1
|
Gałęska E, Kowalczyk A, Wrzecińska M, García MC, Czerniawska-Piątkowska E, Gwoździewicz S, Witkiewicz W, Dobrzański Z. The Importance of Mitochondrial Processes in the Maturation and Acquisition of Competences of Oocytes and Embryo Culture. Int J Mol Sci 2025; 26:4098. [PMID: 40362337 PMCID: PMC12071309 DOI: 10.3390/ijms26094098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondria, as multifunctional and partially independent structures, play a crucial role in determining essential life processes. Recently, their significance in reproductive biology has gained increasing attention. This review aims to comprehensively analyse the role of mitochondrial processes in oocyte maturation and embryo culture. A comprehensive literature review was conducted to highlight the importance of mitochondrial activity in the early stages of life formation. Proper mitochondrial function provides energy, maintains genomic stability, and ensures optimal conditions for fertilisation and embryo progression. Understanding these processes is essential to optimise culture conditions and identify new mitochondrial biomarkers that improve reproductive success and improve assisted reproductive technologies (ARTs). Enhancing mitochondrial function in female reproductive cells is the key to improving oocyte and embryo quality, which can lead to better in vitro fertilisation and embryo transfer. Furthermore, advances in diagnostic techniques, such as mitochondrial genome sequencing, offer a more precise understanding of the relationship between mitochondrial health and oocyte quality. However, fully understanding mitochondrial functions is only part of the challenge. Expanding knowledge of the interactions between mitochondria and other cellular structures is crucial for future advancements in reproductive medicine. Understanding these complex relationships will provide deeper insight into improving reproductive outcomes and embryo development.
Collapse
Affiliation(s)
- Elżbieta Gałęska
- Department of Environment Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, 51-630 Wrocław, Poland; (E.G.); (M.W.); (Z.D.)
| | - Alicja Kowalczyk
- Department of Environment Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, 51-630 Wrocław, Poland; (E.G.); (M.W.); (Z.D.)
| | - Marcjanna Wrzecińska
- Department of Environment Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, 51-630 Wrocław, Poland; (E.G.); (M.W.); (Z.D.)
| | - Mercedes Camiña García
- Department of Physiology, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain;
| | - Ewa Czerniawska-Piątkowska
- Department of Ruminant Science, West Pomeranian University of Technology in Szczecin, 70-310 Szczecin, Poland;
| | - Szymon Gwoździewicz
- Institute of Animal Breeding, Wrocław University of Environmental and Life Sciences, 51-630 Wrocław, Poland
| | - Wojciech Witkiewicz
- Research and Development Center, Voivodeship Specialist Hospital in Wrocław, 51-124 Wrocław, Poland;
| | - Zbigniew Dobrzański
- Department of Environment Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, 51-630 Wrocław, Poland; (E.G.); (M.W.); (Z.D.)
| |
Collapse
|
2
|
Nishigaki A, Kida N, Tsubokura H, Yoshimura M, Shimoi K, Yoshida A, Okada H. Effects of cigarette smoke extract on angiogenesis and aromatase activity in KGN cells. Sci Rep 2025; 15:13967. [PMID: 40263391 PMCID: PMC12015283 DOI: 10.1038/s41598-025-98830-2] [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: 01/11/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Cigarette smoking is associated with negative reproductive outcomes and an increased likelihood of failed in vitro fertilization. We investigated the impact of cigarette smoke on angiogenesis and aromatase activity in human granulosa cells. The human ovarian granulosa-like tumor cell line (KGN) was exposed to different cigarette smoke extract (CSE) concentrations and/or 10-μM forskolin. mRNA and protein levels and their secretion were measured using real-time polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay techniques, respectively. Additionally, KGN cells were transfected with small interfering RNA (siRNA) targeting hypoxia-inducible factor-1α (HIF-1α) for 24 h, followed by treatment with or without (control) CSE for 48 h. Data were statistically analyzed. Compared with the control group, the CSE group showed significantly increased vascular endothelial growth factor (VEGF), N-myc downstream-regulated gene 1 (NDRG1), and HIF-1α expression. Overexpression of VEGF and NDRG1 induced by CSE was inhibited by HIF-1α siRNA transfection. Furthermore, CSE exposure decreased progesterone and estradiol levels and increased testosterone levels. It also affected the expression of genes associated with steroid hormone synthesis, decreased anti-Müllerian hormone and anti-Müllerian hormone receptor II levels, and caused SMAD signaling pathway desensitization. Thus, CSE adversely affects the maturation of human granulosa cells, affecting both angiogenesis and aromatase activity.
Collapse
Affiliation(s)
- Akemi Nishigaki
- Department of Obstetrics and Gynecology, Kansai Medical University, 2-5-1 Shinmachi-cho, Hirakata, Osaka, 573-1010, Japan
| | - Naoko Kida
- KAWA Ladies Clinic, 3-2-3 Wakamatsudai, Minami Ward, Sakai, 590-0116, Japan
| | - Hiroaki Tsubokura
- Department of Obstetrics and Gynecology, Kansai Medical University, 2-5-1 Shinmachi-cho, Hirakata, Osaka, 573-1010, Japan
| | - Masahiro Yoshimura
- Kansai Medical University Hirakata Hospital, 2-3-1 Shinmachi-cho, Hirakata, 573-1191, Japan
| | - Kayo Shimoi
- Kansai Medical University Hirakata Hospital, 2-3-1 Shinmachi-cho, Hirakata, 573-1191, Japan
| | - Aya Yoshida
- Department of Obstetrics and Gynecology, Kansai Medical University, 2-5-1 Shinmachi-cho, Hirakata, Osaka, 573-1010, Japan
| | - Hidetaka Okada
- Department of Obstetrics and Gynecology, Kansai Medical University, 2-5-1 Shinmachi-cho, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|
3
|
Hashim NT, Babiker R, Chaitanya NCSK, Mohammed R, Priya SP, Padmanabhan V, Ahmed A, Dasnadi SP, Islam MS, Gismalla BG, Rahman MM. New Insights in Natural Bioactive Compounds for Periodontal Disease: Advanced Molecular Mechanisms and Therapeutic Potential. Molecules 2025; 30:807. [PMID: 40005119 PMCID: PMC11858609 DOI: 10.3390/molecules30040807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/18/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Periodontal disease is a chronic inflammatory condition that destroys the tooth-supporting structures due to the host's immune response to microbial biofilms. Traditional periodontal treatments, such as scaling and root planing, pharmacological interventions, and surgical procedures, have significant limitations, including difficulty accessing deep periodontal pockets, biofilm recolonization, and the development of antibiotic resistance. In light of these challenges, natural bioactive compounds derived from plants, herbs, and other natural sources offer a promising alternative due to their anti-inflammatory, antioxidant, antimicrobial, and tissue-regenerative properties. This review focuses on the molecular mechanisms through which bioactive compounds, such as curcumin, resveratrol, epigallocatechin gallate (EGCG), baicalin, carvacrol, berberine, essential oils, and Gum Arabic, exert therapeutic effects in periodontal disease. Bioactive compounds inhibit critical inflammatory pathways like NF-κB, JAK/STAT, and MAPK while activating protective pathways such as Nrf2/ARE, reducing cytokine production and oxidative stress. They also inhibit the activity of matrix metalloproteinases (MMPs), preventing tissue degradation and promoting healing. In addition, these compounds have demonstrated the potential to disrupt bacterial biofilms by interfering with quorum sensing, targeting bacterial cell membranes, and enhancing antibiotic efficacy.Bioactive compounds also modulate the immune system by shifting the balance from pro-inflammatory to anti-inflammatory responses and promoting efferocytosis, which helps resolve inflammation and supports tissue regeneration. However, despite the promising potential of these compounds, challenges related to their poor bioavailability, stability in the oral cavity, and the absence of large-scale clinical trials need to be addressed. Future strategies should prioritize the development of advanced delivery systems like nanoparticles and hydrogels to enhance bioavailability and sustain release, alongside long-term studies to assess the effects of these compounds in human populations. Furthermore, combining bioactive compounds with traditional treatments could provide synergistic benefits in managing periodontal disease. This review aims to explore the therapeutic potential of natural bioactive compounds in managing periodontal disease, emphasizing their molecular mechanisms of action and offering insights into their integration with conventional therapies for a more comprehensive approach to periodontal health.
Collapse
Affiliation(s)
- Nada Tawfig Hashim
- Department of Periodontics, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Rasha Babiker
- Department of Physiology, RAK College of Medical Sciences, RAK Medical & Health Science University, Ras-AlKhaimah 11127, United Arab Emirates;
| | - Nallan C. S. K. Chaitanya
- Department of Oral Medicine and Radiology, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Riham Mohammed
- Department Oral Surgery, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Sivan Padma Priya
- Oral Pathology Department, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Vivek Padmanabhan
- Department of Pediatric and Preventive Dentistry, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Ayman Ahmed
- Department of Periodontology and Implantology, Nile University, Khartoum 1847, Sudan;
| | - Shahista Parveen Dasnadi
- Department of Orthodontics, RAK College of Dental, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Md Sofiqul Islam
- Department of Operative Dentistry, RAK College of Dental Sciences, RAK Medical and Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| | - Bakri Gobara Gismalla
- Department of Oral Rehabilitation, Faculty of Dentistry, University of Khartoum, Khartoum 11115, Sudan;
| | - Muhammed Mustahsen Rahman
- Department of Periodontics, RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras-AlKhaimah 12973, United Arab Emirates;
| |
Collapse
|
4
|
Nishigaki A, Ishida M, Tsubokura H, Hisamatsu Y, Hirose Y, Okada H. HIF-1α Promotes Luteinization via NDRG1 Induction in the Human Ovary. Biomedicines 2025; 13:328. [PMID: 40002742 PMCID: PMC11852844 DOI: 10.3390/biomedicines13020328] [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: 12/09/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that plays a crucial role in various physiological and pathological processes of the ovary. However, the timing of HIF-1α expression and its specific biological function in the follicular development of the human ovary remain unclear. Therefore, in this study, we aimed to examine whether HIF-1α and its downstream gene, N-myc downstream-regulated gene 1 (NDRG1), exhibit stage-specific expression during the follicular development process in the human ovary. Methods: We used ovarian tissues from eight women with regular menstrual cycles who were not undergoing hormonal treatment. We investigated HIF-1α and NDRG1 expression and localization using immunohistochemistry. Further, we transfected human ovarian granulosa (KGN) cells with HIF-1α small interfering RNA (siRNA) to investigate the influence of HIF-1α on NDRG1 expression and progesterone synthesis. Results: The immunohistochemical analysis of human ovarian tissues revealed that HIF-1α was localized in the cytoplasm of granulosa cells (GCs) at both the primary and secondary follicular stages. Conversely, in tertiary and later developmental stages, HIF-1α was observed exclusively in the nucleus of GCs. Furthermore, while NDRG1 was not detected in primary follicles, it was present in all GCs beyond the tertiary stage. Notably, transfection of KGN cells with HIF-1α siRNA significantly decreased NDRG1 expression, at both the mRNA and protein levels, and in progesterone synthesis. Conclusion: Our results indicate that HIF-1α and NDRG1 are integral to follicular development and the early luteinization of pre-ovulatory follicles.
Collapse
Affiliation(s)
- Akemi Nishigaki
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Osaka, Japan;
| | - Hiroaki Tsubokura
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Yoji Hisamatsu
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Takatsuki 569-8686, Osaka, Japan;
| | - Hidetaka Okada
- Department of Obstetrics and Gynecology, Kansai Medical University, Hirakata 573-1191, Osaka, Japan; (A.N.); (H.T.); (Y.H.); (H.O.)
| |
Collapse
|
5
|
Liu X, Li R, Xiu Z, Tang S, Duan Y. Toxicity mechanism of acrolein on energy metabolism disorder and apoptosis in human ovarian granulosa cells. Toxicology 2024; 506:153861. [PMID: 38866128 DOI: 10.1016/j.tox.2024.153861] [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: 03/21/2024] [Revised: 05/26/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Acrolein (ACR), an unsaturated, highly reactive aldehyde, is a widespread environmental toxin. ACR exerts permanent and irreversible side effects on ovarian functions. Granulosa cells play a crucial role in supporting ovarian function. Thus, in this study, we investigated the toxicity effects of granulosa cells induced by ACR. Following treatment with varying ACR concentrations (0, 12.5, 25, 50, and 100 μM), we observed that ACR exposure induced reactive oxygen species accumulation, mitochondrial energy metabolism disorder, and apoptosis in KGN cells (a human ovarian granulosa cell line) in a dose-dependent manner. In addition, mitochondrial biogenesis in KGN cells displayed biphasic changes after ACR exposure, with activation at a low ACR dose (12.5 μM), but inhibition at higher ACR doses (≥50 μM). SIRT1/PGC-1α-mediated mitochondrial biogenesis is crucial for maintaining intracellular mitochondrial homeostasis and cellular function. The inhibition/activation of the SIRT1/PGC-1α pathway in KGN cells validated its role in ACR-induced damage. The results indicated that the inhibition of the SIRT1/PGC-1α pathway aggravated ACR-induced cell damage, whereas its activation partially counteracted ACR-induced cell damage. This study attempted to uncover a novel mechanism of ACR-induced ovarian toxicity so as to provide an effective treatment option for safeguarding female reproductive health from the adverse effects of ACR.
Collapse
Affiliation(s)
- Xueping Liu
- College of Basic Medical, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China
| | - Rongxia Li
- College of Basic Medical, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Gynecology Medicine, The Second Hospital of Hebei Medicine University, Shijiazhuang, Hebei Province 050004, China
| | - Zi Xiu
- College of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050200, China
| | - Siling Tang
- College of Basic Medical, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China
| | - Yancang Duan
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Hebei Collaborative Innovation Center of Integrated Chinese and Western Medicine on Reproductive Disease, Shijiazhuang, Hebei Province 050091, China; Hebei Key Laboratory of Integrative Medicine on Liver-kidney Patterns, Shijiazhuang, Hebei Province 050091, China.
| |
Collapse
|
6
|
An Z, Xie C, Lu H, Wang S, Zhang X, Yu W, Guo X, Liu Z, Shang D, Wang X. Mitochondrial Morphology and Function Abnormality in Ovarian Granulosa Cells of Patients with Diminished Ovarian Reserve. Reprod Sci 2024; 31:2009-2020. [PMID: 38294667 DOI: 10.1007/s43032-024-01459-1] [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: 10/19/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024]
Abstract
In this study, we examined the changes in the mitochondrial structure and function in cumulus granulosa cells of patients with diminished ovarian reserve (DOR) to explore the causes and mechanisms of decreased mitochondrial quality. The mitochondrial ultrastructure was observed by transmission electron microscope, and the function was determined by detecting the ATP content, reactive oxygen species (ROS) levels, the number of mitochondria, and the mitochondrial membrane potential. The expression of ATP synthases in relation to mitochondrial function was analyzed. Additionally, protein immunoblotting was used to compare the expression levels of mitochondrial kinetic protein, the related channel protein in the two groups. Patients with DOR had abnormal granulosa cell morphology, increased mitochondrial abnormalities, decreased mitochondrial function, and disturbed mitochondrial dynamics. Additionally, the silent information regulator 1 (SIRT1)/phospho-AMP-activated protein kinase (P-AMPK)-peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) pathway expression was decreased, which was speculated to be associated with the decreased mitochondrial mass in the DOR group. The mitochondrial mass was decreased in granulosa cells of patients in the DOR group. The mitochondrial dysfunction observed in granulosa cells of patients in the DOR group may be associated with dysregulation of the SIRT1/P-AMPK-PGC-1α-mitochondrial transcription factor A (TFAM) pathway.
Collapse
Affiliation(s)
- Zhuo An
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China
- Hebei Medical University, No. 361 Zhongshan Road, Chang'An District, Shijiazhuang, 050017, China
| | - Congcong Xie
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China
| | - Hui Lu
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China
| | - Shusong Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China
| | - Xiujia Zhang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China
| | - Wenbo Yu
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China
| | - Xiaoli Guo
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China
| | - Zehao Liu
- Hebei Children's Hospital, Shijiazhuang, 050031, China
| | - Dandan Shang
- Hebei Medical University, No. 361 Zhongshan Road, Chang'An District, Shijiazhuang, 050017, China.
| | - Xueying Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Road, Xinhua District, Shijiazhuang, 050071, China.
| |
Collapse
|
7
|
Wu X, Zhou Y, Xi Y, Zhou H, Tang Z, Xiong L, Qin D. Polyphenols: Natural Food-Grade Biomolecules for the Treatment of Nervous System Diseases from a Multi-Target Perspective. Pharmaceuticals (Basel) 2024; 17:775. [PMID: 38931442 PMCID: PMC11206395 DOI: 10.3390/ph17060775] [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: 05/24/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Polyphenols are the most prevalent naturally occurring phytochemicals in the human diet and range in complexity from simple molecules to high-molecular-weight polymers. They have a broad range of chemical structures and are generally categorized as "neuroprotective", "anti-inflammatory", and "antioxidant" given their main function of halting disease onset and promoting health. Research has shown that some polyphenols and their metabolites can penetrate the blood-brain barrier and hence increase neuroprotective signaling and neurohormonal effects to provide anti-inflammatory and antioxidant effects. Therefore, multi-targeted modulation of polyphenols may prevent the progression of neuropsychiatric disorders and provide a new practical therapeutic strategy for difficult-to-treat neuropsychiatric disorders. Therefore, multi-target modulation of polyphenols has the potential to prevent the progression of neuropsychiatric disorders and provide a new practical therapeutic strategy for such nervous system diseases. Herein, we review the therapeutic benefits of polyphenols on autism-spectrum disorders, anxiety disorders, depression, and sleep disorders, along with in vitro and ex vivo experimental and clinical trials. Although their methods of action are still under investigation, polyphenols are still seldom employed directly as therapeutic agents for nervous system disorders. Comprehensive mechanistic investigations and large-scale multicenter randomized controlled trials are required to properly evaluate the safety, effectiveness, and side effects of polyphenols.
Collapse
Affiliation(s)
- Xinchen Wu
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Yang Zhou
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Yujiang Xi
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Haimei Zhou
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (H.Z.); (Z.T.)
| | - Zhengxiu Tang
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (H.Z.); (Z.T.)
| | - Lei Xiong
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China; (X.W.); (Y.Z.); (Y.X.)
| | - Dongdong Qin
- School of Basic Medical Science, Yunnan University of Chinese Medicine, Kunming 650500, China; (H.Z.); (Z.T.)
- Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Neuropsychiatric Diseases, Yunnan University of Chinese Medicine, Kunming 650500, China
| |
Collapse
|
8
|
Tang W, Zhu X, Chen Y, Yang S, Wu C, Chen D, Xue L, Guo Y, Dai Y, Wei S, Wu M, Wu M, Wang S. Towards prolonging ovarian reproductive life: Insights into trace elements homeostasis. Ageing Res Rev 2024; 97:102311. [PMID: 38636559 DOI: 10.1016/j.arr.2024.102311] [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: 12/04/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Ovarian aging is marked by a reduction in the quantity and quality of ovarian follicles, leading to a decline in female fertility and ovarian endocrine function. While the biological characteristics of ovarian aging are well-established, the exact mechanisms underlying this process remain elusive. Recent studies underscore the vital role of trace elements (TEs) in maintaining ovarian function. Imbalances in TEs can lead to ovarian aging, characterized by reduced enzyme activity, hormonal imbalances, ovulatory disorders, and decreased fertility. A comprehensive understanding of the relationship between systemic and cellular TEs balance and ovarian aging is critical for developing treatments to delay aging and manage age-related conditions. This review consolidates current insights into TEs homeostasis and its impact on ovarian aging, assesses how altered TEs metabolism affects ovarian aging, and suggests future research directions to prolong ovarian reproductive life. These studies are expected to offer novel approaches for mitigating ovarian aging.
Collapse
Affiliation(s)
- Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Xiaoran Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Shuhong Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China
| | - Mingfu Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| |
Collapse
|
9
|
Del Bianco D, Gentile R, Sallicandro L, Biagini A, Quellari PT, Gliozheni E, Sabbatini P, Ragonese F, Malvasi A, D’Amato A, Baldini GM, Trojano G, Tinelli A, Fioretti B. Electro-Metabolic Coupling of Cumulus-Oocyte Complex. Int J Mol Sci 2024; 25:5349. [PMID: 38791387 PMCID: PMC11120766 DOI: 10.3390/ijms25105349] [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: 03/30/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Oocyte-cumulus cell interaction is essential for oocyte maturation and competence. The bidirectional crosstalk network mediated by gap junctions is fundamental for the metabolic cooperation between these cells. As cumulus cells exhibit a more glycolytic phenotype, they can provide metabolic substrates that the oocyte can use to produce ATP via oxidative phosphorylation. The impairment of mitochondrial activity plays a crucial role in ovarian aging and, thus, in fertility, determining the success or failure of assisted reproductive techniques. This review aims to deepen the knowledge about the electro-metabolic coupling of the cumulus-oocyte complex and to hypothesize a putative role of potassium channel modulators in order to improve fertility, promote intracellular Ca2+ influx, and increase the mitochondrial biogenesis and resulting ATP levels in cumulus cells.
Collapse
Affiliation(s)
- Diletta Del Bianco
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
| | - Rosaria Gentile
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Laboratorio Interdipartimentale di Fisiopatologia della Riproduzione, Università degli Studi di Perugia, Edificio C, Piano 3 P.zza Lucio Severi, 1, Sant’Andrea delle Fratte, 06132 Perugia, Italy
| | - Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Biagini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Paola Tiziana Quellari
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
- ASST Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy
| | - Elko Gliozheni
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tirana, AL1005 Tirana, Albania
| | - Paola Sabbatini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Laboratorio Interdipartimentale di Fisiopatologia della Riproduzione, Università degli Studi di Perugia, Edificio C, Piano 3 P.zza Lucio Severi, 1, Sant’Andrea delle Fratte, 06132 Perugia, Italy
| | - Antonio Malvasi
- Department of Biomedical Sciences and Human Oncology, University of Bari, 70121 Bari, Italy;
| | - Antonio D’Amato
- 1st Unit of Obstetrics and Gynecology, University of Bari, 70121 Bari, Italy;
| | | | - Giuseppe Trojano
- Department of Maternal and Child Health, “Madonna delle Grazie” Hospital ASM, 75100 Matera, Italy;
| | - Andrea Tinelli
- Department of Obstetrics and Gynecology and CERICSAL (CEntro di RIcerca Clinico SALentino), Veris delli Ponti Hospital, Via Giuseppina delli Ponti, 73020 Scorrano, Lecce, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Laboratorio Interdipartimentale di Fisiopatologia della Riproduzione, Università degli Studi di Perugia, Edificio C, Piano 3 P.zza Lucio Severi, 1, Sant’Andrea delle Fratte, 06132 Perugia, Italy
| |
Collapse
|
10
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
11
|
Ruparelia AA, Salavaty A, Barlow CK, Lu Y, Sonntag C, Hersey L, Eramo MJ, Krug J, Reuter H, Schittenhelm RB, Ramialison M, Cox A, Ryan MT, Creek DJ, Englert C, Currie PD. The African killifish: A short-lived vertebrate model to study the biology of sarcopenia and longevity. Aging Cell 2024; 23:e13862. [PMID: 37183563 PMCID: PMC10776123 DOI: 10.1111/acel.13862] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/16/2023] Open
Abstract
Sarcopenia, the age-related decline in muscle function, places a considerable burden on health-care systems. While the stereotypic hallmarks of sarcopenia are well characterized, their contribution to muscle wasting remains elusive, which is partly due to the limited availability of animal models. Here, we have performed cellular and molecular characterization of skeletal muscle from the African killifish-an extremely short-lived vertebrate-revealing that while many characteristics deteriorate with increasing age, supporting the use of killifish as a model for sarcopenia research, some features surprisingly reverse to an "early-life" state in the extremely old stages. This suggests that in extremely old animals, there may be mechanisms that prevent further deterioration of skeletal muscle, contributing to an extension of life span. In line with this, we report a reduction in mortality rates in extremely old killifish. To identify mechanisms for this phenomenon, we used a systems metabolomics approach, which revealed that during aging there is a striking depletion of triglycerides, mimicking a state of calorie restriction. This results in the activation of mitohormesis, increasing Sirt1 levels, which improves lipid metabolism and maintains nutrient homeostasis in extremely old animals. Pharmacological induction of Sirt1 in aged animals was sufficient to induce a late life-like metabolic profile, supporting its role in life span extension in vertebrate populations that are naturally long-lived. Collectively, our results demonstrate that killifish are not only a novel model to study the biological processes that govern sarcopenia, but they also provide a unique vertebrate system to dissect the regulation of longevity.
Collapse
Affiliation(s)
- Avnika A. Ruparelia
- Australian Regenerative Medicine Institute, Monash UniversityClaytonAustralia
- Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine Dentistry and Health SciencesUniversity of MelbourneMelbourneAustralia
- Centre for Muscle Research, Department of Anatomy and PhysiologyUniversity of MelbourneMelbourneAustralia
| | - Adrian Salavaty
- Australian Regenerative Medicine Institute, Monash UniversityClaytonAustralia
- Systems Biology Institute Australia, Monash UniversityClaytonAustralia
| | - Christopher K. Barlow
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonAustralia
- Monash Proteomics and Metabolomics FacilityMonash Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Yansong Lu
- Australian Regenerative Medicine Institute, Monash UniversityClaytonAustralia
| | - Carmen Sonntag
- Australian Regenerative Medicine Institute, Monash UniversityClaytonAustralia
| | - Lucy Hersey
- Australian Regenerative Medicine Institute, Monash UniversityClaytonAustralia
| | - Matthew J. Eramo
- Department of Biochemistry and Molecular BiologyMonash Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Johannes Krug
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI)JenaGermany
| | - Hanna Reuter
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI)JenaGermany
| | - Ralf B. Schittenhelm
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonAustralia
- Monash Proteomics and Metabolomics FacilityMonash Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash UniversityClaytonAustralia
- Systems Biology Institute Australia, Monash UniversityClaytonAustralia
| | - Andrew Cox
- Peter MacCallum Cancer CentreMelbourneAustralia
- Department of Biochemistry and PharmacologyThe University of MelbourneMelbourneAustralia
| | - Michael T. Ryan
- Department of Biochemistry and Molecular BiologyMonash Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Darren J. Creek
- Monash Proteomics and Metabolomics FacilityMonash Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
- Drug Delivery, Disposition and DynamicsMonash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Christoph Englert
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI)JenaGermany
- Institute of Biochemistry and Biophysics, Friedrich‐Schiller‐University JenaJenaGermany
| | - Peter D. Currie
- Australian Regenerative Medicine Institute, Monash UniversityClaytonAustralia
- EMBL Australia, Victorian NodeMonash UniversityClaytonAustralia
| |
Collapse
|
12
|
Zhang Z, Tian J, Liu W, Zhou J, Zhang Y, Ding L, Sun H, Yan G, Sheng X. Perfluorooctanoic acid exposure leads to defect in follicular development through disrupting the mitochondrial electron transport chain in granulosa cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:166954. [PMID: 37722425 DOI: 10.1016/j.scitotenv.2023.166954] [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: 04/27/2023] [Revised: 08/21/2023] [Accepted: 09/08/2023] [Indexed: 09/20/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a persistent environmental pollutant that can impair ovarian function, while the underlying mechanism is not fully understood, and effective treatments are lacking. In this study, we established a mouse model of PFOA exposure induced by drinking water and found that PFOA exposure impaired follicle development, increased apoptosis of granulosa cells (GCs), and hindered normal follicular development in a 3D culture system. RNA-seq analysis revealed that PFOA disrupted oxidative phosphorylation in ovaries by impairing the mitochondrial electron transport chain. This resulted in reduced mitochondrial membrane potential and increased mitochondrial reactive oxygen species (mtROS) in isolated GCs or KGN cells. Resveratrol, a mitochondrial nutrient supplement, could improve mitochondrial function and restore normal follicular development by activating FoxO1 through SIRT1/PI3K-AKT pathway. Our results indicate that PFOA exposure impairs mitochondrial function in GCs and affects follicle development. Resveratrol can be a potential therapeutic agent for PFOA-induced ovarian dysfunction.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Jiao Tian
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Wenwen Liu
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Jidong Zhou
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yang Zhang
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Lijun Ding
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Haixiang Sun
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China.
| | - Guijun Yan
- Department of Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| | - Xiaoqiang Sheng
- Center for Reproductive Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
13
|
Ataei-Nazari S, Rahimi A, Bakhtiarizadeh MR, Jahandideh-Golroodbari P, Assadi-Alamouti A, Hajarizadeh A, Haji-Rahimi H, Mansouri-Bahrani B, Afshar-Bahrabad A, Ozturk I, Sharma M, Tvrdá E, Mohammadi-Sangcheshmeh A. Alpha-linolenic acid alleviates the detrimental effects of lipopolysaccharide during in vitro ovine oocyte development. Theriogenology 2023; 212:64-72. [PMID: 37699276 DOI: 10.1016/j.theriogenology.2023.07.038] [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: 05/02/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
During the transition period and early lactation of ruminants with higher production, the reproductive organs are exposed to various stressors, like inflammation stimulators such as lipopolysaccharides (LPS), as a consequence of high concentrate consumption. In this study, we aimed to determine the probable potential of α-linolenic acid (ALA) in alleviating LPS-induced effects in ovine oocytes in vitro as well as the underlying controlling mechanisms. Different concentrations of LPS (0, 0.01, 0.1, 1, and 10 μg/mL) were added to the oocyte maturation medium to evaluate its effect on oocyte developmental competence. Likewise, different concentrations of ALA (0, 10, 50, 100, and 200 μM/mL) were added to the maturation medium to define its effects on oocyte developmental competence. Accordingly, a combination of ALA and LPS in a dose-dependent manner was added to the maturation medium to elucidate their effect on oocyte developmental competence and uncover any possible potential of ALA to alleviate the detrimental effect induced by the presence of LPS. The expressions of candidate genes were measured in mature oocytes treated either with ALA, LPS, or ALA plus LPS. Adding LPS to the maturation medium decreased the cleavage rate of the treated oocytes, and those oocytes reached the blastocyst stage at a lower rate. Adding ALA to the maturation medium in the presence of LPS alleviated the detrimental effects of LPS in a dose-dependent manner, which ultimately led to higher cleavage and blastocyst formation. A higher expression of Trim26, GRHPR, NDUFA, PGC-1α, SOD, CS, SDH, p53, and CAT was observed in LPS-treated oocytes compared with the ALA and control groups. Additionally, CS and CAT transcripts were down-regulated in oocytes in LPS plus ALA-treated group compared to that of the LPS-treated group. These findings revealed that ALA has the potential to alleviate the detrimental effects induced by LPS on in ovine oocytes during maturation in vitro. Thus, LPS-detrimental effect and ALA-preventing mechanisms seem to be regulated through the expression of genes involved in mitochondrial biogenesis and function, oxidative stress, and antioxidant systems.
Collapse
Affiliation(s)
- Sara Ataei-Nazari
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Amin Rahimi
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran; Chaltasian Agri.-Animal Production Complex, Varamin, Tehran, Iran
| | | | | | - Ali Assadi-Alamouti
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Atieh Hajarizadeh
- Stem Cell Technology Research Center (STRC), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Hanieh Haji-Rahimi
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran; Chaltasian Agri.-Animal Production Complex, Varamin, Tehran, Iran
| | - Banafsheh Mansouri-Bahrani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran; Chaltasian Agri.-Animal Production Complex, Varamin, Tehran, Iran
| | | | - Irfan Ozturk
- Department of Animal Science, Biometry Genetics Unit, Harran University, Şanlıurfa, Turkey
| | - Manjita Sharma
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, 79409, USA
| | - Eva Tvrdá
- Institute of Biotechnology, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 94976, Nitra, Slovakia
| | - Abdollah Mohammadi-Sangcheshmeh
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran; Chaltasian Agri.-Animal Production Complex, Varamin, Tehran, Iran.
| |
Collapse
|
14
|
Adu-Agyeiwaah Y, Vieira CP, Asare-Bediako B, Li Calzi S, DuPont M, Floyd J, Boye S, Chiodo V, Busik JV, Grant MB. Intravitreal Administration of AAV2-SIRT1 Reverses Diabetic Retinopathy in a Mouse Model of Type 2 Diabetes. Transl Vis Sci Technol 2023; 12:20. [PMID: 37070938 PMCID: PMC10123324 DOI: 10.1167/tvst.12.4.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/24/2023] [Indexed: 04/19/2023] Open
Abstract
Purpose The expression of silent information regulator (SIRT) 1 is reduced in diabetic retinopathy (DR). Previous studies showed that alterations in SIRT1 messenger RNA (mRNA) and protein expression are implicated in progressive inflammation and formation of retinal acellular capillaries. Treatment with the SIRT1 agonist, SRT1720, improved visual response by restoration of a- and b-wave responses on electroretinogram scotopic measurements in diabetic (db/db) mice. In this study, we investigated the effects of intravitreal SIRT1 delivery on diabetic retinal pathology. Methods Nine-month-old db/db mice received one intravitreal injection of either AAV2-SIRT1 or AAV2-GFP control virus, and after 3 months, electroretinography and optomotor responses were measured. Their eyes were then removed and analyzed by immunohistochemistry and flow cytometry. Results SIRT1 mRNA and protein levels were increased following AAV2-SIRT1 administration compared to control virus AAV2-GFP injected mice. IBA1+ and caspase 3 expression were decreased in retinas of db/db mice injected with AAV2-SIRT1, and reductions in scotopic a- and b-waves and high spatial frequency in optokinetic response were prevented. Retinal hypoxia inducible factor 1α (HIF-1α) protein levels were reduced in the AAV2-SIRT1-injected mice compared to control-injected mice. Using flow cytometry to assess changes in intracellular HIF-1α levels, endothelial cells (CD31+) from AAV-2 SIRT1 injected mice demonstrated reduced HIF-1α expression compared to db/db mice injected with the control virus. Conclusions Intravitreal AAV2-SIRT1 delivery increased retina SIRT1 and transduced neural and endothelial cells, thus reversing functional damage and improving overall visual function. Translational Relevance AAV2-SIRT1 gene therapy represents a beneficial approach for the treatment of chronic retinal conditions such as DR.
Collapse
Affiliation(s)
- Yvonne Adu-Agyeiwaah
- Department of Vision Science, School of Optometry, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cristiano P. Vieira
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bright Asare-Bediako
- Department of Vision Science, School of Optometry, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sergio Li Calzi
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mariana DuPont
- Department of Vision Science, School of Optometry, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason Floyd
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sanford Boye
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Vince Chiodo
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Julia V. Busik
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Maria B. Grant
- Department of Ophthalmology and Visual Sciences, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
15
|
Su LY, Huang WC, Kan NW, Tung TH, Huynh LBP, Huang SY. Effects of Resveratrol on Muscle Inflammation, Energy Utilisation, and Exercise Performance in an Eccentric Contraction Exercise Mouse Model. Nutrients 2023; 15:249. [PMID: 36615906 PMCID: PMC9824440 DOI: 10.3390/nu15010249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Eccentric contraction can easily cause muscle damage and an inflammatory response, which reduces the efficiency of muscle contraction. Resveratrol causes anti-inflammatory effects in muscles, accelerates muscle repair, and promotes exercise performance after contusion recovery. However, whether resveratrol provides the same benefits for sports injuries caused by eccentric contraction is unknown. Thus, we explored the effects of resveratrol on inflammation and energy metabolism. In this study, mice were divided into four groups: a control group, an exercise group (EX), an exercise with low-dose resveratrol group (EX + RES25), and an exercise with high-dose resveratrol group (EX + RES150). The results of an exhaustion test showed that the time before exhaustion of the EX + RES150 group was greater than that of the EX group. Tumour necrosis factor-α (Tnfα) mRNA expression was lower in the EX + RES150 group than in the EX group. The energy utilisation of the EX + RES150 group was greater than that of the EX + RES25 group in different muscles. High-dose resveratrol intervention decreased Tnfα mRNA expression and enhanced the mRNA expressions of sirtuin 1, glucose transporter 4, AMP-activated protein kinase α1, and AMP-activated protein kinase α2 in muscles. These results revealed that high-dose resveratrol supplementation can reduce inflammation and oxidation and improve energy utilisation during short-duration high-intensity exercise.
Collapse
Affiliation(s)
- Liang-Yu Su
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan
| | - Wen-Ching Huang
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan
| | - Nai-Wen Kan
- Office of Physical Education Affairs, Taipei Medical University Hospital, Taipei 110301, Taiwan
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan
| | - Te-Hsuan Tung
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan
| | - Linh Ba Phuong Huynh
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan
- Department of Public Health, Nutrition and Food Safety, Lien Chieu Hospital, Danang 551000, Vietnam
| | - Shih-Yi Huang
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
16
|
Shaito A, Al-Mansoob M, Ahmad SM, Haider MZ, Eid AH, Posadino AM, Pintus G, Giordo R. Resveratrol-Mediated Regulation of Mitochondria Biogenesis-associated Pathways in Neurodegenerative Diseases: Molecular Insights and Potential Therapeutic Applications. Curr Neuropharmacol 2023; 21:1184-1201. [PMID: 36237161 PMCID: PMC10286596 DOI: 10.2174/1570159x20666221012122855] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/22/2022] [Accepted: 10/09/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative disorders include different neurological conditions that affect nerve cells, causing the progressive loss of their functions and ultimately leading to loss of mobility, coordination, and mental functioning. The molecular mechanisms underpinning neurodegenerative disease pathogenesis are still unclear. Nonetheless, there is experimental evidence to demonstrate that the perturbation of mitochondrial function and dynamics play an essential role. In this context, mitochondrial biogenesis, the growth, and division of preexisting mitochondria, by controlling mitochondria number, plays a vital role in maintaining proper mitochondrial mass and function, thus ensuring efficient synaptic activity and brain function. Mitochondrial biogenesis is tightly associated with the control of cell division and variations in energy demand in response to extracellular stimuli; therefore, it may represent a promising therapeutic target for developing new curative approaches to prevent or counteract neurodegenerative disorders. Accordingly, several inducers of mitochondrial biogenesis have been proposed as pharmacological targets for treating diverse central nervous system conditions. The naturally occurring polyphenol resveratrol has been shown to promote mitochondrial biogenesis in various tissues, including the nervous tissue, and an ever-growing number of studies highlight its neurotherapeutic potential. Besides preventing cognitive impairment and neurodegeneration through its antioxidant and anti-inflammatory properties, resveratrol has been shown to be able to enhance mitochondria biogenesis by acting on its main effectors, including PGC-1α, SIRT1, AMPK, ERRs, TERT, TFAM, NRF-1 and NRF-2. This review aims to present and discuss the current findings concerning the impact of resveratrol on the machinery and main effectors modulating mitochondrial biogenesis in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Abdullah Shaito
- Biomedical Research Center, College of Medicine, Qatar University, Doha, 2713, Qatar
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, 2713, Qatar
| | - Maryam Al-Mansoob
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar
| | - Salma M.S. Ahmad
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, 2713, Qatar
| | | | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, 2713, Qatar
| | - Anna Maria Posadino
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, 505055, United Arab Emirates
| |
Collapse
|
17
|
Zheng Y, Shi Y, Yang X, Gao J, Nie Z, Xu G. Effects of resveratrol on lipid metabolism in liver of red tilapia Oreochromis niloticus. Comp Biochem Physiol C Toxicol Pharmacol 2022; 261:109408. [PMID: 35820615 DOI: 10.1016/j.cbpc.2022.109408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022]
Abstract
Resveratrol (RES), as a polyphenol natural plant extract, mainly accumulates in the root of Polygonum cuspidatum, which can alleviate liver injury in mammals. Our study aims to explore the effects and potential mechanism of RES on lipid metabolism of red tilapia, and the effects of RES on liver structure, fat synthesis and metabolism of red tilapia were determined. The present study designed four groups named as 8 % fat (8%CK), 10 % fat (10 % HF), 10 % HF + RES and 10 % HF + RES + EX527 (selisistat). The liver tissues of red tilapia were collected at 3 (3 W), 6 (6 W) and 9 (9 W) weeks for parameter determination. Compared to the normal diet group, the hepatocyte of tilapia showed nuclear shift and vacuoles of different sizes when fed a high-fat diet. Meanwhile, the high-fat diet increased the contents of LDL, TC and TG significantly at 6 W, and significantly decreased the content of NAD+ at 9 W. Compared to the high-fat group, the nuclei of tilapia fed with RES were increased and visible, the degree of steatosis and the number of vacuoles were both reduced. At 3/6/9 W, RES significantly decreased the contents of LDL, TG and TMAO, and significantly increased the content of NAD+. A total of 1416 genes were up-regulated and 1928 genes were down-regulated in the group with added RES when compared to the 10 % HF group. The pathways related to lipid metabolism including PPAR signaling pathway have been enriched. Interestingly, the expressions of sirt1, pparα, fabp7 and cpt1b genes were up-regulated in RES diet group, while the expressions of pparγ, me1, scd and lpl genes were down-regulated. After the addition of an inhibitor (EX527), the above indexes showed an opposite trend when compared to the group with added RES. The overall results showed that the high-fat diet could cause fatty liver lesions in the liver of red tilapia, and RES could activate the sirt1 gene, regulate the PPARα/γ pathway and related genes, and thus regulate liver fat synthesis and metabolism leading to the alleviation of damage to liver tissue.
Collapse
Affiliation(s)
- Yao Zheng
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu 214081, China
| | - Yulu Shi
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China
| | - Xiaoxi Yang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu 214081, China
| | - Jiancao Gao
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China
| | - Zhijuan Nie
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China
| | - Gangchun Xu
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center (FFRC), Chinese Academy of Fishery Sciences (CAFS), Wuxi, Jiangsu 214081, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu 214081, China.
| |
Collapse
|
18
|
Takagi H, Tamura I, Fujimura T, Doi-Tanaka Y, Shirafuta Y, Mihara Y, Maekawa R, Taketani T, Sato S, Tamura H, Sugino N. Transcriptional coactivator PGC-1α contributes to decidualization by forming a histone-modifying complex with C/EBPβ and p300. J Biol Chem 2022; 298:101874. [PMID: 35358514 PMCID: PMC9048111 DOI: 10.1016/j.jbc.2022.101874] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/17/2022] Open
Abstract
We previously reported that CCAAT/enhancer-binding protein beta (C/EBPβ) is the pioneer factor inducing transcription enhancer mark H3K27 acetylation (H3K27ac) in the promoter and enhancer regions of genes encoding insulin-like growth factor–binding protein-1 (IGFBP-1) and prolactin (PRL) and that this contributes to decidualization of human endometrial stromal cells (ESCs). Peroxisome proliferator–activated receptor gamma coactivator 1-alpha (PGC-1α; PPARGC1A) is a transcriptional coactivator known to regulate H3K27ac. However, although PGC-1α is expressed in ESCs, the potential role of PGC-1α in mediating decidualization is unclear. Here, we investigated the involvement of PGC-1α in the regulation of decidualization. We incubated ESCs with cAMP to induce decidualization and knocked down PPARGC1A to inhibit cAMP-induced expression of IGFBP-1 and PRL. We found cAMP increased the recruitment of PGC-1α and p300 to C/EBPβ-binding sites in the promoter and enhancer regions of IGFBP-1 and PRL, corresponding with increases in H3K27ac. Moreover, PGC-1α knockdown inhibited these increases, suggesting PGC-1α forms a histone-modifying complex with C/EBPβ and p300 at these regions. To further investigate the regulation of PGC-1α, we focused on C/EBPβ upstream of PGC-1α. We found cAMP increased C/EBPβ recruitment to the novel enhancer regions of PPARGC1A. Deletion of these enhancers decreased PGC-1α expression, indicating that C/EBPβ upregulates PGC-1α expression by binding to novel enhancer regions. In conclusion, PGC-1α is upregulated by C/EBPβ recruitment to novel enhancers and contributes to decidualization by forming a histone-modifying complex with C/EBPβ and p300, thereby inducing epigenomic changes in the promoters and enhancers of IGFBP-1 and PRL.
Collapse
Affiliation(s)
- Haruka Takagi
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Isao Tamura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan.
| | - Taishi Fujimura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yumiko Doi-Tanaka
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yuichiro Shirafuta
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yumiko Mihara
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Ryo Maekawa
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Toshiaki Taketani
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shun Sato
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hiroshi Tamura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Norihiro Sugino
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|
19
|
Melatonin Signaling Pathways Implicated in Metabolic Processes in Human Granulosa Cells (KGN). Int J Mol Sci 2022; 23:ijms23062988. [PMID: 35328408 PMCID: PMC8950389 DOI: 10.3390/ijms23062988] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 02/01/2023] Open
Abstract
Female reproduction depends on the metabolic status, especially during the period of folliculogenesis. Even though it is believed that melatonin can improve oocyte competence, there is still limited knowledge of how it can modulate metabolic processes during folliculogenesis and which signaling pathways are involved in regulating gene expression. To investigate the effects of melatonin on metabolic signals during the antral stage of follicular development, human granulosa-like tumor cells (KGN) were treated with melatonin or forskolin, and gene expression was analyzed with RNA-seq technology. Following appropriate normalization and the application of a fold change cut-off of 1.5 (FC 1.5, p ≤ 0.05), 1009 and 922 genes were identified as differentially expressed in response to melatonin and forskolin, respectively. Analysis of major upstream regulators suggested that melatonin may activate PKB/mTOR signaling pathways to program the metabolism of KGN cells to support slower growth and differentiation and to prevent follicular atresia. Similarly, PKA activation through stimulation of cAMP synthesis with FSK seemed to exert the same effects as melatonin in reducing follicular growth and regulating differentiation. This study suggests that melatonin may act through PKA and PKB simultaneously in human granulosa cells to prevent follicular atresia and early luteinization at the antral stage.
Collapse
|
20
|
Wan B, Huang L, Jing C, Li Y, Jiao N, Liang M, Jiang S, Yang W. Zearalenone promotes follicle development through activating the SIRT1/PGC-1α signaling pathway in the ovaries of weaned gilts. J Anim Sci 2022; 100:6537148. [PMID: 35213700 PMCID: PMC9030242 DOI: 10.1093/jas/skac058] [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: 01/07/2022] [Accepted: 02/24/2022] [Indexed: 11/12/2022] Open
Abstract
This study aimed to investigate the effect of zearalenone (ZEA) exposure on follicular development in weaned gilts, and its mechanism based on the silent information regulator 1 (SIRT1)/peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α) signaling pathway. A total of 32 healthy female weaned piglets (Landrace × Yorkshire × Duroc) with an average body weight of 12.39 ± 0.24 kg were randomly allotted to a basal diet supplemented with 0, 0.15, 1.5, or 3.0 mg/kg ZEA for a 32-d feeding test. Blood and ovarian samples were obtained at the end of the experiment to determine serum toxin concentrations, ovarian histology, and the expressions of proliferating cell nuclear antigen (PCNA) and SIRT1/PGC-1α signaling pathway-related genes. Results showed that the vulva area, serum concentrations of ZEA, α-zearalenol and β-zearalenol, the thickness of the growing follicular layer, and the diameter of the largest growing follicles, as well as the expressions of SIRT1, PGC-1α, estrogen-related receptor α (ERRα), ATP synthase subunit beta (ATP5B), and PCNA, increased linearly (P < 0.05) with increasing dietary ZEA, whereas the thickness of the primordial follicle layer decreased linearly (P < 0.05). Immunohistochemical analysis showed that the immunoreactive substances of SIRT1 and PGC-1α in the ovaries enhanced with the increasing dietary ZEA (P < 0.05). In addition, the thickness of the growing follicular layer and the diameter of the largest growing follicle were positively correlated with relative mRNA and protein expressions of SIRT1, PGC-1α, ERRα, ATP5B, and PCNA (P < 0.05). However, the thickness of the primordial follicle layer was negatively correlated with the mRNA and protein expression of SIRT1, PGC-1α, ERRα, ATP5B, and PCNA (P < 0.05). Interestingly, the 1.5 mg/kg ZEA treatment had highly hyperplastic follicles, whereas 3.0 mg/kg ZEA resulted in a large number of follicular atresia, which indicated that low-dose ZEA exposure accelerated follicular proliferation, while high-dose ZEA promoted follicular atresia, although the critical value interval needs further confirmation. Results provide a theoretical basis for finding the therapeutic target of ZEA-induced reproductive disorders in weaned gilts.
Collapse
Affiliation(s)
- Boyang Wan
- Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Libo Huang
- Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Changwei Jing
- Technical Department, Shandong Chinwhiz Co., Weifang, Shandong 262400, China
| | - Yang Li
- Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Ning Jiao
- Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Ming Liang
- Department of Feeding Microecology, Shandong Baolaililai Bioengineering Co., Ltd., Taian, Shandong 271001, China
| | - Shuzhen Jiang
- Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Weiren Yang
- Department of Animal Sciences and Technology and Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, Shandong 271018, China,Corresponding author:
| |
Collapse
|
21
|
Nishigaki A, Tsubokura H, Ishida M, Hashimoto Y, Yoshida A, Hisamatsu Y, Tsuzuki‐Nakao T, Murata H, Okada H. NDRG1 is expressed in human granulosa cells: An implicative role of NDRG1 in the ovary. Reprod Med Biol 2022; 21:e12437. [PMID: 35386369 PMCID: PMC8967294 DOI: 10.1002/rmb2.12437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 11/08/2022] Open
Abstract
Purpose N-myc downstream-regulated gene 1 (NDRG1) is expressed in various human tissues and plays a role in regulating cellular proliferation, angiogenesis, and hypoxia sensing. However, the role of NDRG1 in the ovary remains poorly understood. Therefore, we investigated NDRG1 expression and the role of NDRG1 in the human ovary. Methods Follicular fluid (FF) and luteinized granulosa cells were collected from follicles during oocyte retrieval. KGN cells were cultured with cobalt chloride (CoCl2, a hypoxia-mimicking agent) and/or echinomycin. mRNA, protein levels and secretion, and localization were assessed by real-time PCR, Western blotting, ELISA, and immunohistochemical analysis, respectively. KGN cells were also transfected with NDRG1 siRNA for 72 h. Results NDRG1 protein was expressed in luteinized granulosa cells. NDRG1 concentration was positively correlated with vascular endothelial growth factor (VEGF) and progesterone concentrations in FF. CoCl2-induced hypoxic stress significantly increased NDRG1 and VEGF mRNA and protein and hypoxia-inducible factor-1α expression compared with those in the controls. The CoCl2-induced overexpression of NDRG1 and VEGF was suppressed by echinomycin. Transfection with NDRG1 siRNA significantly suppressed the release of progesterone in the culture medium. Conclusions These results indicate that ovarian NDRG1 may play important roles in follicular development, especially in the early luteinization of pre-ovulatory follicles.
Collapse
Affiliation(s)
- Akemi Nishigaki
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Hiroaki Tsubokura
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Mitsuaki Ishida
- Department of Pathology and Laboratory MedicineKansai Medical UniversityOsakaJapan
| | - Yoshiko Hashimoto
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Aya Yoshida
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Yoji Hisamatsu
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | | | - Hiromi Murata
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Hidetaka Okada
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| |
Collapse
|
22
|
Nishigaki A, Tsubokura H, Tsuzuki-Nakao T, Okada H. Hypoxia: Role of SIRT1 and the protective effect of resveratrol in ovarian function. Reprod Med Biol 2021; 21:e12428. [PMID: 34934403 PMCID: PMC8656197 DOI: 10.1002/rmb2.12428] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/09/2021] [Indexed: 12/27/2022] Open
Abstract
Background Ovarian function is closely related to the degree of vascular network development surrounding the ovary. Maternal aging‐related construction defects in this vascular network can cause ovarian hypoxia, which impedes oocyte nutrient supply, leading to physiological changes in the ovaries and oocytes. The anti‐aging gene Sirtuin 1 (SIRT1) senses and adapts to ambient stress and is associated with hypoxic environments and mitochondrial biogenesis. Methods The present study is a literature review focusing on investigations involving the changes in SIRT1 and mitochondrial expression during hypoxia and the cytoprotective effects of the SIRT1 activator, resveratrol. Main findings Hypoxia suppresses SIRT1 and mitochondrial expression. Resveratrol can reverse the hypoxia‐induced decrease in mitochondrial and SIRT1 activity. Resveratrol suppresses the production of hypoxia‐inducible factor‐1α and vascular endothelial growth factor proteins. Conclusion Resveratrol exhibits protective activity against hypoxic stress and may prevent hypoxia‐ or aging‐related mitochondrial dysfunction. Resveratrol treatment may be a potential option for infertility therapy.
Collapse
Affiliation(s)
- Akemi Nishigaki
- Department of Obstetrics and Gynecology Kansai Medical University Osaka Japan
| | - Hiroaki Tsubokura
- Department of Obstetrics and Gynecology Kansai Medical University Osaka Japan
| | | | - Hidetaka Okada
- Department of Obstetrics and Gynecology Kansai Medical University Osaka Japan
| |
Collapse
|
23
|
Qin J, Guo S, Yang J, Qazi IH, Pan B, Lv T, Zang S, Fang Y, Zhou G. Melatonin Promotes in vitro Development of Vitrified-Warmed Mouse GV Oocytes, Potentially by Modulating Phosphorylation of Drp1. Front Vet Sci 2021; 8:752001. [PMID: 34631868 PMCID: PMC8497800 DOI: 10.3389/fvets.2021.752001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/30/2021] [Indexed: 12/03/2022] Open
Abstract
Previous studies have shown that melatonin can mitigate cryopreservation-induced mitochondrial dysfunction in oocytes; however, the underlying molecular mechanism remains unclear. The objective of the present study was to investigate whether melatonin can improve the mitochondrial function during in vitro maturation of vitrified-warmed mouse germinal vesicle (GV) oocytes by modulating phosphorylation of dynamin related protein 1 (Drp1). Vitrification/warming procedures resulted in the following: (1) After cryopreservation of mouse GV oocytes, the phosphorylation level of Drp1 at Ser616 (p-Drp1 Ser616) in metaphase II (MII) oocytes was increased (P < 0.05). Furthermore, the rates of in vitro maturation, cleavage and blastocyst formation after parthenogenetic activation were decreased (P < 0.05). (2) In MII oocytes, the expression levels of translocase of the mitochondrial outer membrane 20 (TOMM20), mitochondrial membrane potential (MMP), adenosine triphosphate (ATP) content, and mRNA levels of mitochondrial biogenesis-related genes (Sirt1, Pgc-1α, Tfam) were all decreased (P < 0.05), and (3) Reactive oxygen species (ROS) level, early apoptosis level, Cytochrome C release and mRNA levels of pro-apoptotic related genes (Bax, Caspase9, Caspase3) in MII oocytes were all increased (P < 0.05). The results of this study further revealed that negative impacts of GV oocyte cryopreservation were mitigated by supplementation of warming and in vitro maturation media with 10−7mol /L melatonin or 2 x 10−5mol/L Mdivi-1 (Drp1 inhibitor). Therefore, we concluded that 10−7mol/L melatonin improved mitochondrial function, reduced oxidative stress and inhibited apoptosis by regulating phosphorylation of Drp1, thereby enhancing in vitro development of vitrified-warmed mouse GV oocytes.
Collapse
Affiliation(s)
- Jianpeng Qin
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shichao Guo
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jinyu Yang
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Izhar Hyder Qazi
- Department of Veterinary Anatomy and Histology, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand, Pakistan
| | - Bo Pan
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Tianyi Lv
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shengqin Zang
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yi Fang
- Department of Grassland Resources and Animal Husbandry, Jilin Provincial Key Laboratory of Grassland Farming, Northeast Institute of Geography and Agoecology, Chinese Academy of Sciences, Changchun, China
| | - Guangbin Zhou
- Department of Animal Science, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
24
|
Wang H, Su X, Zhang QQ, Zhang YY, Chu ZY, Zhang JL, Ren Q. MicroRNA-93-5p participates in type 2 diabetic retinopathy through targeting Sirt1. Int Ophthalmol 2021; 41:3837-3848. [PMID: 34313929 DOI: 10.1007/s10792-021-01953-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 07/09/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate the role of miR-93-5p in rats with type 2 diabetic retinopathy (DR) through targeting Sirt1. METHODS The targeting correlation between miR-93-5p and Sirt1 was validated by dual-luciferase reporter gene assay. Type 2 diabetes mellitus (T2DM) rat models were received intravitreal injection of antagomir NC (negative control), miR-93-5p antagomir, miR-93-5p agomir and/or recombinant Sirt1, followed by observation of pathological changes in retina via HE staining. Besides, retinal vascular permeability was determined by fluorescein isothiocyanate-bovine serum albumin (FITC-BSA), while the retinal vasculature was observed through retinal trypsin digestion. Expression of miR-93-5p and Sirt1 was measured by qRT-PCR and Western blotting, while the levels of VEGF, proinflammatory cytokines and anti-oxidative indicators were determined using corresponding kits. RESULTS MiR-93-5p could target Sirt1 as analyzed by the luciferase reporter gene assay. Rats in the T2DM group presented the up-regulation of miR-93-5p and down-regulation of Sirt1 in the retina, and miR-93-5p inhibition could up-regulate Sirt1 expression in the T2DM rats. Recombinant Sirt1 decreased retinal vascular permeability and acellular capillaries with improved pathological changes in retina from T2DM rats, which was abolished by miR-93-5p agomir. Moreover, miR-93-5p inhibition or Sirt1 overexpression decreased the levels of VEGF and proinflammatory cytokines while enhancing the activity of anti-oxidative indicators. However, indicators above had no significant differences between T2DM group and T2DM + agomir + Sirt1 group. CONCLUSION MiR-93-5p, via targeting Sirt1, could affect the vascular permeability and acellular capillaries and mitigate the inflammation and oxidative stress in the retinas, which may play a critical role in DR.
Collapse
Affiliation(s)
- Hui Wang
- Department of Ophthalmology, Shijiazhuang People's Hospital, No. 365, Jianhua South Street, Yuhua District, Shijiazhuang, 050030, Hebei Province, China
| | - Xian Su
- Department of Ophthalmology, Shijiazhuang People's Hospital, No. 365, Jianhua South Street, Yuhua District, Shijiazhuang, 050030, Hebei Province, China
| | - Qian-Qian Zhang
- Department of Ophthalmology, Shijiazhuang People's Hospital, No. 365, Jianhua South Street, Yuhua District, Shijiazhuang, 050030, Hebei Province, China
| | - Ying-Ying Zhang
- Department of Ophthalmology, Shijiazhuang People's Hospital, No. 365, Jianhua South Street, Yuhua District, Shijiazhuang, 050030, Hebei Province, China
| | - Zhan-Ya Chu
- Department of Ophthalmology, Shijiazhuang People's Hospital, No. 365, Jianhua South Street, Yuhua District, Shijiazhuang, 050030, Hebei Province, China
| | - Jin-Ling Zhang
- Department of Ophthalmology, Shijiazhuang People's Hospital, No. 365, Jianhua South Street, Yuhua District, Shijiazhuang, 050030, Hebei Province, China
| | - Qian Ren
- Department of Ophthalmology, Shijiazhuang People's Hospital, No. 365, Jianhua South Street, Yuhua District, Shijiazhuang, 050030, Hebei Province, China.
| |
Collapse
|
25
|
Xu R, Luo X, Ye X, Li H, Liu H, Du Q, Zhai Q. SIRT1/PGC-1α/PPAR-γ Correlate With Hypoxia-Induced Chemoresistance in Non-Small Cell Lung Cancer. Front Oncol 2021; 11:682762. [PMID: 34381712 PMCID: PMC8351465 DOI: 10.3389/fonc.2021.682762] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
Resistance is the major cause of treatment failure and disease progression in non-small cell lung cancer (NSCLC). There is evidence that hypoxia is a key microenvironmental stress associated with resistance to cisplatin, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), and immunotherapy in solid NSCLCs. Numerous studies have contributed to delineating the mechanisms underlying drug resistance in NSCLC; nevertheless, the mechanisms involved in the resistance associated with hypoxia-induced molecular metabolic adaptations in the microenvironment of NSCLC remain unclear. Studies have highlighted the importance of posttranslational regulation of molecular mediators in the control of mitochondrial function in response to hypoxia-induced metabolic adaptations. Hypoxia can upregulate the expression of sirtuin 1 (SIRT1) in a hypoxia-inducible factor (HIF)-dependent manner. SIRT1 is a stress-dependent metabolic sensor that can deacetylate some key transcriptional factors in both metabolism dependent and independent metabolic pathways such as HIF-1α, peroxisome proliferator-activated receptor gamma (PPAR-γ), and PPAR-gamma coactivator 1-alpha (PGC-1α) to affect mitochondrial function and biogenesis, which has a role in hypoxia-induced chemoresistance in NSCLC. Moreover, SIRT1 and HIF-1α can regulate both innate and adaptive immune responses through metabolism-dependent and -independent ways. The objective of this review is to delineate a possible SIRT1/PGC-1α/PPAR-γ signaling-related molecular metabolic mechanism underlying hypoxia-induced chemotherapy resistance in the NSCLC microenvironment. Targeting hypoxia-related metabolic adaptation may be an attractive therapeutic strategy for overcoming chemoresistance in NSCLC.
Collapse
Affiliation(s)
- Rui Xu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, China
| | - Xin Luo
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuan Ye
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huan Li
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyue Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiong Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Zhai
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, China.,Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Cacciottola L, Donnez J, Dolmans MM. Ovarian tissue damage after grafting: systematic review of strategies to improve follicle outcomes. Reprod Biomed Online 2021; 43:351-369. [PMID: 34384692 DOI: 10.1016/j.rbmo.2021.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/14/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022]
Abstract
Frozen-thawed human ovarian tissue endures large-scale follicle loss in the early post-grafting period, characterized by hypoxia lasting around 7 days. Tissue revascularization occurs progressively through new vessel invasion from the host and neoangiogenesis from the graft. Such reoxygenation kinetics lead to further potential damage caused by oxidative stress. The aim of the present manuscript is to provide a systematic review of proangiogenic growth factors, hormones and various antioxidants administered in the event of ovarian tissue transplantation to protect the follicle pool from depletion by boosting revascularization or decreasing oxidative stress. Although almost all investigated studies revealed an advantage in terms of revascularization and reduction in oxidative stress, far fewer demonstrated a positive impact on follicle survival. As the cascade of events driven by ischaemia after transplantation is a complex process involving numerous players, it appears that acting on specific molecular mechanisms, such as concentrations of proangiogenic growth factors, is not enough to significantly mitigate tissue damage. Strategies exploiting the activated tissue response to ischaemia for tissue healing and remodelling purposes, such as the use of antiapoptotic drugs and adult stem cells, are also discussed in the present review, since they yielded promising results in terms of follicle pool protection.
Collapse
Affiliation(s)
- Luciana Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques Donnez
- Prof. Emeritus, Université Catholique de Louvain, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Department of Gynecology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
27
|
Ragonese F, Monarca L, De Luca A, Mancinelli L, Mariani M, Corbucci C, Gerli S, Iannitti RG, Leonardi L, Fioretti B. Resveratrol depolarizes the membrane potential in human granulosa cells and promotes mitochondrial biogenesis. Fertil Steril 2021; 115:1063-1073. [PMID: 33487442 DOI: 10.1016/j.fertnstert.2020.08.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To study the biological effects of resveratrol on the growth, electrophysiology, and mitochondrial function of human granulosa cells (h-GCs). DESIGN Preclinical study. SETTING Electrophysiology laboratory and in vitro fertilization unit. PATIENT(S) This study included h-GCs from seven infertile women undergoing assisted reproductive techniques. INTERVENTION(S) Human ovarian Granulosa Cell Tumor (GCT) cell line COV434 and h-GCs obtained after oocyte retrieval were cultured in the absence or presence of resveratrol. MAIN OUTCOME MEASURE(S) Granulosa cells were evaluated for cell viability and mitochondrial activity. Electrophysiological recordings and evaluation of potassium current (IKur) and Ca2+ concentration were also performed. RESULT(S) Resveratrol induced mitochondrial activity in a bell-shaped, dose-effect-dependent manner. Specifically, resveratrol treatment (3 μM, 48 hours) increased ATP production and cell viability and promoted the induction of cellular differentiation. These biological changes were associated with mitochondrial biogenesis. Electrophysiological recordings showed that resveratrol reduced the functional expression of an ultra rapid activating, slow inactivating, delayed rectifier potassium current (IKur) that is associated with a plasma membrane depolarization and that promotes an increase in intracellular Ca2+. CONCLUSION(S): The effects of resveratrol on potassium current and mitochondrial biogenesis in h-GCs could explain the beneficial effects of this polyphenol on the physiology of the female reproductive system. These findings suggest there are therapeutic implications of resveratrol in a clinical setting.
Collapse
Affiliation(s)
- Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy; Department of Experimental Medicine, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Antonella De Luca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Loretta Mancinelli
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Monica Mariani
- Centre of Assisted Reproductive Technologies, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Cristina Corbucci
- Centre of Assisted Reproductive Technologies, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Sandro Gerli
- Department of Surgical and Biomedical Sciences, Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | | | | | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy.
| |
Collapse
|
28
|
Bordoni L, Gabbianelli R. Mitochondrial DNA and Neurodegeneration: Any Role for Dietary Antioxidants? Antioxidants (Basel) 2020; 9:E764. [PMID: 32824558 PMCID: PMC7466149 DOI: 10.3390/antiox9080764] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
The maintenance of the mitochondrial function is essential in preventing and counteracting neurodegeneration. In particular, mitochondria of neuronal cells play a pivotal role in sustaining the high energetic metabolism of these cells and are especially prone to oxidative damage. Since overproduction of reactive oxygen species (ROS) is involved in the pathogenesis of neurodegeneration, dietary antioxidants have been suggested to counteract the detrimental effects of ROS and to preserve the mitochondrial function, thus slowing the progression and limiting the extent of neuronal cell loss in neurodegenerative disorders. In addition to their role in the redox-system homeostasis, mitochondria are unique organelles in that they contain their own genome (mtDNA), which acts at the interface between environmental exposures and the molecular triggers of neurodegeneration. Indeed, it has been demonstrated that mtDNA (including both genetics and, from recent evidence, epigenetics) might play relevant roles in modulating the risk for neurodegenerative disorders. This mini-review describes the link between the mitochondrial genome and cellular oxidative status, with a particular focus on neurodegeneration; moreover, it provides an overview on potential beneficial effects of antioxidants in preserving mitochondrial functions through the protection of mtDNA.
Collapse
Affiliation(s)
- Laura Bordoni
- Unit of Molecular Biology, School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | | |
Collapse
|