1
|
Silva-Palacios A, Zúñiga-Muñoz AM, Soria-Castro E, Álvarez-León E, Nieto M, Navarrete-Anastasio G, Carbó R, García-Niño WR, López-Cervantes SP, Salas-Venegas V, Flores-Torres RP, Luna-López A, Zazueta C, Königsberg M. Cardioprotective effect of senotherapy in chronically obese middle-aged female rats may be mediated by a MERCSs/Nrf2 interaction. J Nutr Biochem 2025; 142:109923. [PMID: 40250489 DOI: 10.1016/j.jnutbio.2025.109923] [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: 12/18/2024] [Revised: 03/16/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
Hypercaloric intake promotes the development of obesity, a risk factor for cardiovascular disease (CVD). In recent years, it has been suggested that senescent cells have negative implications for the outcome of these chronic pathologies, and senotherapy has emerged as a novel intervention to reduce damage to the organism. However, it is unclear whether the accumulation of senescent cells induces alterations at the cardiac level in rats fed a hypercaloric diet (HD) and if the use of senotherapeutics can reverse it. To address this question, we used middle-aged female rats fed HD from 21 days to 15 months of age. Under our experimental conditions, rats exhibited cardiac hypertrophy and fibrosis, accumulation of senescent cells, changes in mitochondrial morphology, and oxidative stress. Rats were treated for 2 months with senolytic (dasatinib + quercetin, DQ) or senomorphic (sulforaphane, SFN) agents. Interestingly, the HD rats showed cardiac improvement after the treatment. Our data suggest a possible link mechanism between Nrf2 activation and mitochondria-endoplasmic reticulum contact sites (MERCSs) preservation, activated by SFN rather than by the DQ combination, which allowed cardiac structure maintenance in HD rats decreasing the harmful effects of senescent cells.
Collapse
Affiliation(s)
- Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.
| | - Alejandra María Zúñiga-Muñoz
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Elizabeth Soria-Castro
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Edith Álvarez-León
- Subdirección de Investigación Básica y Tecnológica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Mario Nieto
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Gabriela Navarrete-Anastasio
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Wylly Ramsés García-Niño
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Stefanie Paola López-Cervantes
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico; Posgrado en Biología Experimental, Universidad Autónoma Metropolitana Iztapalala, Mexico City, Mexico
| | - Verónica Salas-Venegas
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Rosa Pamela Flores-Torres
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico; Posgrado en Biología Experimental, Universidad Autónoma Metropolitana Iztapalala, Mexico City, Mexico
| | - Armando Luna-López
- Departamento de Investigación Básica, Instituto Nacional de Geriatria, Mexico City, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Mina Königsberg
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autonóma Metropolitana Iztapalapa, Mexico City, Mexico.
| |
Collapse
|
2
|
Guo X, Wang L, Xuan J, Chen T, Du Y, Qiao H, Zhang S, Sun Z, Wang J, Niu R. Fluoride induces spermatocyte apoptosis by IP3R1/MCU-mediated mitochondrial calcium overload through MAMs. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137514. [PMID: 39929132 DOI: 10.1016/j.jhazmat.2025.137514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/16/2025] [Accepted: 02/04/2025] [Indexed: 04/16/2025]
Abstract
Excessive fluoride exposure has been shown to induce diminished sperm quality and mitochondrial dysfunction. The interaction between mitochondria and the endoplasmic reticulum (ER) is critical for regulating mitochondrial function in spermatogenic cells. Therefore, this study was designed to investigate the molecular events involved in mitochondria-associated ER membranes (MAMs) in mice exposed to 25, 50, and 100 mg/L NaF for 60 days, and in GC-2spd treated with 1.5, 2.0, and 2.5 mM NaF for 24 hours. Mitochondrial stress tests revealed a significant reduction in basal respiration, maximal respiration, and ATP production, suggesting mitochondrial dysfunction following fluoride exposure. Results further indicated that fluoride exposure significantly enhanced ER-mitochondria contacts, mitochondrial Ca2+ levels, and the expressions of IP3R1, GRP75, VDAC1, and MCU, while reduced the levels of MFN1, MFN2, VAPB, and PTPIP51, along with an increase in Cytochrome C and Caspase-3. Treatment with the Ru360 and IP3R1 siRNA restored mitochondrial membrane potential, while reduced mitochondrial Ca2+ levels and apoptosis rates, indicating that both MCU and IP3R1 play a role in regulating fluoride-induced the formation of MAMs. Collectively, these findings proved that fluoride promoted Ca2+ transfer through MAMs in spermatocytes via the IP3R1-GRP75-VDAC1-MCU axis, and inhibiting IP3R1/MCU might be a potential therapeutic target in fluorosis.
Collapse
Affiliation(s)
- Xin Guo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Linyuan Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Jingyan Xuan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Tong Chen
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Yu Du
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Hanxing Qiao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Shaosan Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Jundong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Ruiyan Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China.
| |
Collapse
|
3
|
Liu X, Zhang H, Xu L, Ye H, Huang J, Jing Xiang, He Y, Zhou H, Fang L, Zhang Y, Xiang X, Cannon RD, Ji P, Zhai Q. cGAMP-targeting injectable hydrogel system promotes periodontal restoration by alleviating cGAS-STING pathway activation. Bioact Mater 2025; 48:55-70. [PMID: 40303968 PMCID: PMC12038443 DOI: 10.1016/j.bioactmat.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 05/02/2025] Open
Abstract
The impaired function of periodontal ligament stem cells (PDLSCs) impedes restoration of periodontal tissues. The cGAS-cGAMP-STING pathway is an innate immune pathway that sensing cytosolic double-stranded DNA (dsDNA), but its role in regulating the function of PDLSCs is still unclear. In this study, we found that mitochondrial DNA (mtDNA) was released into the cytoplasm through the mitochondrial permeability transition pore (mPTP) in PDLSCs upon inflammation, which binds to cGAS and activated the STING pathway by promoting the production of cGAMP, and ultimately impaired the osteogenic differentiation of PDLSCs. Additionally, it is first found that inflammation can down-regulate the level of the ATP-binding cassette membrane subfamily member C1 (ABCC1, a cGAMP exocellular transporter) and ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1, a cGAMP hydrolase), which further aggravated the accumulation of intracellular cGAMP, leading to the persistent activation of the cGAS-STING pathway and thus the impaired the differentiation capacity of PDLSCs. Furthermore, we designed a hydrogel system loaded with a mPTP blocker, an ABCC1 agonist and ENPP1 to promote periodontal tissue regeneration by modulating the production, exocytosis, and clearance of cGAMP. In conclusion, our results highlight the profound effects, and specific mechanisms, of the cGAS-STING pathway on the function of stem cells and propose a new strategy to promote periodontal tissue restoration based on the reestablishment of cGAMP homeostasis.
Collapse
Affiliation(s)
- Xiang Liu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Department of Stomatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hua Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Huayu Ye
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Jinghuan Huang
- Orthopedic Department of Shanghai Sixth People's Hospital, Shanghai, China
| | - Jing Xiang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Yunying He
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Huan Zhou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Lingli Fang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Yunyan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Xuerong Xiang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Richard D. Cannon
- Department of Oral Sciences, Sir John Walsh Research Institute, Dentistry, University of Otago, Dunedin, 9054, New Zealand
| | - Ping Ji
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| | - Qiming Zhai
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
| |
Collapse
|
4
|
Zhou J, Dong Q. Testicular aging: mechanism, management and future therapy. Exp Cell Res 2025; 449:114603. [PMID: 40373850 DOI: 10.1016/j.yexcr.2025.114603] [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: 03/21/2025] [Revised: 05/03/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Testicular aging results in degeneration in testicular function, including decreased testosterone production and quality of sperm. Decreased testosterone level is associated with a range of systemic diseases and comorbidities, including cardiovascular disease, cognitive decline, depression, osteoporosis, frailty, increased body fat, and metabolic syndrome. In addition, with the rapid development of industrialization and increasing environmental pollution, the quality of male semen continues to decline globally. Currently, the average age of first marriage and childbirth for men is delayed, and the birth rate has been declining year by year. At present, the therapies for testosterone level decline in clinical practice are relatively limited. Therefore, studying the triggering and delaying mechanisms of testicular aging is significant for improving male health and protecting male fertility. This review summarizes the mechanisms and treatment strategies for male reproductive aging.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, 610000, Sichuan Province, China
| | - Qiang Dong
- Department of Urology, Institute of Urology, West China Hospital of Sichuan University, Chengdu, 610000, Sichuan Province, China.
| |
Collapse
|
5
|
Huang Y, Prastyaningrum LL, Wang X, Xu F, Wang Z, Wang Z, Tan X, Dai G, Chen G, Gong X, Yang L. MICU1 is the nexus for Ca V3.3 regulation of mitochondrial calcium, redox balance and chondrocyte viability. Int J Biol Macromol 2025; 312:144127. [PMID: 40354861 DOI: 10.1016/j.ijbiomac.2025.144127] [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: 03/08/2025] [Revised: 04/17/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Voltage-gated calcium channels are emerging regulators of cellular homeostasis, but their molecular interplay with mitochondrial bioenergetics in chondrocytes remains poorly characterized. This study elucidates how the T-type calcium channel CaV3.3 governs mitochondrial calcium-redox coupling through structural interactions with MICU1, the regulatory subunit of the mitochondrial calcium uniporter (MCU) complex. The absence of the CaV3.3 precipitated mitochondrial ultrastructural disorganization characterized, coupled with MICU1 downregulation and consequent loss of MCU gating fidelity. Through integrated transcriptomic-proteomic profiling and live-cell imaging, we demonstrate that CaV3.3 deficiency induces pathological mitochondrial calcium influx, triggering Reactive oxygen species (ROS) overproduction and bioenergetic collapse, these metabolic derangements activated intrinsic apoptosis. Notably, lentiviral overexpression of MICU1 in CaV3.3 knockout cells restored the mitochondrial calcium set point and inhibited ROS burst, while rescued cell proliferation and inhibited apoptosis execution. Our findings establish CaV3.3 as a redox rheostat coordinating MICU1-mediated mitochondrial calcium buffering, with direct implications for cartilage matrix maintenance and osteoarthritis therapy targeting calcium-handling macromolecules.
Collapse
Affiliation(s)
- Yumengfei Huang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China; Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Lucky Laras Prastyaningrum
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xin Wang
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Fa Xu
- Knorigene Technologies, Chongqing 400084, China
| | - Zonghan Wang
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Zhi Wang
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Xin Tan
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Gang Dai
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Guangxing Chen
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China.
| | - Xiaoyuan Gong
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China.
| | - Liu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China; Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China.
| |
Collapse
|
6
|
Li T, Huang L, Guo C, Ren J, Chen X, Ke Y, Xun Z, Hu W, Qi Y, Wang H, Gong Z, Liang X, Xue X. Massage-Mimicking Nanosheets Mechanically Reorganize Inter-organelle Contacts to Restore Mitochondrial Functions in Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413376. [PMID: 40223359 PMCID: PMC12120710 DOI: 10.1002/advs.202413376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Parkinson's disease (PD) is exacerbated by dysfunction of inter-organelle contact, which depends on cellular responses to the mechanical microenvironment and can be regulated by external mechanical forces. Delivering dynamic mechanical forces to neural cells proves challenging due to the skull. Inspired by the effects of massage; here PEGylated black phosphorus nanosheets (PEG-BPNS), known for their excellent biocompatibility, biodegradability, specific surface area, mechanical strength, and flexibility, are introduced, which are capable of adhering to neural cell membrane and generating mechanical stimulation with their lateral size of 200 nm, exhibiting therapeutic potential in a 1-methyl-4-phenyl-1,2,3,6-te-trahydropyridine-induced PD mouse model by regulating inter-organelle contacts. Specifically, it is found that 200 nm PEG-BPNS, acting as "NanoMassage," significantly increase plasma membrane tension, as evidenced by fluorescent lipid tension reporter fluorescence lifetime analysis. This mechanical force modulates actin reorganization, subsequently regulating the contacts between actin, mitochondria, and endoplasmic reticulum, further controlling mitochondrial fission and mitigating mitochondrial dysfunction in PD, exhibiting therapeutic efficacy via intranasal administration. These findings provide a noninvasive strategy for applying mechanical stimulation to deep brain areas and elucidate the mechanism of NanoMassage mediating inter-organelle contacts, suggesting the rational design of "NanoMassage" to remodel inter-organelle communications in neurodegenerative disease treatment.
Collapse
Affiliation(s)
- Tianqi Li
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Liwen Huang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
- Present address:
Department of ChemistryShanghai Key Laboratory of Molecular Catalysis and Innovative MaterialsState Key Laboratory of Molecular Engineering of Polymers and iChemFudan UniversityShanghai200438P. R. China
| | - Chenxiao Guo
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Jing Ren
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Xi Chen
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Yachu Ke
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Zengyu Xun
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Wenzhuo Hu
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Yilin Qi
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| | - Heping Wang
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
- Present address:
State Key Laboratory of Advanced Medical Materials and DevicesTianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineKey Laboratory of Radiopharmacokinetics for Innovative DrugsTianjin Institutes of Health ScienceInstitute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300192P. R. China
| | - Zhongying Gong
- Department of NeurologyTianjin First Central HospitalSchool of MedicineNankai UniversityTianjin300192P. R. China
| | - Xing‐Jie Liang
- Laboratory of Controllable NanopharmaceuticalsChinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyNational Center for Nanoscience and TechnologyBeijing100190P. R. China
- University of Chinese Academy of SciencesBeijing101408P. R. China
| | - Xue Xue
- State Key Laboratory of Medicinal Chemical BiologyCollege of PharmacyNankai UniversityTianjin300350P. R. China
| |
Collapse
|
7
|
Wang B, Yu R, Zhang Z, Peng Y, Li L. Exosomes secreted from adipose-derived stem cells inhibit M1 macrophage polarization ameliorate chronic endometritis by regulating SIRT2/NLRP3. Mol Cell Biochem 2025:10.1007/s11010-025-05283-2. [PMID: 40257720 DOI: 10.1007/s11010-025-05283-2] [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: 11/13/2024] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
Chronic endometritis (CE) is a key factor in adverse pregnancy outcomes such as miscarriage and infertility. Macrophages are an important immune cell type that secrete pro-inflammatory and anti-inflammatory cytokines that are essential for maintaining endometrial function. This study aimed to investigate the key mechanisms by which exosomes derived from adipose-derived mesenchymal stem cells (ADSCs) regulate macrophage polarization through the sirtuin 2 (SIRT2)/NOD-like receptor pyrin containing 3 (NLRP3) axis and exert a protective effect on CE. Exosomes were obtained from ADSCs (ADSCs-exo) using the classical ultracentrifugation method and characterized using transmission electron microscopy, nanoparticle tracking analysis, and western blotting. ADSCs-exo protective effects on CE mice and RAW 264.7 cells and its related molecular mechanisms were investigated using real-time quantitative polymerase chain reaction, western blotting, enzyme-linked immunosorbent assay, flow cytometry, immunofluorescence, immunoprecipitation, hematoxylin and eosin staining, and immunohistochemistry. ADSCs-exo significantly inhibited M1 macrophage polarization, as evidenced by a 54% reduction in tumor necrosis factor alfa (TNF-α), a 46% reduction in interleukin 1β (IL-1β), and a 36% reduction in interleukin 6 (IL-6) levels in LPS-induced RAW264.7 cells. In vivo, ADSCs-exo treatment reduced the expression of TNF-α by 50%, IL-1β by 58%, and IL-6 by 49% in the uterine tissues of CE mice. Moreover, ADSCs-exo upregulated the expression of SIRT2, promoted the deacetylation modification of NLRP3 to inhibit NLRP3 inflammasome activation, and further suppressed M1 macrophage polarization. However, these trends were reversed after SIRT2 silencing. Our experimental results demonstrate that ADSCs-exo alleviate CE by regulating the SIRT2/NLRP3 axis to inhibit M1 macrophage polarization. This provides a potential theoretical basis for the therapeutic role of stem cells in CE.
Collapse
Affiliation(s)
- Bin Wang
- Department of Reproduction, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), No.78, Wandao Road, Wanjiang District, Dongguan, 523059, Guangdong Province, China
| | - Ruizhu Yu
- Department of Reproduction, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), No.78, Wandao Road, Wanjiang District, Dongguan, 523059, Guangdong Province, China
| | - Zhao Zhang
- Department of Reproduction, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), No.78, Wandao Road, Wanjiang District, Dongguan, 523059, Guangdong Province, China
| | - Yuhong Peng
- Department of Reproduction, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), No.78, Wandao Road, Wanjiang District, Dongguan, 523059, Guangdong Province, China.
| | - Li Li
- Department of Rheumatology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), No.78, Wandao Road, Wanjiang District, Dongguan, 523059, Guangdong Province, China.
| |
Collapse
|
8
|
Wu J, He R, Xu Z, Yang H, Guan Y, Sun L, Lv W, Huang J, Wang J. Mechanical signal-mediated mitochondria-endoplasmic reticulum contacts modulate Leydig cell testosterone biosynthesis during testis development. Mol Hum Reprod 2025; 31:gaaf017. [PMID: 40419464 DOI: 10.1093/molehr/gaaf017] [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: 12/13/2024] [Revised: 04/07/2025] [Indexed: 05/28/2025] Open
Abstract
In males, 95% of testosterone is synthesized by Leydig cells, and a deficiency in this synthesis will cause metabolic disorders and multiple organ dysfunction. Testosterone deficiency is not only affected by aged or diseased Leydig cells, which have been studied extensively, but is also closely related to the development of the testis. At present, the focus on the mechanism of testis development includes epigenetic and hormone regulation. However, testicular development is constrained by the external tough tunica albuginea, suggesting that mechanical signals may also play an important role in the regulation of testis development; however, this is not yet well understood. In this in vitro study, we found that a gradual increase in extracellular substrate stiffness for testis development leads to the activation of mechanical signals to promote cytoskeleton remodeling. Eventually, the mechanical signal mediates changes in the mitochondrial-endoplasmic reticulum and affects the synthesis of testosterone in Leydig cells. Through organoid and animal experiments, we found that targeting mechanical signaling pathways that regulate testosterone biosynthesis is feasible. This provides a new angle for further exploration of testis development and new insights into how substrate stiffness affects the testis, raising new clues for clinical applications.
Collapse
Affiliation(s)
- Jiahong Wu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Ruiling He
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Zeyu Xu
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Huan Yang
- Department of Gynecology, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Yupeng Guan
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Lu Sun
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Wantong Lv
- School of Medicine, Sun Yat-Sen University, Shenzhen, P. R. China
| | - Jiayu Huang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, P. R. China
| |
Collapse
|
9
|
Cui L, Nie X, Guo Y, Ren P, Guo Y, Wang X, Li R, Hotaling JM, Cairns BR, Guo J. Single-cell transcriptomic atlas of the human testis across the reproductive lifespan. NATURE AGING 2025; 5:658-674. [PMID: 40033047 PMCID: PMC12003174 DOI: 10.1038/s43587-025-00824-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025]
Abstract
Testicular aging is associated with declining reproductive health, but the molecular mechanisms are unclear. Here we generate a dataset of 214,369 single-cell transcriptomes from testicular cells of 35 individuals aged 21-69, offering a resource for studying testicular aging and physiology. Machine learning analysis reveals a stronger aging response in somatic cells compared to germ cells. Two waves of aging-related changes are identified: the first in peritubular cells of donors in their 30s, marked by increased basement membrane thickness, indicating a priming state for aging. In their 50s, testicular cells exhibit functional changes, including altered steroid metabolism in Leydig cells and immune responses in macrophages. Further analyses reveal the impact of body mass index on spermatogenic capacity as age progresses, particularly after age 45. Altogether, our findings illuminate molecular alterations during testis aging and their relationship with body mass index, providing a foundation for future research and offering potential diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Lina Cui
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xichen Nie
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
- Division of Urology, Department of Surgery, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Yixuan Guo
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Pengcheng Ren
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yifei Guo
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Xiaoyan Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Ran Li
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - James M Hotaling
- Division of Urology, Department of Surgery, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Bradley R Cairns
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, USA.
| | - Jingtao Guo
- State Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of the Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
10
|
Sato K, Koyanagi-Aoi M, Uehara K, Yamashita Y, Shinohara M, Lee S, Reinhardt A, Woltjen K, Chiba K, Miyake H, Fujisawa M, Aoi T. Efficient differentiation of human iPSCs into Leydig-like cells capable of long-term stable secretion of testosterone. Stem Cell Reports 2025; 20:102392. [PMID: 39824187 PMCID: PMC11864132 DOI: 10.1016/j.stemcr.2024.102392] [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: 04/05/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/20/2025] Open
Abstract
Late-onset hypogonadism (LOH) syndrome is characterized by age-related testosterone deficiency and negatively affects the quality of life of older men. A promising therapeutic approach for LOH syndrome is transplantation of testosterone-producing Leydig-like cells (LLCs) derived from human induced pluripotent stem cells (hiPSCs). However, previous studies have encountered obstacles, such as limited cell longevity, insufficient testosterone production, and inefficiency of differentiation. To address these issues, we developed a novel protocol that includes forced NR5A1 expression, a cytokine cocktail promoting mesoderm differentiation, and a transitional shift from 3D to 2D cultures. The resultant cells survived on culture dishes for over 16 weeks, produced 22-fold more testosterone than the conventional method, and constituted a homogeneous population of LLCs with a differentiation efficiency exceeding 99% without purification. Furthermore, these LLCs were successfully engrafted subcutaneously into mice, resulting in increased serum testosterone levels. Our study will facilitate innovative therapeutic strategies for LOH syndrome.
Collapse
Affiliation(s)
- Katsuya Sato
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Michiyo Koyanagi-Aoi
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan
| | - Keiichiro Uehara
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Department of Diagnostic Pathology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yosuke Yamashita
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Masakazu Shinohara
- The Integrated Center for Mass Spectrometry, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Molecular Epidemiology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Suji Lee
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Anika Reinhardt
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Knut Woltjen
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Koji Chiba
- Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Hideaki Miyake
- Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Masato Fujisawa
- Division of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Takashi Aoi
- Division of Stem Cell Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan; Division of Advanced Medical Science, Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan; Center for Human Resource Development for Regenerative Medicine, Kobe University Hospital, Kobe, Japan; Division of Signal Pathways, Biosignal Research Center, Kobe University, Kobe, Japan.
| |
Collapse
|
11
|
Dinevska M, McAloney L, Widodo SS, Filiz G, Anderson J, Dworkin S, Windley SP, Wilhelm D, Mantamadiotis T. Testicular sex cord-stromal tumors in mice with constitutive activation of PI3K and loss of Pten. Carcinogenesis 2025; 46:bgae077. [PMID: 39672957 PMCID: PMC11879163 DOI: 10.1093/carcin/bgae077] [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: 09/16/2024] [Revised: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 12/15/2024] Open
Abstract
Testicular tumors are the most common malignancy of young men, and tumors affecting the testis are caused by somatic mutations in germ or germ-like cells. The PI3K pathway is constitutively activated in about one-third of testicular cancers. To investigate the role of the PI3K pathway in transforming stem-like cells in the testis, we investigated tumors derived from mice with post-natal, constitutive activation of PI3K signaling and homozygous deletion of tumor suppressor Pten, targeted to Nestin-expressing cells. Mice developed aggressive tumors, exhibiting heterogeneous histopathology and hemorrhaging. The tumors resemble the rare testis tumor type, testicular sex cord-stromal Leydig cell tumors. Single-cell resolution spatial tissue analysis demonstrated that T-cells are the dominant tumor-infiltrating immune cell type, with very few infiltrating macrophages observed in the tumor tissue, with CD8+ T-cells predominating. Further analysis showed that immune cells preferentially localize to, or accumulate within stromal regions.
Collapse
Affiliation(s)
- Marija Dinevska
- Department of Surgery (RMH), The University of Melbourne, Parkville, VIC 3052, Australia
| | - Lachlan McAloney
- Department of Microbiology & Immunology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Samuel S Widodo
- Department of Surgery (RMH), The University of Melbourne, Parkville, VIC 3052, Australia
| | - Gulay Filiz
- School of Biosciences, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Jeremy Anderson
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Flemington Road, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia
| | - Simon P Windley
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Dagmar Wilhelm
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Theo Mantamadiotis
- Department of Surgery (RMH), The University of Melbourne, Parkville, VIC 3052, Australia
- Department of Microbiology & Immunology, The University of Melbourne, Parkville, VIC 3052, Australia
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
12
|
Yang F, Li X, Wang M, Lan X, Zhang J, Li J, Chang D, Yu X. The Role of Environmental Endocrine Disruptors on Leydig Cell Death and Senescen. World J Mens Health 2025; 43:43.e5. [PMID: 39843178 DOI: 10.5534/wjmh.240254] [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: 10/08/2024] [Revised: 11/11/2024] [Accepted: 11/26/2024] [Indexed: 01/24/2025] Open
Abstract
Environmental endocrine disruptors, as exogenous chemicals that interfere with hormonal behavior, are known to cause testicular Leydig cell death and senescence. The incidence of diseases of the male reproductive system has been increasing over the past half-century. Genetic defects alone cannot explain the rapid increase in incidence, and there is growing evidence that environmental factors or lifestyle changes are responsible for the high incidence in recent years. Testicular Leydig cells occupy an important role in the male reproductive system. In this study, we review the mechanisms by which environmental endocrine disruptors promote both death and senescence of testicular Leydig cells, refine the former into two programmed death modes, apoptosis, and autophagy, and further explore the interactions among them, thus summarizing the advances of the toxic effects of environmental endocrine disruptors on testicular Leydig cells, and expecting to provide a new therapeutic idea.
Collapse
Affiliation(s)
- Fang Yang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoya Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meijing Wang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiucheng Lan
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingyi Zhang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junjun Li
- Chengdu Fifth People's Hospital, Chengdu, China
| | - Degui Chang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xujun Yu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
13
|
Chi A, Yang C, Liu J, Zhai Z, Shi X. Reconstructing the Stem Leydig Cell Niche via the Testicular Extracellular Matrix for the Treatment of Testicular Leydig Cell Dysfunction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410808. [PMID: 39555675 PMCID: PMC11727238 DOI: 10.1002/advs.202410808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/24/2024] [Indexed: 11/19/2024]
Abstract
Therapies involving the use of stem Leydig cells (SLCs), as testicular mesenchymal stromal cells, have shown great promise in the treatment of Leydig cell (LC) dysfunction in aging males. However, the outcomes of these therapies are not satisfactory. In this study, it is demonstrated that the aging microenvironment of the testicular interstitium impairs the function of SLCs, leading to poor regeneration of LCs and, consequently, inefficient functional restoration. The study develops a decellularized testicular extracellular matrix (dTECM) hydrogel from young pigs and evaluates its safety and feasibility as a supportive niche for the expansion and differentiation of SLCs. dTECM hydrogel facilitates the steroidogenic differentiation of SLCs into LCs, the primary producers of testosterone. The combination of SLCs with a dTECM hydrogel leads to a significant and sustained increase in testosterone levels, which promotes the restoration of spermatogenesis and fertility in an LC-deficient and aged mouse model. Mechanistically, collagen 1 within the dTECM is identified as a key factor contributing to these effects. Notably, symptoms associated with testosterone deficiency syndrome are significantly alleviated in aged mice. These findings may aid the design of therapeutic interventions for patients with testosterone deficiency in the clinic.
Collapse
Affiliation(s)
- Ani Chi
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
| | - Chao Yang
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
| | - Jie Liu
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
| | - Zhichen Zhai
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510006P. R. China
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640P. R. China
| | - Xuetao Shi
- National Engineering Research Centre for Tissue Restoration and ReconstructionKey Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510640P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong ProvinceSouth China University of TechnologyGuangzhou510006P. R. China
- School of Materials Science and EngineeringSouth China University of TechnologyGuangzhou510640P. R. China
| |
Collapse
|
14
|
Ren C, Hu C, Hu M, Wu Y, Yang Y, Lu F. Melatonin protects RPE cells from necroptosis and NLRP3 activation via promoting SERCA2-related intracellular Ca 2+ homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156088. [PMID: 39341129 DOI: 10.1016/j.phymed.2024.156088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Melatonin is an antioxidant that also has anti-inflammatory effects. It has been reported to delay the progression of age-related macular degeneration (AMD), however, the mechanism has not been fully recognized. PURPOSE The aim of the present study was to investigate the effects of melatonin on sodium iodate (SI)-induced retinal degeneration and elucidate the specific mechanisms, then, provide novel targets in AMD treatment. METHODS Retinal degeneration mouse model and in vitro retinal pigment epithelium (RPE) death model were established by SI treatment. Melatonin was administrated intraperitoneally at a concentration of 20, 40 or 80 mg/kg for in vivo study or treated at 48 h before SI treatment. To confirm the therapeutic effects of melatonin on mouse, the retinal structure and visual function were evaluated. The specific cell death rates were determined by CCK-8 assay, PI staining and protein level of RIPK3. The cytosolic or mitochondrial calcium levels were determined by Fluo-4AM or Rhod-2AM staining. Mitochondrial functions including mitochondrial dynamics, mitochondrial membrane potential, or mitochondrial permeability pore opening were evaluated. The proteins involved in endoplasmic reticulum (ER) stress were measured by western blot assay while the genes expression in calcium signaling pathway were measured by RT-qPCR. RESULTS We show that melatonin protects RPE cells from necroptosis and NLRP3 inflammasome activation induced by SI. Mechanistically, melatonin suppresses ER stress and intracellular calcium overload triggered by SI through restoring the function of SERCA2. Silencing of SERCA2 or blocking of melatonin receptors inhibit the protective effects of melatonin. Melatonin reduces mitochondrial Ca2+ levels and restores mitochondrial membrane potential. Constant mitochondrial Ca2+ overload directly promote cell necroptosis through mitochondrial fission. Inhibition of mitochondrial fission by Mdivi-1 prevent necroptosis induced by SI without altering the level of mitochondrial Ca2+. CONCLUSIONS The results confirmed that melatonin protects RPE cells from SI-induced injury by regulates MT2/SERCA2/Ca2+ axis. This study highlighted the potential of melatonin in the treatment of AMD and elucidated the mechanism and signaling pathway that mediate the protective effects.
Collapse
Affiliation(s)
- Chengda Ren
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| | - Chengyu Hu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, PR China.
| | - Ming Hu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yan Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Yang Yang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, PR China.
| | - Fang Lu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
15
|
Winstanley YE, Stables JS, Gonzalez MB, Umehara T, Norman RJ, Robker RL. Emerging therapeutic strategies to mitigate female and male reproductive aging. NATURE AGING 2024; 4:1682-1696. [PMID: 39672895 DOI: 10.1038/s43587-024-00771-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/29/2024] [Indexed: 12/15/2024]
Abstract
People today are choosing to have children later in life, often in their thirties and forties, when their fertility is in decline. We sought to identify and compile effective methods for improving either male or female fertility in this context of advanced reproductive age. We found few clinical studies with strong evidence for therapeutics that mitigate reproductive aging or extend fertility; however, this Perspective summarizes the range of emerging experimental strategies under development. Preclinical studies, in mouse models of aging, have identified pharmaceutical candidates that improve egg and sperm quality. Further, a diverse array of medically assisted reproduction methodologies, including those that stimulate rare ovarian follicles and rejuvenate egg quality using mitochondria, may have future utility for older patients. Finally, we highlight the many knowledge gaps and possible future directions in the field of therapeutics to extend the age of healthy human reproduction.
Collapse
Affiliation(s)
- Yasmyn E Winstanley
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia
| | - Jennifer S Stables
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia
| | - Macarena B Gonzalez
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia
| | - Takashi Umehara
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Robert J Norman
- Robinson Research Institute, Adelaide Medical School; The University of Adelaide, Adelaide, South Australia, Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Biomedicine; The University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
16
|
Cheng H, Zhang X, Li Y, Cao D, Luo C, Zhang Q, Zhang S, Jiao Y. Age-related testosterone decline: mechanisms and intervention strategies. Reprod Biol Endocrinol 2024; 22:144. [PMID: 39543598 PMCID: PMC11562514 DOI: 10.1186/s12958-024-01316-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Contemporary societies exhibit delayed reproductive age and increased life expectancy. While the male reproductive system demonstrates relatively delayed aging compared to that of females, increasing age substantially impacts its function. A characteristic manifestation is age-induced testosterone decline. Testosterone, a crucial male sex hormone, plays pivotal roles in spermatogenesis and sexual function, and contributes significantly to metabolism, psychology, and cardiovascular health. Aging exerts profound effects on the hypothalamic-pituitary-gonadal axis and Leydig cells, precipitating testosterone reduction, which adversely affects male health. Exogenous testosterone supplementation can partially ameliorate age-related testosterone deficiency; however, its long-term safety remains contentious. Preserving endogenous testosterone production capacity during the aging process warrants further investigation as a potential intervention strategy.
Collapse
Affiliation(s)
- Haoyang Cheng
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoyan Zhang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Yongheng Li
- Jiading Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Dezhong Cao
- First People's Hospital of Dongcheng District, Beijing, China
| | - Chenglong Luo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Sizheng Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongzheng Jiao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
17
|
Lv ZM, Liu C, Wang P, Chen YH. Dysregulation of mitochondrial dynamics and mitophagy are involved in high-fat diet-induced steroidogenesis inhibition. J Lipid Res 2024; 65:100639. [PMID: 39236859 PMCID: PMC11467671 DOI: 10.1016/j.jlr.2024.100639] [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: 03/05/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
Male obesity is a pandemic health issue and can disrupt testicular steroidogenesis. Here, we explored the mechanism by which a high-fat diet (HFD) induced steroidogenic inhibition. As expected, HFD induced lipid droplet accumulation and reduced the expression of StAR, P450scc, and 3β-HSD, three steroidogenic enzymes, in mouse testes. Palmitic acid (PA), a saturated fatty acid usually used to trigger lipotoxicity in vitro, induced greater accumulation of lipid droplets and the downregulation of steroidogenic enzymes in TM3 cells. Mechanistically, both HFD and PA disturbed mitochondrial fusion/fission dynamics and then induced mitochondrial dysfunction and mitophagy inhibition in mouse Leydig cells. Additionally, mitochondrial fusion promoter M1 attenuated PA-induced imbalance of mitochondrial dynamics, mitophagy inhibition, mitochondrial reactive oxygen species (ROS) production, and mitochondrial dysfunction in TM3 cells. Mitofusin 2 (Mfn2) knock-down further aggravated the PA-induced imbalance of mitochondrial dynamics, mitochondrial ROS production, and mitochondrial dysfunction in TM3 cells. Importantly, M1 rescued PA-induced downregulation of steroidogenic enzymes, whereas Mfn2 knock-down further aggravated PA-induced downregulation of steroidogenic enzymes in TM3 cells. Overall, our results provide laboratory evidence that mitochondrial dysfunction and mitophagy inhibition caused by dysregulation of mitochondrial fusion may be involved in HFD-induced steroidogenesis inhibition in mouse Leydig cells.
Collapse
Affiliation(s)
- Zheng-Mei Lv
- Department of Histology and Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Chao Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ping Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Yuan-Hua Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
18
|
Chi A, Yang B, Dai H, Li X, Mo J, Gao Y, Chen Z, Feng X, Ma M, Li Y, Yang C, Liu J, Liu H, Wang Z, Gao F, Liao Y, Shi X, Deng C, Zhang M. Stem Leydig cells support macrophage immunological homeostasis through mitochondrial transfer in mice. Nat Commun 2024; 15:2120. [PMID: 38459012 PMCID: PMC10924100 DOI: 10.1038/s41467-024-46190-2] [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: 06/03/2023] [Accepted: 02/16/2024] [Indexed: 03/10/2024] Open
Abstract
As testicular mesenchymal stromal cells, stem Leydig cells (SLCs) show great promise in the treatment of male hypogonadism. The therapeutic functions of mesenchymal stromal cells are largely determined by their reciprocal regulation by immune responses. However, the immunoregulatory properties of SLCs remain unclear. Here, we observe that SLCs transplantation restore male fertility and testosterone production in an ischemia‒reperfusion injury mouse model. SLCs prevent inflammatory cascades through mitochondrial transfer to macrophages. Reactive oxygen species (ROS) released from activated macrophages inducing mitochondrial transfer from SLCs to macrophages in a transient receptor potential cation channel subfamily member 7 (TRPM7)-mediated manner. Notably, knockdown of TRPM7 in transplanted SLCs compromised therapeutic outcomes in both testicular ischemia‒reperfusion and testicular aging mouse models. These findings reveal a new mechanism of SLCs transplantation that may contribute to preserve testis function in male patients with hypogonadism related to immune disorders.
Collapse
Affiliation(s)
- Ani Chi
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Bicheng Yang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Dai
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Xinyu Li
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiahui Mo
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yong Gao
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhihong Chen
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin Feng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Menghui Ma
- Center of Reproductive Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yanqing Li
- Center of Reproductive Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Chao Yang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Jie Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Hanchao Liu
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhenqing Wang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Feng Gao
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan Liao
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Xuetao Shi
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province South China University of Technology, Guangzhou, 510640, China.
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, 518054, China.
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Min Zhang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
19
|
Liu W, Du L, Cui Y, He C, He Z. WNT5A regulates the proliferation, apoptosis and stemness of human stem Leydig cells via the β-catenin signaling pathway. Cell Mol Life Sci 2024; 81:93. [PMID: 38367191 DOI: 10.1007/s00018-023-05077-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/20/2023] [Accepted: 11/16/2023] [Indexed: 02/19/2024]
Abstract
Stem Leydig cells (SLCs) are essential for maintaining normal spermatogenesis as the significant component of testis microenvironment and gonadal aging. Although progress has been achieved in the regulation of male germ cells in mammals and humans, it remains unknown about the genes and signaling pathways of human SLCs. Here we have demonstrated, for the first time, that WNT5A (Wnt family member 5a) mediates the proliferation, apoptosis, and stemness of human SLCs, namely NGFR+ Leydig cells. We revealed that NGFR+ Leydig cells expressed NGFR, PDGFRA, NES, NR2F2, and THY1, hallmarks for SLCs. RNA-sequencing showed that WNT5A was expressed at a higher level in human SLCs than non-SLCs, while immunohistochemistry and Western blots further illustrated that WNT5A was predominantly expressed in human SLCs. Notably, CCK-8, EdU and Western blots displayed that WNT5A enhanced the proliferation and DNA synthesis and retained stemness of human SLCs, whereas flow cytometry and TUNEL analyses demonstrated that WNT5A inhibited the apoptosis of these cells. WNT5A knockdown caused an increase in LC lineage differentiation of human SLCs and reversed the effect of WNT5A overexpression on fate decisions of human SLCs. In addition, WNT5A silencing resulted in the decreases in nuclear translocation of β-catenin and expression levels of c-Myc, CD44, and Cyclin D1. Collectively, these results implicate that WNT5A regulates the proliferation, apoptosis and stemness of human SLCs through the activation of the β-catenin signaling pathway. This study thus provides a novel molecular mechanism underlying the fate determinations of human SLCs, and it offers a new insight into the niche regulation of human testis.
Collapse
Affiliation(s)
- Wei Liu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Li Du
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yinghong Cui
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Caimei He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Hunan Normal University School of Medicine, Changsha, 410013, Hunan, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
20
|
Schwarz N, Leube RE. Plasticity of cytoplasmic intermediate filament architecture determines cellular functions. Curr Opin Cell Biol 2023; 85:102270. [PMID: 37918274 DOI: 10.1016/j.ceb.2023.102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023]
Abstract
Cytoplasmic intermediate filaments endow cells with mechanical stability. They are subject to changes in morphology and composition if needed. This remodeling encompasses entire cells but can also be restricted to specific intracellular regions. Intermediate filaments thereby support spatially and temporally defined cell type-specific functions. This review focuses on recent advances in our understanding of how intermediate filament dynamics affect the underlying regulatory pathways. We will elaborate on the role of intermediate filaments for the formation and maintenance of surface specializations, cell migration, contractility, organelle positioning, nucleus protection, stress responses and axonal conduction velocity. Together, the selected examples highlight the modulatory role of intermediate filament plasticity for multiple cellular functions.
Collapse
Affiliation(s)
- Nicole Schwarz
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany.
| |
Collapse
|
21
|
Qigen X, Haiming C, Kai X, Yong G, Chunhua D. Prenatal DEHP Exposure Induces Premature Testicular Aging by Promoting Leydig Cell Senescence through the MAPK Signaling Pathways. Adv Biol (Weinh) 2023; 7:e2300130. [PMID: 37246248 DOI: 10.1002/adbi.202300130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/02/2023] [Indexed: 05/30/2023]
Abstract
Previous studies show that prenatal di-(2-ethylhexyl) phthalate (DEHP) exposure induces premature testicular aging. However, the evidence is weak, and the underlying mechanisms remain unclear. p38/extracellular signal-regulated kinase (ERK)/c-Jun NH(2)-terminal kinase (JNK) MAPK pathways participate in aging. Leydig cell (LC) senescence results in testicular aging. Whether prenatal DEHP exposure induces premature testicular aging by promoting LC senescence warrants further study. Here, male mice undergo prenatal exposure to 500 mg per kg per day DEHP, and TM3 LCs are treated with 200 µm mono (2-ethylhexyl) phthalate (MEHP). MAPK pathways, testicular toxicity, and senescent phenotypes (β-gal activity, p21, p16, and cell cycle) of male mice and LCs are explored. Prenatal DEHP exposure induces premature testicular aging in middle-aged mice (poor genital development, reduced testosterone synthesis, poor semen quality, increased β-gal activity, and upregulated expression of p21 and p16). MEHP induces LCs senescence (cell cycle arrest, increased β-gal activity, and upregulated expression of p21). p38 and JNK pathways are activated, and the ERK pathway is inactivated. In conclusion, prenatal DEHP exposure induces premature testicular aging by promoting LC senescence through MAPK signaling pathways.
Collapse
Affiliation(s)
- Xie Qigen
- Department of Pediatric Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
- Department of Andrology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Cao Haiming
- Department of Andrology, Reproductive Center of the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518000, China
| | - Xia Kai
- Department of Andrology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Gao Yong
- Department of Andrology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Deng Chunhua
- Department of Andrology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| |
Collapse
|
22
|
Monageng E, Offor U, Takalani NB, Mohlala K, Opuwari CS. A Review on the Impact of Oxidative Stress and Medicinal Plants on Leydig Cells. Antioxidants (Basel) 2023; 12:1559. [PMID: 37627554 PMCID: PMC10451682 DOI: 10.3390/antiox12081559] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/03/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Leydig cells are essential for steroidogenesis and spermatogenesis. An imbalance in the production of reactive oxygen species (ROS) and the cellular antioxidant level brings about oxidative stress. Oxidative stress (OS) results in the dysfunction of Leydig cells, thereby impairing steroidogenesis, spermatogenesis, and ultimately, male infertility. To prevent Leydig cells from oxidative insults, there needs to be a balance between the ROS production and the cellular protective capacity of antioxidants. Evidence indicates that medicinal plants could improve Leydig cell function at specific concentrations under basal or OS conditions. The increased usage of medicinal plants has been considered a possible alternative treatment for male infertility. This review aims to provide an overview of the impact of oxidative stress on Leydig cells as well as the effects of various medicinal plant extracts on TM3 Leydig cells. The medicinal plants of interest include Aspalathus linearis, Camellia sinensis, Moringa oleifera, Morinda officinale, Taraxacum officinale, Trichilia emetica, Terminalia sambesiaca, Peltophorum africanum, Ximenia caffra, Serenoa repens, Zingiber officinale, Eugenia jambolana, and a combination of dandelion and fermented rooibos (CRS-10). According to the findings obtained from studies conducted on the evaluated medicinal plants, it can, therefore, be concluded that the medicinal plants maintain the antioxidant profile of Leydig cells under basal conditions and have protective or restorative effects following exposure to oxidative stress. The available data suggest that the protective role exhibited by the evaluated plants may be attributed to their antioxidant content. Additionally, the use of the optimal dosage or concentration of the extracts in the management of oxidative stress is of the utmost importance, and the measurement of their oxidation reduction potential is recommended.
Collapse
Affiliation(s)
- Elizabeth Monageng
- Department of Medical Biosciences, Faculty of Natural Science, University of Western Cape, Cape Town 7535, South Africa
| | - Ugochukwu Offor
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Ndivhuho Beauty Takalani
- Department of Medical Biosciences, Faculty of Natural Science, University of Western Cape, Cape Town 7535, South Africa
| | - Kutullo Mohlala
- Department of Medical Biosciences, Faculty of Natural Science, University of Western Cape, Cape Town 7535, South Africa
| | - Chinyerum Sylvia Opuwari
- Department of Medical Biosciences, Faculty of Natural Science, University of Western Cape, Cape Town 7535, South Africa
| |
Collapse
|
23
|
Zhang KY, Li CN, Zhang NX, Gao XC, Shen JM, Cheng DD, Wang YL, Zhang H, Lv JW, Sun JM. UPLC-QE-Orbitrap-Based Cell Metabolomics and Network Pharmacology to Reveal the Mechanism of N-Benzylhexadecanamide Isolated from Maca ( Lepidium meyenii Walp.) against Testicular Dysfunction. Molecules 2023; 28:molecules28104064. [PMID: 37241805 DOI: 10.3390/molecules28104064] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Testicular dysfunction (TDF) is characterized by testosterone deficiency and is caused by oxidative stress injury in Leydig cells. A natural fatty amide named N-benzylhexadecanamide (NBH), derived from cruciferous maca, has been shown to promote testosterone production. Our study aims to reveal the anti-TDF effect of NBH and explore its potential mechanism in vitro. This study examined the effects of H2O2 on cell viability and testosterone levels in mouse Leydig cells (TM3) under oxidative stress. In addition, cell metabolomics analysis based on UPLC-Q-Exactive-MS/MS showed that NBH was mainly involved in arginine biosynthesis, aminoacyl-tRNA biosynthesis, phenylalanine, tyrosine and tryptophan biosynthesis, the TCA cycle and other metabolic pathways by affecting 23 differential metabolites, including arginine and phenylalanine. Furthermore, we also performed network pharmacological analysis to observe the key protein targets in NBH treatment. The results showed that its role was to up-regulate ALOX5, down-regulate CYP1A2, and play a role in promoting testicular activity by participating in the steroid hormone biosynthesis pathway. In summary, our study not only provides new insights into the biochemical mechanisms of natural compounds in the treatment of TDF, but also provides a research strategy that integrates cell metabolomics and network pharmacology in order to promote the screening of new drugs for the treatment of TDF.
Collapse
Affiliation(s)
- Kai-Yue Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Chun-Nan Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Nan-Xi Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xiao-Chen Gao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jia-Ming Shen
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Duan-Duan Cheng
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yue-Long Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Hui Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jing-Wei Lv
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jia-Ming Sun
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|