applications in reproduction and regenerative medicine. Abnormal spermatogenesis results in male infertility, which seriously affects human reproduction and health. Spermatogenesis depends on the epigenetic and genetic regulation of male germ cells and somatic cells. A number of microRNAs (miRNAs) have been identified in human testicular tissues, and they are closely related to male fertility. Significantly, we and peers have recently demonstrated that numerous miRNAs are essential for regulating the self-renewal and apoptosis of human SSCs and Sertoli cells through controlling their mRNA and lncRNA targets. In this review, we critically discuss these findings regarding the important functions and mechanisms of miRNAs in mediating the fate determinations of human SSCs and Sertoli cells. Meanwhile, we illustrate the regulatory networks for miRNAs by forming the upstream and downstream regulators of mRNAs and lncRNAs in human SSCs, and we address that miRNAs regulate the decisions of Sertoli cells by targeting genes and via N6-methyladenosine (m6A). We also point out the future directions for further studies on this field. This review could offer an update on novel molecular targets for treating male infertility and new insights into epigenetic regulation of human spermatogenesis.
Collapse
Affiliation(s)
- Wei Chen
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China
| | - Yinghong Cui
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China
| | - Minqi Ning
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China
| | - Haorui Zhang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China
| | - Chenjun Yin
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China
| | - Zuping He
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China.
| |
Collapse
|
30
|
Jiang H, Fan C, Lu Y, Cui X, Liu J. Astragaloside regulates lncRNA LOC100912373 and the miR‑17‑5p/PDK1 axis to inhibit the proliferation of fibroblast‑like synoviocytes in rats with rheumatoid arthritis. Int J Mol Med 2021; 48:130. [PMID: 34013364 PMCID: PMC8136124 DOI: 10.3892/ijmm.2021.4963] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Previous studies have confirmed that astragaloside (AST) exerts a positive effect on alleviating synovial and joint injury in rheumatoid arthritis (RA). However, the precise mechanisms through which AST acts in the treatment of RA remain unclear. Long non-coding RNA (lncRNA) LOC100912373 was identified as a key gene related to RA and has been proven to interact with miR-17-5p, in order to regulate the pyruvate dehydrogenase kinase 1 and protein kinase B axis (PDK1/AKT axis). The present study aimed to determine whether AST may treat RA through the interaction between lncRNA LOC100912373 and the miR-17-5p/PDK1 axis. MTT assays and flow cytometry were used to detect the proliferation and cell cycle progression of AST-treated fibroblast-like synoviocytes (FLSs). The expression of lncRNA LOC100912373 and miR-17-5p, as well as relative the mRNA expression of the PDK1 and AKT genes following AST intervention was detected by reverse transcription-quantitative PCR (RT-qPCR), immunofluorescence and western blot analysis. The results revealed that AST inhibited FLS proliferation, reduced lncRNA LOC100912373 expression levels, increased miR-17-5p expression levels, and decreased the PDK1 and p-AKT expression levels. Additionally, consecutive rescue experiments revealed that AST counteracted the effects of lncRNA LOC100912373 overexpression on FLS proliferation and cell cycle progression. On the whole, the present study demonstrates that AST inhibits FLS proliferation by regulating the expression of lncRNA LOC100912373 and the miR-17-5p/PDK1 axis.
Collapse
Affiliation(s)
- Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Chang Fan
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Yunqi Lu
- Department of Biochemistry, Drew University, Madison, NJ 07940, USA
| | - Xiaoya Cui
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| | - Jian Liu
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
31
|
Novel Gene Regulation in Normal and Abnormal Spermatogenesis. Cells 2021; 10:cells10030666. [PMID: 33802813 PMCID: PMC8002376 DOI: 10.3390/cells10030666] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/01/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
Spermatogenesis is a complex and dynamic process which is precisely controlledby genetic and epigenetic factors. With the development of new technologies (e.g., single-cell RNA sequencing), increasingly more regulatory genes related to spermatogenesis have been identified. In this review, we address the roles and mechanisms of novel genes in regulating the normal and abnormal spermatogenesis. Specifically, we discussed the functions and signaling pathways of key new genes in mediating the proliferation, differentiation, and apoptosis of rodent and human spermatogonial stem cells (SSCs), as well as in controlling the meiosis of spermatocytes and other germ cells. Additionally, we summarized the gene regulation in the abnormal testicular microenvironment or the niche by Sertoli cells, peritubular myoid cells, and Leydig cells. Finally, we pointed out the future directions for investigating the molecular mechanisms underlying human spermatogenesis. This review could offer novel insights into genetic regulation in the normal and abnormal spermatogenesis, and it provides new molecular targets for gene therapy of male infertility.
Collapse
|
32
|
Up-regulation of P21-activated kinase 1 in osteoarthritis chondrocytes is responsible for osteoarthritic cartilage destruction. Biosci Rep 2021; 40:221716. [PMID: 31868209 PMCID: PMC6954364 DOI: 10.1042/bsr20191017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 11/19/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis is mainly caused by a degenerative joint disorder, which is characterized by the gradual degradation of articular cartilage and synovial inflammation. The chondrocyte, the unique resident cell type of articular cartilage, is crucial for the development of osteoarthritis. Previous studies revealed that P21-activated kinase-1 (PAK1) was responsible for the initiation of inflammation. The purpose of the present study was to determine the potential role of PAK1 in osteoarthritis. The level of PAK1 expression was measured by Western blot and quantitative real-time PCR in articular cartilage from osteoarthritis model rats and patients with osteoarthritis. In addition, the functional role of aberrant PAK1 expression was detected in the chondrocytes. We found that the expression of PAK1 was significantly increased in chondrocytes treated with osteoarthritis-related factors. Increased expression of PAK1 was also observed in knee articular cartilage samples from patients with osteoarthritis and osteoarthritis model rats. PAK1 was found to inhibit chondrocytes proliferation and to promote the production of inflammatory cytokines in cartilages chondrocytes. Furthermore, we found that PAK1 modulated the production of extracellular matrix and cartilage degrading enzymes in chondrocytes. Results of the present studies demonstrated that PAK1 might play an important role in the pathogenesis of osteoarthritis.
Collapse
|
33
|
Wu X, Lin D, Sun F, Cheng CY. Male Infertility in Humans: An Update on Non-obstructive Azoospermia (NOA) and Obstructive Azoospermia (OA). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:161-173. [PMID: 34453736 DOI: 10.1007/978-3-030-77779-1_8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Non-obstructive azoospermia (NOA) and obstructive azoospermia (OA) are two common causes of infertility that affect a considerable number of men. However, few studies were performed to understand the molecular etiology of these disorders. Studies based on bioinformatics and genetic analyses in recent years, however, have yielded insightful information and have identified a number of genes that are involved in these disorders. In this review, we briefly summarize and evaluate these findings. We also discuss findings based on epigenetic modifications of sperm DNAs that affect a number of genes pertinent to NOA and OA. The information summarized in this Chapter should be helpful to investigators in future functional studies of NOA and OA.
Collapse
Affiliation(s)
- Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Dengfeng Lin
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Fei Sun
- Sir Run Run Shaw Hospital (SRRSH), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - C Yan Cheng
- Sir Run Run Shaw Hospital (SRRSH), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
34
|
Jabari A, Sadighi Gilani MA, Koruji M, Gholami K, Mohsenzadeh M, Rastegar T, Khadivi F, Ghanami Gashti N, Nikmahzar A, Mojaverrostami S, Talebi A, Ashouri Movassagh S, Rezaie MJ, Abbasi M. Three-dimensional co-culture of human spermatogonial stem cells with Sertoli cells in soft agar culture system supplemented by growth factors and Laminin. Acta Histochem 2020; 122:151572. [PMID: 32622422 DOI: 10.1016/j.acthis.2020.151572] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022]
Abstract
Application of a three-dimensional (3D) culture system for in vitro proliferation and differentiation of human spermatogonial stem cells (SSCs) is a useful tool for the investigation of the spermatogenesis process and the management of male infertility particularly in prepubertal cancer patients. The main purpose of this study was to investigate the proliferation of human SSCs co-cultured with Sertoli cells in soft agar culture system (SACS) supplemented by Laminin and growth factors. Testicular cells were isolated from testes of brain-dead patients and cultured in two-dimensional (2D) culture system for 3 weeks. After 3 weeks, functional SSCs were evaluated by xenotransplantation and also identification of cells was assessed by immunocytochemistry, flow cytometry, and RT-PCR. Then, SSCs and Sertoli cells were transferred to the upper layer of SACS for 3 weeks. After 3 weeks, the number of colonies and the expression of specific SSCs and Sertoli cell markers, as well as apoptotic genes were evaluated. Our results showed that transplanted SSCs, migrated into the basement membrane of seminiferous tubules of recipient mice. The expression of PLZF, α6-Integrin, and Vimentin proteins in SSCs and Sertoli cells were observed in 2D and 3D culture systems. The expression rate of PLZF, α6-Integrin, Bcl2, and colony number in SACS supplemented by Laminin and growth factors group were significantly higher than non-supplemented groups (P ≤ 0.01), but the expression rate of c-kit and Bax in supplemented group were significantly lower than non-supplemented groups (P ≤ 0.05). This 3D co-culture system decreased apoptosis and increased propagation of human SSCs. Therefore, this designed system can be utilized to increase the proliferation of human SSCs in prepubertal male cancer and azoospermic men to obtain an adequate SSCs number to outotransplant success and in vitro spermatogenesis.
Collapse
Affiliation(s)
- Ayob Jabari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Morteza Koruji
- Cellular and Molecular Research Center & Department of Anatomical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Keykavos Gholami
- Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mojtaba Mohsenzadeh
- Iranian Tissue Bank and Research Center of Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Rastegar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Khadivi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Ghanami Gashti
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aghbibi Nikmahzar
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Talebi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Sepideh Ashouri Movassagh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Mohammad Jafar Rezaie
- Department of Embryology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mehdi Abbasi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Liu C, Li P, Zhou W, Ma X, Wang X, Xu Y, Jiang N, Zhao M, Zhou T, Yin Y, Ren J, Huang R. Genome Data Uncover Conservation Status, Historical Relatedness and Candidate Genes Under Selection in Chinese Indigenous Pigs in the Taihu Lake Region. Front Genet 2020; 11:591. [PMID: 32582299 PMCID: PMC7296076 DOI: 10.3389/fgene.2020.00591] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 05/15/2020] [Indexed: 12/17/2022] Open
Abstract
Chinese indigenous pig breeds in the Taihu Lake (TL) region of Eastern China are well documented by their exceptional prolificacy. There are seven breeds in this region including Meishan (MS), Erhualian (EHL), Jiaxing Black (JXB), Fengjing (FJ), Shawutou (SWT), Mi (MI), and Hongdenglong (HDL). At present, these breeds are facing a great threat of population decline, inbreeding depression and lineage admixture since Western commercial pigs have dominated in Chinese pig industry. To provide better conservation strategies and identify candidate genes under selection for these breeds, we explored genome-wide single nucleotide polymorphism (SNP) markers to uncover genetic variability and relatedness, population structure, historical admixture and genomic signature of selection of 440 pigs representing the most comprehensive lineages of these breeds in TL region in a context of 1228 pigs from 45 Eurasian breeds. We showed that these breeds were more closely related to each other as compared to other Eurasian breeds, defining one of the main ancestral lineages of Chinese indigenous pigs. These breeds can be divided into two subgroups, one including JXB and FJ, and the other comprising of EHL, MI, HDL, MS, and SWT. In addition, HDL was highly inbred whereas EHL and MS had more abundant genetic diversity owing to their multiple conservation populations. Moreover, we identified a list of candidate genes under selection for body size and prolificacy. Our results would benefit the conservation of these valuable breeds and improve our understanding of the genetic mechanisms of body size and fecundity in pigs.
Collapse
Affiliation(s)
- Chenxi Liu
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China
| | - Pinghua Li
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China.,Huaian Academy, Nanjing Agricultural University, Huaian, China
| | - Wuduo Zhou
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China
| | - Xiang Ma
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China
| | - Xiaopeng Wang
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yan Xu
- Jiangsu Provincial Station of Animal Husbandry, Nanjing, China
| | - Nengjing Jiang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China
| | - Moran Zhao
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China
| | - Tianwei Zhou
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China
| | - Yanzhen Yin
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China
| | - Jun Ren
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ruihua Huang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, China.,Huaian Academy, Nanjing Agricultural University, Huaian, China
| |
Collapse
|
36
|
Mao H, Nian J, Wang Z, Li X, Huang C. KDELR2 is an unfavorable prognostic biomarker and regulates CCND1 to promote tumor progression in glioma. Pathol Res Pract 2020; 216:152996. [PMID: 32534703 DOI: 10.1016/j.prp.2020.152996] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/07/2020] [Accepted: 04/22/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND The KDEL receptor is a seven-transmembrane-domain protein, which plays a key role in ER quality control and in the ER stress response, KDELR2 involved in regulation of cellular functions, including cell proliferation, survival, promotes glioblastoma tumorigenesis. The aim of this study was to investigate the clinicpathological value and biological role of KDELR2 in glioma. METHODS We studied the expression of KEDLR2 and its association with the prognosis through the TCGA, CGGA, and GSE16011 database. To explore the role of KDELR2 in glioma, KDELR2 siRNA was constructed and transfected into U87 glioma cells. CCK-8, colony formation and Transwell assays were used to investigate the roles of KDELR2 on GBM cell proliferation. We further studied the effect of KDELR2 on tumorigenesis in animal model. Additionally, flow cytometry was used to monitor the changes in the cell cycle and apoptosis following transfection with KDELR2 siRNA. We applied GeneChip primeview expression array to analysis the differential gene expression profiling. Ingenuity Pathway Analysis to show that KDELR2 has a significant impact in canonical pathway in cell cycle regulation and participate in multiple pathways. And we detected the cell cycle proteins CCND1 expression by Western blot analysis. RESULTS Our results showed that KDELR2 was up-regulated in glioma tissue and cell lines. Knockdown KDELR2 was able to reduce cell viability, promote cell cycle arrest at the G1 phase, and induce apoptotic cell death. Moreover, our results suggested that KDELR2 regulated the cellular functions of U87 cells by targeting CCND1. Therefore, we demonstrated that KDELR2 is a novel biomarker in glioma. CONCLUSIONS KDELR2 is highly expressed in human glioma tissues and cell lines, a higher expression of KDELR2 is associated with a poor prognosis of glioma patients. Moreover, KDELR2 regulated the cellular functions of U87 cells by targeting CCND1. The KDELR2/CCND1 axis may provide a new therapeutic target for the treatment of glioma and deepen our understanding of glioma mechanisms.
Collapse
Affiliation(s)
- Hui Mao
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China
| | - Jiang Nian
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhao Wang
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China
| | - XueJun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - ChunHai Huang
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China.
| |
Collapse
|
37
|
Liu C, Tang M, Zhang X, Li J, Cao G. Knockdown of miR-665 Protects Against Cardiomyocyte Ischemia/Reperfusion Injury-Induced ROS Accumulation and Apoptosis Through the Activation of Pak1/Akt Signaling in Myocardial Infarction. Int Heart J 2020; 61:347-354. [DOI: 10.1536/ihj.19-416] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Chuanzhen Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Mengmeng Tang
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Xiquan Zhang
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Jianhua Li
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| | - Guangqing Cao
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University
| |
Collapse
|
38
|
Qian Y, Wu X, Wang H, Hou G, Han X, Song W. PAK1 silencing is synthetic lethal with CDK4/6 inhibition in gastric cancer cells via regulating PDK1 expression. Hum Cell 2020; 33:377-385. [PMID: 31919718 DOI: 10.1007/s13577-019-00317-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. The prognosis of GC is unsatisfied owning to widespread metastasis. P21-activated kinase 1 (PAK1), a member of serine/threonine kinases, is associated with the progression of multiple types of human cancers. Here, we demonstrated that CDK4/6 inhibitor reduced GC cell viability and decreased PAK1 expression. Consistently, PAK1 ablation increased GC cell sensitivity exposed to CDK4/6 inhibitor and promoted DNA damage. We also revealed PAK1 depletion notably affected PDK1-AKT pathway, and PDK1 overexpression totally abrogated the effect of PAK1 deletion on DNA damage in GC cells. Additionally, PDK1 overexpression also rescued the increased GC cell sensitivity towards CDK4/6 inhibitor and the cell cycle arrest caused by PAK1 depletion. Our findings, therefore, suggested that PAK1 silencing increased sensitivity to CDK4/6 inhibition in gastric cancer cells via PDK1-AKT pathway. We, therefore, thought PAK1 as a promising therapeutic target for the treatment of CDK4/6 inhibitor-resistant gastric cancer.
Collapse
Affiliation(s)
- Yan Qian
- Department of Gastric, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, Jiangsu, China
| | - Xu Wu
- Department of Gastric, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, Jiangsu, China
| | - Haixiao Wang
- Department of Gastric, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, Jiangsu, China
| | - Guowei Hou
- Department of Gastric, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, Jiangsu, China
| | - Xiao Han
- Department of Gastric, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, Jiangsu, China
| | - Wei Song
- Department of Gastroenterlogy, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Huanghe West Road, Huaiyin, Huai'an, 223300, Jiangsu, China.
| |
Collapse
|
39
|
Qiu Q, Yu X, Yao C, Hao Y, Fan L, Li C, Xu P, An G, Li Z, He Z. FOXP3 pathogenic variants cause male infertility through affecting the proliferation and apoptosis of human spermatogonial stem cells. Aging (Albany NY) 2019; 11:12581-12599. [PMID: 31855573 PMCID: PMC6949051 DOI: 10.18632/aging.102589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/26/2019] [Indexed: 12/11/2022]
Abstract
Genetic causes of male infertility that is associated with aging are largely unknown. This study was designed to identify novel pathogenic variants of FOXP3 gene causing azoospermia. One homozygous (c.155 G > T) pathogenic variant of FOXP3 was identified in nine non-obstructive azoospermia patients, and one heterozygous (c.691 C > A) of FOXP3 was found in one non-obstructive azoospermia patient. Pedigrees studies indicated that the homozygous (c.155 G > T) FOXP3 pathogenic variant was inherited, while heterozygous (c.691 C > A) FOXP3 pathogenic variant was acquired. Human testis carrying pathogenic variant exhibited abnormal spermatogenesis. FOXP3 protein was expressed at a lower level or undetected in spermatocytes of mutant testis of non-obstructive azoospermia patients compared to obstructive azoospermia patients. FOXP3 stimulated the proliferation and inhibited the apoptosis of human spermatogonial stem cells, and we further analyzed the targets of FOXP3. We have identified two new pathogenic variants of FOXP3 in non-obstructive azoospermia patients with high incidence, and FOXP3 silencing inhibits the proliferation and enhances the apoptosis of human spermatogonial stem cells. This study provides new insights into the etiology of azoospermia and offers novel pathogenic variants for gene targeting of male infertility.
Collapse
Affiliation(s)
- Qianqian Qiu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Yu
- Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Chencheng Yao
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yujun Hao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liqing Fan
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Chunyi Li
- Fertility Center, Shenyang Dongfang Jinghua Hospital, Shenyang, Liaoning, China
| | - Peng Xu
- Fertility Center, Shenyang Dongfang Jinghua Hospital, Shenyang, Liaoning, China
| | - Geng An
- Department of Reproductive Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zheng Li
- Department of Andrology, The Center for Men's Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zuping He
- Hunan Normal University School of Medicine, Changsha, Hunan, China.,Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai, China
| |
Collapse
|
40
|
Fu H, Zhou F, Yuan Q, Zhang W, Qiu Q, Yu X, He Z. miRNA-31-5p Mediates the Proliferation and Apoptosis of Human Spermatogonial Stem Cells via Targeting JAZF1 and Cyclin A2. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 14:90-100. [PMID: 30583099 PMCID: PMC6305686 DOI: 10.1016/j.omtn.2018.11.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/10/2018] [Accepted: 11/11/2018] [Indexed: 01/15/2023]
Abstract
Several lines of evidence highlight the important application of human spermatogonial stem cells (SSCs) in translational medicine. The fate decisions of SSCs are mainly mediated by genetic and epigenetic factors. We have recently demonstrated that PAK1 regulates the proliferation, DNA synthesis, and early apoptosis of human SSCs through the PDK1/KDR/ZNF367 and ERK1/2 and AKT pathway. However, the underlying epigenetic mechanism of PAK1 in human SSCs remains unknown. In this study, we found that the level of miRNA-31-5p was elevated by PAK1 knockdown. CCK-8, PCNA, and 5-ethynyl-2′-deoxyuridine (EDU) assays revealed that miRNA-31-5p mimics inhibited cell proliferation and DNA synthesis of human SSCs. Annexin V/propidium iodide (PI) staining and flow cytometry showed that miRNA-31-5p increased the early and late apoptosis of human SSCs. Furthermore, JAZF1 was predicted and verified as a target of miRNA-31-5p, and the three-dimensional (3D) structure model of JAZF1 protein was illustrated. JAZF1 silencing led to a reduction of cell proliferation and DNA synthesis as well as an enhancement of the early and late apoptosis of human SSCs. Finally, miRNA-31-5p mimics decreased the level of cyclin A2 rather than cyclin D1 or cyclin E1, and JAZF1 knockdown led to the reduction of cyclin A2 in human SSCs. Collectively, miRNA-31-5p regulates the proliferation, DNA synthesis, and apoptosis of human SSCs by the PAK1-JAZF1-cyclin A2 pathway. This study thus offers a novel insight into the molecular mechanisms underlying the fate determinations of human SSCs and might provide novel targets for molecular therapy of male infertility.
Collapse
Affiliation(s)
- Hongyong Fu
- Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China; The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, Henan 450008, China
| | - Fan Zhou
- Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China
| | - Qingqing Yuan
- Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China
| | - Wenhui Zhang
- Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China
| | - Qianqian Qiu
- Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China
| | - Xing Yu
- Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China
| | - Zuping He
- Hunan Normal University School of Medicine, 371 Tongzipo Road, Changsha, Hunan 410013, China; Renji- Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai 200127, China; Shanghai Key Laboratory of Assisted Reproduction and Reproductive Genetics, Shanghai 200127, China; Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China.
| |
Collapse
|