1
|
Nguyen CLK, Kuba Y, Le HT, Shawki HH, Mikami N, Aoki M, Yasuhara N, Suzuki H, Mizuno-Iijima S, Ayabe S, Osawa Y, Fujiyama T, Dinh TTH, Ishida M, Daitoku Y, Tanimoto Y, Murata K, Kang W, Ema M, Hirao Y, Ogura A, Takahashi S, Sugiyama F, Mizuno S. Exocyst complex component 1 (Exoc1) loss in dormant oocyte disrupts c-KIT and growth differentiation factor (GDF9) subcellular localization and causes female infertility in mice. Cell Death Discov 2025; 11:17. [PMID: 39833146 PMCID: PMC11747099 DOI: 10.1038/s41420-025-02291-5] [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: 09/18/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
A limited number of female germ cells support reproduction in many mammals. The follicle, composed of oocytes and supporting granulosa cells, forms the basis of oogenesis. Crosstalk between oocytes and granulosa cells is essential for the formation, dormancy, re-awakening, and maturation of oocytes. The oocyte expresses c-KIT and growth differentiation factor-9 (GDF-9), which are major factors in this crosstalk. The downstream signalling pathways of c-KIT and GDF-9 have been well-documented; however, their intra-oocyte trafficking pathway remains unclear. Our study reveals that the exocyst complex, a heterotetrameric protein complex important for tethering in vesicular transport, is important for proper intra-oocyte trafficking of c-KIT and GDF9 in mice. We found that depletion of oocyte-specific EXOC1, a component of the exocyst complex, impaired oocyte re-awakening and cyst breakdown, and inhibited granulosa cell proliferation during follicle growth. The c-KIT receptor is localised on the oocyte plasma membrane. The oocyte-specific Kit conditional knockout mice were reported to exhibit impaired oocyte re-awakening and reduced oocyte cyst breakdown. GDF9 is a protein secreted extracellularly in the oocyte. Previous studies have shown that Gdf9 knockout mice impaired proliferation and granulosa cell multilayering in growing follicles. We found that both c-KIT and GDF9 abnormally stuck in the EXOC1-depleted oocyte cytoplasm. These abnormal phenotypes were also observed in oocytes depleted of exocyst complex members EXOC3 and EXOC7. These results clearly show that the exocyst complex is essential for proper intra-oocyte trafficking of c-KIT and GDF9. Inhibition of this complex causes complete loss of female fertility in mice. Our findings build a platform for research related to trafficking mechanisms of vital crosstalk factors for oogenesis.
Collapse
Affiliation(s)
- Chi Lieu Kim Nguyen
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yumeno Kuba
- Master's Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hoai Thu Le
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hossam Hassan Shawki
- Department of Comparative and Experimental Medicine, Nagoya City University Graduate School of Medical Sciences, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Natsuki Mikami
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
- Research Fellow of the Japan Society for the Promotion of Science, Kojimachi Business Center Building, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Madoka Aoki
- College of Medical Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Nanako Yasuhara
- College of Biological Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Hayate Suzuki
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Saori Mizuno-Iijima
- Experimental Animal Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Shinya Ayabe
- Experimental Animal Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Yuki Osawa
- Master's Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Tomoyuki Fujiyama
- International Institute for Integrative Sleep Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Tra Thi Huong Dinh
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
- Next Generation Human Disease Model Team, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Miyuki Ishida
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yoko Daitoku
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Yoko Tanimoto
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kazuya Murata
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Woojin Kang
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Yuji Hirao
- Division of Dairy Cattle Feeding and Breeding Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, 2 Ikenodai, Tsukuba, Ibaraki, 305-0901, Japan
| | - Atsuo Ogura
- Bioresource Engineering Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Trans-Border Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
2
|
Ru Y, Deng X, Chen J, Zhang L, Xu Z, Lv Q, Long S, Huang Z, Kong M, Guo J, Jiang M. Maternal age enhances purifying selection on pathogenic mutations in complex I genes of mammalian mtDNA. NATURE AGING 2024; 4:1211-1230. [PMID: 39075271 DOI: 10.1038/s43587-024-00672-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/14/2024] [Indexed: 07/31/2024]
Abstract
Mitochondrial diseases, caused mainly by pathogenic mitochondrial DNA (mtDNA) mutations, pose major challenges due to the lack of effective treatments. Investigating the patterns of maternal transmission of mitochondrial diseases could pave the way for preventive approaches. In this study, we used DddA-derived cytosine base editors (DdCBEs) to generate two mouse models, each haboring a single pathogenic mutation in complex I genes (ND1 and ND5), replicating those found in human patients. Our findings revealed that both mutations are under strong purifying selection during maternal transmission and occur predominantly during postnatal oocyte maturation, with increased protein synthesis playing a vital role. Interestingly, we discovered that maternal age intensifies the purifying selection, suggesting that older maternal age may offer a protective effect against the transmission of deleterious mtDNA mutations, contradicting the conventional notion that maternal age correlates with increased transmitted mtDNA mutations. As collecting comprehensive clinical data is needed to understand the relationship between maternal age and transmission patterns in humans, our findings may have profound implications for reproductive counseling of mitochondrial diseases, especially those involving complex I gene mutations.
Collapse
Affiliation(s)
- Yanfei Ru
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
| | - Xiaoling Deng
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
- Fudan University, Shanghai, China
| | - Jiatong Chen
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Leping Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Zhe Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
| | - Qunyu Lv
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Shiyun Long
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Zijian Huang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
- Fudan University, Shanghai, China
| | - Minghua Kong
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Jing Guo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Min Jiang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences,Westlake University, Hangzhou, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China.
| |
Collapse
|
3
|
Altassan R, AlQudairy H, AlJebreen S, AlMuhaizea M, Al-Hindi H, Pena-Guerra KA, Ghebeh H, Almzroua A, Albakheet A, AlDosary M, Colak D, Arold ST, Kaya N. Expanding the phenotypic and genotypic spectrum of GGPS1 related congenital muscular dystrophy. Am J Med Genet A 2024; 194:e63498. [PMID: 38129970 DOI: 10.1002/ajmg.a.63498] [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: 08/07/2023] [Revised: 10/24/2023] [Accepted: 11/26/2023] [Indexed: 12/23/2023]
Abstract
Congenital muscular dystrophies are a group of progressive disorders with wide range of symptoms associated with diverse cellular mechanisms. Recently, biallelic variants in GGPS1 were linked to a distinct autosomal recessive form of muscular dystrophy associated with hearing loss and ovarian insufficiency. In this report, we present a case of a young patient with a homozygous variant in GGPS1. The patient presented with only proximal muscle weakness, and elevated liver transaminases with spared hearing function. The hepatic involvement in this patient caused by a novel deleterious variant in the gene extends the phenotypic and genotypic spectrum of GGPS1 related muscular dystrophy.
Collapse
Affiliation(s)
- Ruqaiah Altassan
- Department of Medical Genomics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Hanan AlQudairy
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Sarah AlJebreen
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Mohammed AlMuhaizea
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
- Center for Neurosciences, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Hindi Al-Hindi
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Karla A Pena-Guerra
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal, Kingdom of Saudi Arabia
| | - Hazem Ghebeh
- Stem Cell and Tissue Re-Engineering Program Department, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Amer Almzroua
- Stem Cell and Tissue Re-Engineering Program Department, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Albandary Albakheet
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Mazhor AlDosary
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Dilek Colak
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Division of Biological and Environmental Sciences and Engineering (BESE), Thuwal, Kingdom of Saudi Arabia
- Centre de Biochimie Structurale, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Namik Kaya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
4
|
Nandi S, Kumar B S, Gupta PSP, Mondal S, Kumar VG. Influence of phenolic flavonols (Kaempferol, Querectin and Myricetin) on the survival and growth of ovine preantral follicles and granulosa cells cultured in vitro. Theriogenology 2024; 214:266-272. [PMID: 37948816 DOI: 10.1016/j.theriogenology.2023.10.023] [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: 03/31/2023] [Revised: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Study was carried out to examine the influence of plant bioactive compounds [Kaempferol (KAE), Querectin (QUE) and Myricetin (MYR)] on the survival and growth parameters of cultured ovine preantral follicles (PFs) granulosa cells (GCs) and expression of some key developmental genes. Ovine PFs were isolated from slaughterhouse derived ovaries and KAE, QUE and MYR were supplemented to the standard culture medium of GCs and PFs at concentrations of 0, 5, 10, 25, 50 and 100 μM and cultured for 5 and 7 days respectively. PFs morphological and functional parameters [follicle and enclosed oocyte growth rate, viability of follicles, antrum formation rate, oocyte maturation rate, estradiol concentration, reactive oxygen species (ROS) production] and GC growth parameters (metabolic activity, viability rate, cell number increment, ROS production) were measured after culture. Significantly higher PF growth, viability rate and estradiol concentration was observed at 10 μM, 25 μM and 10 μM concentration of KAE, MYR and QUE respectively compared to the control. ROS production was significantly decreased in the PF culture media treated with 10 μM KAE or MYR 25 μM or 10 μM QUE compared to those observed in the control group. Likewise, metabolic activity of GCs, viability rate and cell number increment cultured with KAE, MYR and QUE was significantly higher at 10, 25 and 10 μM concentrations respectively compared to those observed in control group. ROS production was significantly lower in the GC cultured with KAE, MYR and QUE at 10, 25 and 10 μM concentrations respectively compared to the control. Based on the results of the growth parameters, gene expression of PFs and GCs were studied by qPCR at selected concentrations (KAE, MYR and QUE at 10, 25 and 10 μM concentrations respectively) in the cultured PFs and GCs. Gene expression of GDF9, FGF2, CYP19A1 was significantly higher and Bax, Bcl2 expression was significantly lower in the PFs and GCs cultured with the KAE or QUE at 10 μM concentration. KAE, MYR and QUE have dose dependant responses on PFs and GCs morphological and functional parameters; however, KAE is more potent amongst the three in augmenting the ovarian functions.
Collapse
Affiliation(s)
- S Nandi
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India.
| | - Sampath Kumar B
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India; Veterinary College, Bangalore Campus, Hebbal, Bangalore, India
| | - P S P Gupta
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India
| | - S Mondal
- ICAR-National Institute of Animal Nutrition and Physiology, Bangalore, India
| | - V Girish Kumar
- Veterinary College, Bangalore Campus, Hebbal, Bangalore, India
| |
Collapse
|
5
|
Zhang JL, Lv M, Yang CF, Zhu YX, Li CJ. Mevalonate pathway and male reproductive aging. Mol Reprod Dev 2023; 90:774-781. [PMID: 37733694 DOI: 10.1002/mrd.23705] [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: 07/17/2022] [Revised: 08/27/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Abstract
Male fertility declines with age. The mevalonate pathway, through which cholesterol and nonsteroidal isoprenoids are synthesized, plays key role in metabolic processes and is an essential pathway for cholesterol production and protein prenylation. Male reproductive aging is accompanied by dramatic changes in the metabolic microenvironment of the testis. Since the mevalonate pathway has an important role in spermatogenesis, we attempted to explore the association between male reproductive aging and the mevalonate pathway to explain the mechanism of male reproductive aging. Alterations in the mevalonate pathway may affect male reproductive aging by decreasing cholesterol synthesis and altering testis protein prenylation. Decreased cholesterol levels affect cholesterol modification, testosterone production, and remodeling of germ cell membranes. Aging-related metabolic disorders also affect the metabolic coupling between somatic cells and spermatogenic cells, leading to male fertility decline. Therefore, we hypothesized that alterations in the mevalonate pathway represent one of the metabolic causes of reproductive aging.
Collapse
Affiliation(s)
- Jia-Le Zhang
- State Key Laboratory of Reproductive Medicine and China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meng Lv
- State Key Laboratory of Reproductive Medicine and China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chao-Fan Yang
- State Key Laboratory of Reproductive Medicine and China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ying-Xi Zhu
- State Key Laboratory of Reproductive Medicine and China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chao-Jun Li
- State Key Laboratory of Reproductive Medicine and China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Sang Y, Yang Q, Guo Y, Liu X, Shen D, Jiang C, Wang X, Li K, Wang H, Yang C, Ding L, Sun H, Guo X, Li C. Oocytes orchestrate protein prenylation for mitochondrial function through selective inactivation of cholesterol biosynthesis in murine species. J Biol Chem 2023; 299:105183. [PMID: 37611828 PMCID: PMC10534227 DOI: 10.1016/j.jbc.2023.105183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 07/18/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023] Open
Abstract
Emerging research and clinical evidence suggest that the metabolic activity of oocytes may play a pivotal role in reproductive anomalies. However, the intrinsic mechanisms governing oocyte development regulated by metabolic enzymes remain largely unknown. Our investigation demonstrates that geranylgeranyl diphosphate synthase1 (Ggps1), the crucial enzyme in the mevalonate pathway responsible for synthesizing isoprenoid metabolite geranylgeranyl pyrophosphate from farnesyl pyrophosphate, is essential for oocyte maturation in mice. Our findings reveal that the deletion of Ggps1 that prevents protein prenylation in fully grown oocytes leads to subfertility and offspring metabolic defects without affecting follicle development. Oocytes that lack Ggps1 exhibit disrupted mitochondrial homeostasis and the mitochondrial defects arising from oocytes are inherited by the fetal offspring. Mechanistically, the excessive farnesylation of mitochondrial ribosome protein, Dap3, and decreased levels of small G proteins mediate the mitochondrial dysfunction induced by Ggps1 deficiency. Additionally, a significant reduction in Ggps1 levels in oocytes is accompanied by offspring defects when females are exposed to a high-cholesterol diet. Collectively, this study establishes that mevalonate pathway-protein prenylation is vital for mitochondrial function in oocyte maturation and provides evidence that the disrupted protein prenylation resulting from an imbalance between farnesyl pyrophosphate and geranylgeranyl pyrophosphate is the major mechanism underlying impairment of oocyte quality induced by high cholesterol.
Collapse
Affiliation(s)
- Yongjuan Sang
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China
| | - Qiwen Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Yueshuai Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Xiaofei Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Di Shen
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China
| | - Chen Jiang
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China
| | - Xinying Wang
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China
| | - Kang Li
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China
| | - Haiquan Wang
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China
| | - Chaofan Yang
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China.
| | - Chaojun Li
- Modern Animal Research Center of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
7
|
Muehlebach ME, Holstein SA. Geranylgeranyl diphosphate synthase: Role in human health, disease and potential therapeutic target. Clin Transl Med 2023; 13:e1167. [PMID: 36650113 PMCID: PMC9845123 DOI: 10.1002/ctm2.1167] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023] Open
Abstract
Geranylgeranyl diphosphate synthase (GGDPS), an enzyme in the isoprenoid biosynthesis pathway, is responsible for the production of geranylgeranyl pyrophosphate (GGPP). GGPP serves as a substrate for the post-translational modification (geranylgeranylation) of proteins, including those belonging to the Ras superfamily of small GTPases. These proteins play key roles in signalling pathways, cytoskeletal regulation and intracellular transport, and in the absence of the prenylation modification, cannot properly localise and function. Aberrant expression of GGDPS has been implicated in various human pathologies, including liver disease, type 2 diabetes, pulmonary disease and malignancy. Thus, this enzyme is of particular interest from a therapeutic perspective. Here, we review the physiological function of GGDPS as well as its role in pathophysiological processes. We discuss the current GGDPS inhibitors under development and the therapeutic implications of targeting this enzyme.
Collapse
Affiliation(s)
- Molly E. Muehlebach
- Cancer Research Doctoral ProgramUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Sarah A. Holstein
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
8
|
Flavanol-Rich Cocoa Supplementation Inhibits Mitochondrial Biogenesis Triggered by Exercise. Antioxidants (Basel) 2022; 11:antiox11081522. [PMID: 36009241 PMCID: PMC9405215 DOI: 10.3390/antiox11081522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
The potential role of cocoa supplementation in an exercise context remains unclear. We describe the effects of flavanol-rich cocoa supplementation during training on exercise performance and mitochondrial biogenesis. Forty-two male endurance athletes at the beginning of the training season received either 5 g of cocoa (425 mg of flavanols) or maltodextrin (control) daily for 10 weeks. Two different doses of cocoa (equivalent to 5 g and 15 g per day of cocoa for a 70 kg person) were tested in a mouse exercise training study. In the athletes, while both groups had improved exercise performance, the maximal aerobic speed increased only in the control group. A mitochondrial DNA analysis revealed that the control group responded to training by increasing the mitochondrial load whereas the cocoa group showed no increase. Oxidative stress was lower in the cocoa group than in the control group, together with lower interleukin-6 levels. In the muscle of mice receiving cocoa, we corroborated an inhibition of mitochondrial biogenesis, which might be mediated by the decrease in the expression of nuclear factor erythroid-2-related factor 2. Our study shows that supplementation with flavanol-rich cocoa during the training period inhibits mitochondrial biogenesis adaptation through the inhibition of reactive oxygen species generation without impacting exercise performance.
Collapse
|
9
|
Pramanick R, Nathani N, Warke H, Mayadeo N, Aranha C. Vaginal Dysbiotic Microbiome in Women With No Symptoms of Genital Infections. Front Cell Infect Microbiol 2022; 11:760459. [PMID: 35096634 PMCID: PMC8790106 DOI: 10.3389/fcimb.2021.760459] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
The vaginal microbiome plays a critical role in determining the progression of female genital tract infections; however, little is known about the vaginal microbiota of Indian women. We aimed to investigate the vaginal microbial architecture of women with asymptomatic bacterial vaginosis (BV) (n=20) and normal microbiota (n=19). Microbial diversity was analyzed in vaginal swabs from regularly menstruating women (18-45yrs) by 16S rRNA V3-V4 amplicon (MiSeq Illumina) sequencing. Rarefaction analysis showed a higher number of species in normal flora compared to BV. Alpha diversity as measured by Pielou’s evenness revealed microbial diversity was significantly greater in BV samples than normal microbiota (p= 0.0165). Beta diversity comparison using UniFrac metrics indicated distinct microbial communities clustering between normal and BV flora. Firmicutes were the major phyla observed in vaginal specimens of normal microbiota whereas Actinobacteria, Fusobacteria, Bacteroidetes were significantly abundant in BV samples. Notably, the relative abundance of Lactobacillus was significantly high in normal microbiota. Conversely Gardnerella, Sneathia, Prevotella, Atopobium, Ureaplasma, Dialister significantly dominated dysbiotic microbiota. Relative frequency of Lactobacillus decreased significantly in BV (6%) as compared to normal microbiota (35.2%). L. fermentum, L. gasseri, L. iners, L. jensenii, L. mucosae, L. ruminis, L. salivarius, L. coleohominis was more exclusively present in normal microbiota. L. iners was detected from both the groups with a relative frequency of 50.4% and 17.2% in normal and BV microbiota respectively. Lefse analysis indicated Atopobium vaginae, Sneathia amnii, Mycoplasma hominis Prevotella disiens in the vaginal microbiota as a biomarker for dysbiosis and L. jensenii as a biomarker of a healthy microbiota. Firmicutes were negatively correlated to Tenericutes, Actinobacteria, Bacteroidetes, and Fusobacteria. Proteobacteria positively correlated to Tenericutes, and Bacteroidetes were shown to be positively correlated to Fusobacteria. Predicted functional analysis indicated differences in the functional profiles between BV and normal microbiota. Normal microbiota utilized pathways essential for phosphatidylglycerol biosynthesis I & II, peptidoglycan biosynthesis, geranylgeranyl diphosphate biosynthesis I, mevalonate pathway, CoA biosynthesis pathway I and pyrimidine nucleotide salvage; whereas BV bacteria had characteristic aromatic amino acid biosynthesis, pentose phosphate pathway, carbohydrate degradation. In conclusion, women with asymptomatic BV have vaginal microbiota significantly different than women with normal microbiota. Furthermore, the study provides insights into the vaginal microbial structure of Indian women that will enable us to explore the prospective candidates for restoring the vaginal microbiota.
Collapse
Affiliation(s)
- Rinku Pramanick
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive and Child Health, Mumbai, India
| | - Neelam Nathani
- School of Applied Sciences & Technology (SAST-GTU), Gujarat Technological University, Ahmedabad, India
| | - Himangi Warke
- Department of Obstetrics and Gynecology, King Edward Memorial Hospital and Seth Gordhandas Sunderdas Medical College, Mumbai, India
| | - Niranjan Mayadeo
- Department of Obstetrics and Gynecology, King Edward Memorial Hospital and Seth Gordhandas Sunderdas Medical College, Mumbai, India
| | - Clara Aranha
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive and Child Health, Mumbai, India
| |
Collapse
|
10
|
Rooda I, Kaselt B, Liivrand M, Smolander OP, Salumets A, Velthut-Meikas A. Hsa-mir-548 family expression in human reproductive tissues. BMC Genom Data 2021; 22:40. [PMID: 34625017 PMCID: PMC8501715 DOI: 10.1186/s12863-021-00997-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hsa-miR-548ba expressed in ovarian granulosa cells targets PTEN and LIFR, which are essential for ovarian follicle activation and growth. The expression pattern of hsa-miR-548ba correlates with its host gene follicle-stimulating hormone receptor (FSHR), and FSH has a positive influence on hsa-miR-548ba expression. However, hsa-miR-548ba is a member of a large hsa-mir-548 family with potentially overlapping targets. The current study aims to investigate the co-expression of hsa-mir-548 family members in FSHR-positive reproductive tissues and to explore the potential co-regulation of pathways. RESULTS For the above-described analysis, small RNA sequencing data from public data repositories were used. Sequencing results revealed that hsa-miR-548ba was expressed at the highest level in the ovarian granulosa cells and uterine myometrial samples together with another twelve and one hsa-miR-548 family members, respectively. Pathway enrichment analysis of microRNA targets in the ovarian samples revealed the hsa-miR-548ba and hsa-miR-548b-5p co-regulation of RAB geranylgeranylation in mural granulosa cells. Moreover, other hsa-mir-548 family members co-regulate pathways essential for ovarian functions (PIP3 activates AKT signalling and signalling by ERBB4). In addition to hsa-miR-548ba, hsa-miR-548o-3p is expressed in the myometrium, which separately targets the peroxisome proliferator-activated receptor alpha (PPARA) pathway. CONCLUSION This study reveals that hsa-mir-548 family members are expressed in variable combinations in the reproductive tract, where they potentially fulfil different regulatory roles. The results provide a reference for further studies of the hsa-mir-548 family role in the reproductive tract.
Collapse
Affiliation(s)
- Ilmatar Rooda
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia.
| | - Birgitta Kaselt
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Maria Liivrand
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Olli-Pekka Smolander
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 14186, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, L. Puusepa St. 8, 50406, Tartu, Estonia
- Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| |
Collapse
|
11
|
Peng SL, Wu QF, Xie Q, Tan J, Shu KY. PATL2 regulated the apoptosis of ovarian granulosa cells in patients with PCOS. Gynecol Endocrinol 2021; 37:629-634. [PMID: 34008465 DOI: 10.1080/09513590.2021.1928066] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AIM PCOS often showed abnormal follicular development. Previous studies have found that the increased apoptosis of granulosa cells (GCs) is one of the key factors leading to follicular dysplasia. It has been found that the decrease or deletion of PATL2 function can significantly inhibit the development and maturation of human oocytes. We found that PATL2 was also expressed in human ovarian GCs, suggesting that PATL2 may be involved in the regulation of related biological events in GCs. This study aims to explore the function of PATL2 on regulation of GCs apoptosis, and the potential role of PATL2 in the development of PCOS-related abnormal follicles. MATERIALS AND METHODS The follicular GCs of PCOS patients and normal ovulating female patients were collected. Moreover, human granular cell line (KGN) was used for in vitro experiments. RESULTS (1) The maturation rate and fertilization rate of oocytes in the PCOS group were significantly lower than those in the normal control group (p<0.05). (2) Flow cytometry and TUNEL staining showed that the apoptosis level of GCs in the PCOS group was significantly increased. (3) Immunofluorescence and Western Blot showed that the PATL2 expression level of GCs in the PCOS group was significantly reduced. (4) Knocking down the expression of PATL2 by siRNA significantly prevented the apoptosis of GCs. CONCLUSIONS Reduced PATL2 could resulted in the increased apoptosis level of ovarian GCs, which might be closely related to the occurrence and development of abnormal follicles in PCOS.
Collapse
Affiliation(s)
- Shao-Lan Peng
- Maternal and Child Health Hospital Affiliated to Nanchang University, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P. R. China
- Yongkang Maternity and Child Health Care Hospital, Yongkang, Zhejiang, P. R. China
| | - Qiong-Fang Wu
- Maternal and Child Health Hospital Affiliated to Nanchang University, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P. R. China
- Reproductive Medicine Center, Maternal and Child Health Hospital Affiliated to Nanchang University, Jiangxi Maternal andChild Health Hospital, Nanchang, P. R. China
| | - Qi Xie
- Maternal and Child Health Hospital Affiliated to Nanchang University, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P. R. China
- Jiangxi Medical College, Nanchang University, Nanchang, P. R. China
| | - Jun Tan
- Maternal and Child Health Hospital Affiliated to Nanchang University, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P. R. China
- Reproductive Medicine Center, Maternal and Child Health Hospital Affiliated to Nanchang University, Jiangxi Maternal andChild Health Hospital, Nanchang, P. R. China
| | - Kuan-Yong Shu
- Maternal and Child Health Hospital Affiliated to Nanchang University, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P. R. China
- Department of Gynecology, Maternal and Child Health Hospital of Nanchang University, Jiangxi Maternal and Child Health Hospital, Nanchang, P. R. China
| |
Collapse
|
12
|
Chong D, Chen Z, Guan S, Zhang T, Xu N, Zhao Y, Li C. Geranylgeranyl pyrophosphate-mediated protein geranylgeranylation regulates endothelial cell proliferation and apoptosis during vasculogenesis in mouse embryo. J Genet Genomics 2021; 48:300-311. [PMID: 34049800 DOI: 10.1016/j.jgg.2021.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 11/17/2022]
Abstract
Vascular development is essential for the establishment of the circulatory system during embryonic development and requires the proliferation of endothelial cells. However, the underpinning regulatory mechanisms are not well understood. Here, we report that geranylgeranyl pyrophosphate (GGPP), a metabolite involved in protein geranylgeranylation, plays an indispensable role in embryonic vascular development. GGPP is synthesized by geranylgeranyl pyrophosphate synthase (GGPPS) in the mevalonate pathway. The selective knockout of Ggpps in endothelial cells led to aberrant vascular development and embryonic lethality, resulting from the decreased proliferation and enhanced apoptosis of endothelial cells during vasculogenesis. The defect in protein geranylgeranylation induced by GGPP depletion inhibited the membrane localization of RhoA and enhanced yes-associated protein (YAP) phosphorylation, thereby prohibiting the entry of YAP into the nucleus and the expression of YAP target genes related to cell proliferation and the antiapoptosis process. Moreover, inhibition of the mevalonate pathway by simvastatin induced endothelial cell proliferation defects and apoptosis, which were ameliorated by GGPP. Geranylgeraniol (GGOH), a precursor of GGPP, ameliorated the harmful effects of simvastatin on vascular development of developing fetuses in pregnant mice. These results indicate that GGPP-mediated protein geranylgeranylation is essential for endothelial cell proliferation and the antiapoptosis process during embryonic vascular development.
Collapse
Affiliation(s)
- Danyang Chong
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing 210093, China
| | - Zhong Chen
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing 210093, China
| | - Shan Guan
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing 210093, China
| | - Tongyu Zhang
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing 210093, China
| | - Na Xu
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing 210093, China
| | - Yue Zhao
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing 210093, China.
| | - Chaojun Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing 210093, China.
| |
Collapse
|
13
|
Sang YJ, Wang Q, Zheng F, Hua Y, Wang XY, Zhang JZ, Li K, Wang HQ, Zhao Y, Zhu MS, Sun HX, Li CJ. Ggps1 deficiency in the uterus results in dystocia by disrupting uterine contraction. J Mol Cell Biol 2020; 13:116-127. [PMID: 33340314 PMCID: PMC8104943 DOI: 10.1093/jmcb/mjaa066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 12/01/2022] Open
Abstract
Dystocia is a serious problem for pregnant women, and it increases the cesarean section rate. Although uterine dysfunction has an unknown etiology, it is responsible for cesarean delivery and clinical dystocia, resulting in neonatal morbidity and mortality; thus, there is an urgent need for novel therapeutic agents. Previous studies indicated that statins, which inhibit the mevalonate (MVA) pathway of cholesterol synthesis, can reduce the incidence of preterm birth, but the safety of statins for pregnant women has not been thoroughly evaluated. Therefore, to unambiguously examine the function of the MVA pathway in pregnancy and delivery, we employed a genetic approach by using myometrial cell-specific deletion of geranylgeranyl pyrophosphate synthase (Ggps1) mice. We found that Ggps1 deficiency in myometrial cells caused impaired uterine contractions, resulting in disrupted embryonic placing and dystocia. Studies of the underlying mechanism suggested that Ggps1 is required for uterine contractions to ensure successful parturition by regulating RhoA prenylation to activate the RhoA/Rock2/p-MLC pathway. Our work indicates that perturbing the MVA pathway might result in problems during delivery for pregnant females, but modifying protein prenylation with supplementary farnesyl pyrophosphate or geranylgeranyl pyrophosphate might be a strategy to avoid side effects.
Collapse
Affiliation(s)
- Yong-Juan Sang
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Qiang Wang
- Department of Neurosurgery, Jingling Hospital, School of Medicine, Nanjing University, Nanjing 210002, China
| | - Feng Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Yue Hua
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Xin-Ying Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Jing-Zi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Kang Li
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Hai-Quan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Yue Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Hai-Xiang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | - Chao-Jun Li
- State Key Laboratory of Pharmaceutical Biotechnology, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing 210093, China
| |
Collapse
|
14
|
Foley AR, Zou Y, Dunford JE, Rooney J, Chandra G, Xiong H, Straub V, Voit T, Romero N, Donkervoort S, Hu Y, Markello T, Horn A, Qebibo L, Dastgir J, Meilleur KG, Finkel RS, Fan Y, Mamchaoui K, Duguez S, Nelson I, Laporte J, Santi M, Malfatti E, Maisonobe T, Touraine P, Hirano M, Hughes I, Bushby K, Oppermann U, Böhm J, Jaiswal JK, Stojkovic T, Bönnemann CG. GGPS1 Mutations Cause Muscular Dystrophy/Hearing Loss/Ovarian Insufficiency Syndrome. Ann Neurol 2020; 88:332-347. [PMID: 32403198 PMCID: PMC7496979 DOI: 10.1002/ana.25772] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE A hitherto undescribed phenotype of early onset muscular dystrophy associated with sensorineural hearing loss and primary ovarian insufficiency was initially identified in 2 siblings and in subsequent patients with a similar constellation of findings. The goal of this study was to understand the genetic and molecular etiology of this condition. METHODS We applied whole exome sequencing (WES) superimposed on shared haplotype regions to identify the initial biallelic variants in GGPS1 followed by GGPS1 Sanger sequencing or WES in 5 additional families with the same phenotype. Molecular modeling, biochemical analysis, laser membrane injury assay, and the generation of a Y259C knock-in mouse were done. RESULTS A total of 11 patients in 6 families carrying 5 different biallelic pathogenic variants in specific domains of GGPS1 were identified. GGPS1 encodes geranylgeranyl diphosphate synthase in the mevalonate/isoprenoid pathway, which catalyzes the synthesis of geranylgeranyl pyrophosphate, the lipid precursor of geranylgeranylated proteins including small guanosine triphosphatases. In addition to proximal weakness, all but one patient presented with congenital sensorineural hearing loss, and all postpubertal females had primary ovarian insufficiency. Muscle histology was dystrophic, with ultrastructural evidence of autophagic material and large mitochondria in the most severe cases. There was delayed membrane healing after laser injury in patient-derived myogenic cells, and a knock-in mouse of one of the mutations (Y259C) resulted in prenatal lethality. INTERPRETATION The identification of specific GGPS1 mutations defines the cause of a unique form of muscular dystrophy with hearing loss and ovarian insufficiency and points to a novel pathway for this clinical constellation. ANN NEUROL 2020;88:332-347.
Collapse
Affiliation(s)
- A. Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Yaqun Zou
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - James E. Dunford
- Botnar Research Centre, National Institute for Health Research Biomedical Research Centre OxfordUniversity of OxfordOxfordUnited Kingdom
| | - Jachinta Rooney
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Goutam Chandra
- Children's National Health SystemCenter for Genetic Medicine ResearchWashingtonDistrict of ColumbiaUSA
| | - Hui Xiong
- Department of PediatricsPeking University First HospitalBeijingChina
| | - Volker Straub
- Institute of Genetic MedicineInternational Centre for LifeNewcastle upon TyneUnited Kingdom
| | - Thomas Voit
- Great Ormond Street Hospital Biomedical Research CentreGreat Ormond Street Institute of Child Health, University College LondonLondonUnited Kingdom
| | - Norma Romero
- National Institute of Health and Medical Research U974, Sorbonne UniversityInstitute of Myology, APHPParisFrance
- Neuromuscular Morphology UnitInstitute of Myology, Pitié‐Salpêtrière HospitalParisFrance
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Thomas Markello
- National Institutes of Health Undiagnosed Diseases ProgramNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Adam Horn
- Children's National Health SystemCenter for Genetic Medicine ResearchWashingtonDistrict of ColumbiaUSA
| | - Leila Qebibo
- Unit of Medical Genetics and OncogeneticsUniversity HospitalFesMorocco
| | - Jahannaz Dastgir
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
- Department of Pediatric NeurologyGoryeb Children's HospitalMorristownNew JerseyUSA
| | - Katherine G. Meilleur
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
- BiogenCambridgeMassachusettsUSA
| | - Richard S. Finkel
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Translational Neuroscience ProgramSt. Jude Children’s Research HospitalMemphisTennesseeUSA
| | - Yanbin Fan
- Department of PediatricsPeking University First HospitalBeijingChina
| | - Kamel Mamchaoui
- National Institute of Health and Medical Research U974, Sorbonne UniversityInstitute of Myology, APHPParisFrance
| | - Stephanie Duguez
- National Institute of Health and Medical Research U974, Sorbonne UniversityInstitute of Myology, APHPParisFrance
- School of Biomedical SciencesUlster UniversityDerryUnited Kingdom
| | - Isabelle Nelson
- National Institute of Health and Medical Research U974, Sorbonne UniversityInstitute of Myology, APHPParisFrance
| | - Jocelyn Laporte
- Institute of Genetics and Molecular and Cellular Biology, National Institute of Health and Medical Research U1258, National Center for Scientific Research UMR7104University of StrasbourgIllkirchFrance
| | - Mariarita Santi
- Department of Pathology and Laboratory MedicineChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Edoardo Malfatti
- National Institute of Health and Medical Research U974, Sorbonne UniversityInstitute of Myology, APHPParisFrance
- U1179 University of Versailles Saint‐Quentin‐en‐Yvelines‐National Institute of Health and Medical ResearchParis‐Saclay UniversityVersaillesFrance
- Neurology Department, Reference Center for Neuromuscular Diseases North/East/Ile de FranceRaymond‐Poincaré University HospitalGarchesFrance
| | - Thierry Maisonobe
- Department of Clinical NeurophysiologyPitié‐Salpêtrière HospitalParisFrance
| | - Philippe Touraine
- Department of Endocrinology and Reproductive Medicine, Faculty of Medicine, Sorbonne University, Pitié‐Salpêtrière Hospital, APHPReference Center for Rare Endocrine Diseases of Growth and Development and Reference Center for Rare Gynecologic DisordersParisFrance
| | - Michio Hirano
- Department of Neurology, H. Houston Merritt Neuromuscular Research Center Columbia University Medical CenterNew YorkNew YorkUSA
| | - Imelda Hughes
- Department of Paediatric NeurologyRoyal Manchester Children's HospitalManchesterUnited Kingdom
| | - Kate Bushby
- Institute of Genetic MedicineInternational Centre for LifeNewcastle upon TyneUnited Kingdom
| | - Udo Oppermann
- Botnar Research Centre, National Institute for Health Research Biomedical Research Centre OxfordUniversity of OxfordOxfordUnited Kingdom
- Structural Genomics ConsortiumUniversity of OxfordOxfordUnited Kingdom
- Freiburg Institute of Advanced StudiesUniversity of FreiburgFreiburgGermany
| | - Johann Böhm
- Institute of Genetics and Molecular and Cellular Biology, National Institute of Health and Medical Research U1258, National Center for Scientific Research UMR7104University of StrasbourgIllkirchFrance
| | - Jyoti K. Jaiswal
- Children's National Health SystemCenter for Genetic Medicine ResearchWashingtonDistrict of ColumbiaUSA
- Department of Genomics and Precision MedicineGeorge Washington University School of Medicine and Health SciencesWashingtonDistrict of ColumbiaUSA
| | - Tanya Stojkovic
- Faculty of Medicine, Sorbonne University, Pitié‐Salpêtrière Hospital, APHPReference Center for Neuromuscular Diseases North/East/Ile de FranceParisFrance
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
15
|
Li H, You L, Tian Y, Guo J, Fang X, Zhou C, Shi L, Su Y. DPAGT1-Mediated Protein N-Glycosylation Is Indispensable for Oocyte and Follicle Development in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000531. [PMID: 32714760 PMCID: PMC7375233 DOI: 10.1002/advs.202000531] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/29/2020] [Indexed: 05/11/2023]
Abstract
Post-translational modification of proteins by N-linked glycosylation is crucial for many life processes. However, the exact contribution of N-glycosylation to mammalian female reproduction remains largely undefined. Here, DPAGT1, the enzyme that catalyzes the first step of protein N-glycosylation, is identified to be indispensable for oocyte development in mice. Dpagt1 missense mutation (c. 497A>G; p. Asp166Gly) causes female subfertility without grossly affecting other functions. Mutant females ovulate fewer eggs owing to defective development of growing follicles. Mutant oocytes have a thin and fragile zona pellucida (ZP) due to the reduction in glycosylation of ZP proteins, and display poor developmental competence after fertilization in vitro. Moreover, completion of the first meiosis is accelerated in mutant oocytes, which is coincident with the elevation of aneuploidy. Mechanistically, transcriptomic analysis reveals the downregulation of a number of transcripts essential for oocyte meiotic progression and preimplantation development (e.g., Pttgt1, Esco2, Orc6, and Npm2) in mutant oocytes, which could account for the defects observed. Furthermore, conditional knockout of Dpagt1 in oocytes recapitulates the phenotypes observed in Dpagt1 mutant females, and causes complete infertility. Taken together, these data indicate that protein N-glycosylation in oocytes is essential for female fertility in mammals by specific control of oocyte development.
Collapse
Affiliation(s)
- Hui Li
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Liji You
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Yufeng Tian
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Jing Guo
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Xianbao Fang
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Chenmin Zhou
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Lanying Shi
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - You‐Qiang Su
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjing211166P. R. China
- Women's Hospital of Nanjing Medical UniversityNanjing Maternity and Child Health HospitalNanjing Medical UniversityNanjing211166P. R. China
- Collaborative Innovation Center of Genetics and DevelopmentFudan UniversityShanghai200433P. R. China
- Key Laboratory of Model Animal ResearchNanjing Medical UniversityNanjing211166P. R. China
| |
Collapse
|
16
|
Genomic sequencing highlights the diverse molecular causes of Perrault syndrome: a peroxisomal disorder (PEX6), metabolic disorders (CLPP, GGPS1), and mtDNA maintenance/translation disorders (LARS2, TFAM). Hum Genet 2020; 139:1325-1343. [PMID: 32399598 DOI: 10.1007/s00439-020-02176-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/02/2020] [Indexed: 02/08/2023]
Abstract
Perrault syndrome is a rare heterogeneous condition characterised by sensorineural hearing loss and premature ovarian insufficiency. Additional neuromuscular pathology is observed in some patients. There are six genes in which variants are known to cause Perrault syndrome; however, these explain only a minority of cases. We investigated the genetic cause of Perrault syndrome in seven affected individuals from five different families, successfully identifying the cause in four patients. This included previously reported and novel causative variants in known Perrault syndrome genes, CLPP and LARS2, involved in mitochondrial proteolysis and mitochondrial translation, respectively. For the first time, we show that pathogenic variants in PEX6 can present clinically as Perrault syndrome. PEX6 encodes a peroxisomal biogenesis factor, and we demonstrate evidence of peroxisomal dysfunction in patient serum. This study consolidates the clinical overlap between Perrault syndrome and peroxisomal disorders, and highlights the need to consider ovarian function in individuals with atypical/mild peroxisomal disorders. The remaining patients had variants in candidate genes such as TFAM, involved in mtDNA transcription, replication, and packaging, and GGPS1 involved in mevalonate/coenzyme Q10 biosynthesis and whose enzymatic product is required for mouse folliculogenesis. This genomic study highlights the diverse molecular landscape of this poorly understood syndrome.
Collapse
|
17
|
De Cian MC, Gregoire EP, Le Rolle M, Lachambre S, Mondin M, Bell S, Guigon CJ, Chassot AA, Chaboissier MC. R-spondin2 signaling is required for oocyte-driven intercellular communication and follicular growth. Cell Death Differ 2020; 27:2856-2871. [PMID: 32341451 PMCID: PMC7493947 DOI: 10.1038/s41418-020-0547-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
R-spondin2 (RSPO2) is a member of the R-spondin family, which are secreted activators of the WNT/β-catenin (CTNNB1) signaling pathway. In the mouse postnatal ovary, WNT/CTNNB1 signaling is active in the oocyte and in the neighboring supporting cells, the granulosa cells. Although the role of Rspo2 has been previously studied using in vitro experiments, the results are conflicting and the in vivo ovarian function of Rspo2 remains unclear. In the present study, we found that RSPO2/Rspo2 expression is restricted to the oocyte of developing follicles in both human and mouse ovaries from the beginning of the follicular growth. In mice, genetic deletion of Rspo2 does not impair oocyte growth, but instead prevents cell cycle progression of neighboring granulosa cells, thus resulting in an arrest of follicular growth. We further show this cell cycle arrest to be independent of growth promoting GDF9 signaling, but rather associated with a downregulation of WNT/CTNNB1 signaling in granulosa cells. To confirm the contribution of WNT/CTNNB1 signaling in granulosa cell proliferation, we induced cell type specific deletion of Ctnnb1 postnatally. Strikingly, follicles lacking Ctnnb1 failed to develop beyond the primary stage. These results show that RSPO2 acts in a paracrine manner to sustain granulosa cell proliferation in early developing follicles. Taken together, our data demonstrate that the activation of WNT/CTNNB1 signaling by RSPO2 is essential for oocyte-granulosa cell interactions that drive maturation of the ovarian follicles and eventually female fertility.
Collapse
Affiliation(s)
- Marie-Cécile De Cian
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France.,Université de Corte, Corte, France
| | | | | | | | - Magali Mondin
- Université de Bordeaux, UMS 3420 CNRS-US4 Inserm, Pôle d'imagerie photonique, Bordeaux, France
| | - Sheila Bell
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Céline J Guigon
- Université de Paris, BFA, UMR 8251, CNRS, ERL U1133, Inserm, Paris, France
| | | | | |
Collapse
|
18
|
Chen M, Wan B, Zhu S, Zhang F, Jin J, Li X, Wang X, Lv Y, Chen C, Lv T, Song Y. Geranylgeranyl diphosphate synthase deficiency aggravates lung fibrosis in mice by modulating TGF-β1/BMP-4 signaling. Biol Chem 2020; 400:1617-1627. [PMID: 31120854 DOI: 10.1515/hsz-2019-0168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/12/2019] [Indexed: 02/06/2023]
Abstract
Geranylgeranyl diphosphate synthase (GGPPS) is an enzyme that catalyzes the synthesis of geranylgeranyl pyrophosphate (GGPP). GGPPS is implicated in many disorders, but its role in idiopathic pulmonary fibrosis (IPF) remains unclear. This study aimed to investigate the role of GGPPS in IPF. We established bleomycin-induced lung injury in a lung-specific GGPPS-deficient mouse (GGPPS-/-) and detected GGPPS expression in lung tissues by Western blot and immunohistochemistry analysis. We found that GGPPS expression increased during lung injury and fibrosis in mice induced by bleomycin, and GGPPS deficiency augmented lung fibrosis. GGPPS deficiency activated lung fibroblast by facilitating transforming growth factor β1 while antagonizing bone morphogenetic protein 4 signaling. Notably, the supplementation of exogenous GGPP mitigated lung fibrosis in GGPPS-/- mice induced by bleomycin. In conclusion, our findings suggest that GGPPS provides protection against pulmonary fibrosis and that the restoration of protein geranylgeranylation may benefit statin-induced lung injury.
Collapse
Affiliation(s)
- Meizi Chen
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China.,Department of General Internal Medicine, The First People's Hospital of Chenzhou, Chenzhou 423000, Hunan, P.R. China
| | - Bing Wan
- Department of Respiratory and Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Suhua Zhu
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Fang Zhang
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Jiajia Jin
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Xinying Li
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Xianghai Wang
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Yanling Lv
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Cen Chen
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Tangfeng Lv
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), Nanjing 210002, P.R. China
| | - Yong Song
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing Clinical School of Southern Medical University (Guangzhou), 305 Zhongshan Road, Nanjing 210002, P.R. China
| |
Collapse
|
19
|
Li M, Min W, Wang J, Wang L, Li Y, Zhou N, Yang Z, Qian Q. Effects of mevalonate kinase interference on cell differentiation, apoptosis, prenylation and geranylgeranylation of human keratinocytes are attenuated by farnesyl pyrophosphate or geranylgeranyl pyrophosphate. Exp Ther Med 2020; 19:2861-2870. [PMID: 32256770 PMCID: PMC7086283 DOI: 10.3892/etm.2020.8569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 11/14/2019] [Indexed: 12/20/2022] Open
Abstract
Mevalonate kinase (MVK) mutations were previously identified in disseminated superficial actinic porokeratosis. However, the role of MVK in differentiation, apoptosis and prenylation of keratinocytes requires further investigation. Farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP) of the mevalonate pathway attach to small G proteins, and serve as molecular switches in biochemical pathways. Therefore, the aim of the present study was to investigate the role of MVK in the expression of keratin 1 and involucrin, apoptosis, protein prenylation and the processing of small G proteins. HaCat human keratinocytes were transfected with viruses carrying MVK interference and overexpression vectors, respectively. The mRNA expression of MVK, keratin 1 and involucrin was detected by reverse transcription-quantitative PCR. Protein expression of MVK, keratin 1, involucrin, lamin A, HRAS, KRAS, NRAS, Rho E, Rho B, Rho A, RAC1 and cdc42 in HaCat cells was detected by western blotting. The apoptotic rates of HaCat cells and protein prenylation levels were examined by flow cytometry. The expression of MVK in HaCat cells was significantly decreased in the interference groups, and markedly increased in the overexpression group compared with the negative control groups. The mRNA and protein expression levels of keratin 1 and involucrin were significantly decreased following interference of MVK expression, and the decrease was markedly attenuated by FPP. Furthermore, the apoptotic rate was markedly increased following MVK interference, and the increase was significantly attenuated by GGPP. The overexpression of MVK significantly decreased the apoptotic rate of HaCat cells. The prenylation levels after MVK interference was notably decreased, which was markedly attenuated by GGPP. The overexpression of MVK significantly increased the prenylation levels of HaCat cells. FPP or GGPP reversed MVK interference-induced decrease in geranylgeranylation levels of lamin A, HRAS, KRAS, NRAS, Rho E, Rho B, Rho A, RAC1 and cdc42. In conclusion, MVK interference decreases the expression of differentiation markers, increases apoptosis, and decreases protein prenylation and geranylgeranylation levels in keratinocytes. These changes are attenuated by FPP or GGPP.
Collapse
Affiliation(s)
- Min Li
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wei Min
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jianbo Wang
- Department of Dermatology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, P.R. China
| | - Lu Wang
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yan Li
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Naihui Zhou
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ziliang Yang
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qihong Qian
- Department of Dermatology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
20
|
Zhao Y, Zhao MF, Jiang S, Wu J, Liu J, Yuan XW, Shen D, Zhang JZ, Zhou N, He J, Fang L, Sun XT, Xue B, Li CJ. Liver governs adipose remodelling via extracellular vesicles in response to lipid overload. Nat Commun 2020; 11:719. [PMID: 32024826 PMCID: PMC7002740 DOI: 10.1038/s41467-020-14450-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Lipid overload results in lipid redistribution among metabolic organs such as liver, adipose, and muscle; therefore, the interplay between liver and other organs is important to maintain lipid homeostasis. Here, we show that liver responds to lipid overload first and sends hepatocyte-derived extracellular vesicles (EVs) targeting adipocytes to regulate adipogenesis and lipogenesis. Geranylgeranyl diphosphate synthase (Ggpps) expression in liver is enhanced by lipid overload and regulates EV secretion through Rab27A geranylgeranylation. Consistently, liver-specific Ggpps deficient mice have reduced fat adipose deposition. The levels of several EV-derived miRNAs in the plasma of non-alcoholic fatty liver disease (NAFLD) patients are positively correlated with body mass index (BMI), and these miRNAs enhance adipocyte lipid accumulation. Thus, we highlight an inter-organ mechanism whereby the liver senses different metabolic states and sends corresponding signals to remodel adipose tissue to adapt to metabolic changes in response to lipid overload.
Collapse
Affiliation(s)
- Yue Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
- MOE Key Laboratory of Model Animals for Disease Study, Department of Hepatobiliary Surgery & Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China
| | - Meng-Fei Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
| | - Shan Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
| | - Jing Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Jia Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
| | - Xian-Wen Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
- MOE Key Laboratory of Model Animals for Disease Study, Department of Hepatobiliary Surgery & Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China
| | - Di Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
| | - Jing-Zi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China
| | - Nan Zhou
- MOE Key Laboratory of Model Animals for Disease Study, Department of Hepatobiliary Surgery & Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China
| | - Jian He
- MOE Key Laboratory of Model Animals for Disease Study, Department of Hepatobiliary Surgery & Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China
| | - Lei Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China.
- MOE Key Laboratory of Model Animals for Disease Study, Department of Hepatobiliary Surgery & Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China.
| | - Xi-Tai Sun
- MOE Key Laboratory of Model Animals for Disease Study, Department of Hepatobiliary Surgery & Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China.
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| | - Chao-Jun Li
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University & Model Animal Research Center, Nanjing University, Nanjing, 210093, China.
- MOE Key Laboratory of Model Animals for Disease Study, Department of Hepatobiliary Surgery & Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China.
| |
Collapse
|
21
|
Jia WJ, Tang QL, Jiang S, Sun SQ, Xue B, Qiu YD, Li CJ, Mao L. Conditional loss of geranylgeranyl diphosphate synthase alleviates acute obstructive cholestatic liver injury by regulating hepatic bile acid metabolism. FEBS J 2020; 287:3328-3345. [PMID: 31905247 DOI: 10.1111/febs.15204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 10/31/2019] [Accepted: 01/04/2020] [Indexed: 12/12/2022]
Abstract
Previous studies have suggested that metabolites in the mevalonate pathway are involved in hepatic bile acid metabolism, yet the details of this relationship remain unknown. In this study, we found that the hepatic farnesyl pyrophosphate (FPP) level and the ratio of FPP to geranylgeranyl pyrophosphate (GGPP) were increased in mice with acute obstructive cholestasis compared with mice that underwent a sham operation. In addition, the livers of the mice with acute obstructive cholestasis showed lower expression of geranylgeranyl diphosphate synthase (GGPPS), which synthesizes GGPP from FPP. When Ggps1 was conditionally deleted in the liver, amelioration of liver injury, as shown by downregulation of the hepatic inflammatory response and decreased hepatocellular apoptosis, was found after ligation of the common bile duct and cholecystectomy (BDLC). Subsequently, liquid chromatography/mass spectrometry analysis showed that knocking out Ggps1 decreased the levels of hepatic bile acids, including hydrophobic bile acids. Mechanistically, the disruption of Ggps1 increased the levels of hepatic FPP and its metabolite farnesol, thereby resulting in farnesoid X receptor (FXR) activation, which modulated hepatic bile acid metabolism and reduced hepatic bile acids. It was consistently indicated that digeranyl bisphosphonate, a specific inhibitor of GGPPS, and GW4064, an agonist of FXR, could also alleviate acute obstructive cholestatic liver injury in vivo. In general, GGPPS is critical for modulating acute obstructive cholestatic liver injury, and the inhibition of GGPPS ameliorates acute obstructive cholestatic liver injury by decreasing hepatic bile acids, which is possibly achieved through the activation of FXR-induced bile acid metabolism.
Collapse
Affiliation(s)
- Wen-Jun Jia
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, China.,Ministry of Education Key Laboratory of Model Animal for Disease Study, the School of Medicine and Model Animal Research Center of Nanjing University, China.,Department of General Surgery, the Affiliated Drum Tower Hospital of Medical School of Nanjing University, China
| | - Qiao-Li Tang
- Ministry of Education Key Laboratory of Model Animal for Disease Study, the School of Medicine and Model Animal Research Center of Nanjing University, China
| | - Shan Jiang
- Ministry of Education Key Laboratory of Model Animal for Disease Study, the School of Medicine and Model Animal Research Center of Nanjing University, China
| | - Shi-Quan Sun
- Department of General Surgery, the Affiliated Drum Tower Hospital of Medical School of Nanjing University, China
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yu-Dong Qiu
- Department of General Surgery, the Affiliated Drum Tower Hospital of Medical School of Nanjing University, China
| | - Chao-Jun Li
- Ministry of Education Key Laboratory of Model Animal for Disease Study, the School of Medicine and Model Animal Research Center of Nanjing University, China
| | - Liang Mao
- Department of General Surgery, the Affiliated Drum Tower Hospital of Medical School of Nanjing University, China
| |
Collapse
|
22
|
Qin Y, Tang T, Li W, Liu Z, Yang X, Shi X, Sun G, Liu X, Wang M, Liang X, Cong P, Mo D, Liu X, Chen Y, He Z. Bone Morphogenetic Protein 15 Knockdown Inhibits Porcine Ovarian Follicular Development and Ovulation. Front Cell Dev Biol 2019; 7:286. [PMID: 31803742 PMCID: PMC6877722 DOI: 10.3389/fcell.2019.00286] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
Bone morphogenetic protein 15 (BMP15) is strongly associated with animal reproduction and woman reproductive disease. As a multifunctional oocyte-specific secret factor, BMP15 controls female fertility and follicular development in both species-specific and dosage-sensitive manners. Previous studies found that BMP15 played a critical role in follicular development and ovulation rate in mono-ovulatory mammalian species, especially in sheep and human, but study on knockout mouse model implied that BMP15 possibly has minimal impact on female fertility of poly-ovulatory species. However, this needs to be validated in other poly-ovulatory species. To investigate the regulatory role of BMP15 on porcine female fertility, we generated a BMP15-knockdown pig model through somatic nuclear transfer technology. The BMP15-knockdown gilts showed markedly reduced fertility accompanied by phenotype of dysplastic ovaries containing significantly declined number of follicles, increased number of abnormal follicles, and abnormally enlarged antral follicles resulting in disordered ovulation, which is remarkably different from the unchanged fertility observed in BMP15 knockout mice. Molecular and transcriptome analysis revealed that the knockdown of BMP15 significantly affected both granulosa cells (GCs) and oocytes development, including suppression of cell proliferation, differentiation, and follicle stimulating hormone receptor (Fshr) expression, leading to premature luteinization and reduced estradiol (E2) production in GCs, and simultaneously decreased quality and meiotic maturation of oocyte. Our results provide in vivo evidence of the essential role of BMP15 in porcine ovarian and follicular development, and new insight into the complicated regulatory function of BMP15 in female fertility of poly-ovulatory species.
Collapse
Affiliation(s)
- Yufeng Qin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Tao Tang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhiguo Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaoliang Yang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xuan Shi
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Guanjie Sun
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaofeng Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Min Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xinyu Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peiqing Cong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
23
|
Cao Z, Gao D, Xu T, Zhang L, Tong X, Zhang D, Wang Y, Ning W, Qi X, Ma Y, Ji K, Yu T, Li Y, Zhang Y. Circular RNA profiling in the oocyte and cumulus cells reveals that circARMC4 is essential for porcine oocyte maturation. Aging (Albany NY) 2019; 11:8015-8034. [PMID: 31562810 PMCID: PMC6781969 DOI: 10.18632/aging.102315] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 09/21/2019] [Indexed: 12/13/2022]
Abstract
Thousands of circular RNAs (circRNAs) have been recently discovered in cumulus cells and oocytes from several species. However, the expression and function of circRNA during porcine oocyte meiotic maturation have been never examined. Here, we separately identified 7,067 and 637 circRNAs in both cumulus cells and oocytes via deep sequencing and bioinformatic analysis. Further analysis revealed that a faction of circRNAs is differentially expressed (DE) in a developmental stage-specific manner. The host genes of DE circRNAs are markedly enriched to multiple signaling pathways associated with cumulus cell function and oocyte maturation. Additionally, most DE circRNAs harbor several miRNA targets, suggesting that these DE circRNAs potentially act as miRNA sponge. Importantly, we found that maternal circARMC4 knockdown by siRNA microinjection caused a severely impaired chromosome alignment, and significantly inhibited first polar body extrusion and early embryo development. Taken together, these results demonstrate for the first time that circRNAs are abundantly and dynamically expressed in a developmental stage-specific manner in cumulus cells and oocytes, and maternally expressed circARMC4 is essential for porcine oocyte meiotic maturation and early embryo development.
Collapse
Affiliation(s)
- Zubing Cao
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Di Gao
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Tengteng Xu
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ling Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xu Tong
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Dandan Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yiqing Wang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Wei Ning
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xin Qi
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yangyang Ma
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Kaiyuan Ji
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Tong Yu
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yunsheng Li
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yunhai Zhang
- Anhui Province Key Laboratory of Local Livestock and Poultry, Genetical Resource Conservation and Breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
24
|
Guo T, Zhang J, Yao W, Du X, Li Q, Huang L, Ma M, Li Q, Liu H, Pan Z. CircINHA resists granulosa cell apoptosis by upregulating CTGF as a ceRNA of miR-10a-5p in pig ovarian follicles. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:194420. [PMID: 31476383 DOI: 10.1016/j.bbagrm.2019.194420] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
Mammalian ovarian follicular atresia is a complex and fine-regulated biological process with active involvement of connective tissue growth factor (CTGF). The emergence of studies of endogenous non-coding RNAs has raised a new aspect for exploration of the regulatory mechanisms involved in follicular atresia. Here, we aimed to illustrate a circRNA involved in the CTGF regulatory pathway during the apoptosis and follicular atresia of pig granulosa cells (GCs). We first detected a decreased expression pattern of CTGF during follicular atresia using IHC, FISH and qRT-PCR and confirmed the anti-apoptosis effect of CTGF in GCs in vitro by CTGF siRNA knockdown. Then, we used a dual luciferase activity assay to demonstrate CTGF as a direct functional target of miR-10a-5p, which was upregulated in atresic follicles and promoted the apoptosis of GCs in vitro. The negative effect of miR-10a-5p on GC viability was confirmed by cell cycle assays, cell proliferation/apoptosis assays and the WB detection of marker proteins. More importantly, we identified a novel circRNA, termed circINHA, that was downregulated during atresia in ovarian follicles, and we confirmed a direct interaction between miR-10a-5p and circINHA. Finally, we demonstrated that circINHA promoted GCs proliferation and inhibited GCs apoptosis via CTGF as a competing endogenous RNA (ceRNA) that directly bound to miR-10a-5p. Taken together, this study provides evidence for the circINHA/miR-10a-5p/CTGF regulatory pathway in follicular GC apoptosis and provides novel insights into the role of circRNAs in the modulation of ovarian physiological functions.
Collapse
Affiliation(s)
- Tianya Guo
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Jinbi Zhang
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Wang Yao
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - QiQi Li
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Long Huang
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Menglan Ma
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China
| | - Zengxiang Pan
- College of Animal Science and Technology, Nanjing Agriculture University, 210095, China; National Experimental Teaching Demonstration Center of Animal Science, China.
| |
Collapse
|
25
|
The alteration of RhoA geranylgeranylation and Ras farnesylation breaks the integrity of the blood-testis barrier and results in hypospermatogenesis. Cell Death Dis 2019; 10:450. [PMID: 31171774 PMCID: PMC6554403 DOI: 10.1038/s41419-019-1688-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 05/09/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022]
Abstract
Non-obstructive azoospermia (NOA) severely affects male infertility, however, the deep mechanisms of this disease are rarely interpreted. In this study, we find that undifferentiated spermatogonial stem cells (SSCs) still exist in the basal compartment of the seminiferous tubules and the blood–testis barrier (BTB) formed by the interaction of neighbor Sertoli cells (SCs) is incomplete in NOA patients with spermatogenic maturation arrest. The adhesions between SCs and germ cells (GCs) are also broken in NOA patients. Meanwhile, the expression level of geranylgeranyl diphosphate synthase (Ggpps), a key enzyme in mevalonate metabolic pathway, is lower in NOA patients than that in obstructive azoospermia (OA) patients. After Ggpps deletion specifically in SCs, the mice are infertile and the phenotype of the SC-Ggpps−/− mice is similar to the NOA patients, where the BTB and the SC–GC adhesions are severely destroyed. Although SSCs are still found in the basal compartment of the seminiferous tubules, fewer mature spermatocyte and spermatid are found in SC-Ggpps−/− mice. Further examination suggests that the defect is mediated by the aberrant protein isoprenylation of RhoA and Ras family after Ggpps deletion. The exciting finding is that when the knockout mice are injected with berberine, the abnormal cell adhesions are ameliorated and spermatogenesis is partially restored. Our data suggest that the reconstruction of disrupted BTB is an effective treatment strategy for NOA patients with spermatogenic maturation arrest and hypospermatogenesis.
Collapse
|
26
|
Zhang P, Wang J, Lang H, Wang W, Liu X, Liu H, Tan C, Li X, Zhao Y, Wu X. MicroRNA-205 affects mouse granulosa cell apoptosis and estradiol synthesis by targeting CREB1. J Cell Biochem 2019; 120:8466-8474. [PMID: 30556190 DOI: 10.1002/jcb.28133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023]
Abstract
MicroRNA-205 (miR-205) is involved in various physiological and pathological processes, but its biological function in follicular atresia remains unclear. In this study, we investigated miR-205 expression in mouse granulosa cells (mGCs) and analyzed its functions in primary mGCs by performing a series of in vitro experiments. Quantitative real-time polymerase chain reaction showed that miR-205 expression was significantly higher in early atretic follicles and progressively atretic follicles than in healthy follicles. miR-205 overexpression in mGCs significantly promoted apoptosis and caspase-3/9 activities, as well as inhibited estrogen (E2) release and cytochrome P450 family 19 subfamily A polypeptide 1 (CYP19A1, a key gene in E2 production) expression. Bioinformatics and luciferase reporter assays revealed that the gene encoding cyclic AMP response element (CRE)-binding protein 1 (CREB1) was a direct target of miR-205 in mGCs. CREB1 upregulation partially rescued the effects of miR-205 on apoptosis, caspase-3/9 activities, E2 production, and CYP19A1 expression on mGCs. These results indicate that miR-205 might play an important role in ovarian follicular development and provide new insights into follicular atresia.
Collapse
Affiliation(s)
- Pengju Zhang
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Jun Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Hongyan Lang
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Weixia Wang
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Xiaohui Liu
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Haiyan Liu
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Chengcheng Tan
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Xintao Li
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Yumin Zhao
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Xinghong Wu
- Institute of Animal Sciences, Jilin Academy of Agricultural Sciences, Changchun, Jilin, China
| |
Collapse
|
27
|
Lai SS, Fu X, Cheng Q, Yu ZH, Jiang EZ, Zhao DD, Yu DC, Qiu YD, Gao X, Ju HX, Wang W, Jiang Q, Zhu MS, Li CJ, Xue B. HSC-specific knockdown of GGPPS alleviated CCl 4-induced chronic liver fibrosis through mediating RhoA/Rock pathway. Am J Transl Res 2019; 11:2382-2392. [PMID: 31105844 PMCID: PMC6511779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/07/2019] [Indexed: 06/09/2023]
Abstract
Hepatic stellate cells (HSCs) play a critical role in the pathogenesis and reversal of liver fibrosis. Targeting HSCs is of great significance in the treatment of hepatic fibrosis, and has attracted wide attention of scholars. Here we demonstrated that expression of geranylgeranyldiphosphate synthase (GGPPS) predominantly increased in HSCs in murine fibrotic liver. HSC-specific knockdown of GGPPS using vitamin A-coupled liposome carrying siRNA-ggpps decreased activation of HSCs and alleviated fiber accumulation in vivo. Furthermore, our in vitro studies showed that GGPPS was up-regulated during HSCs activation in TGF-β1-dependent manner. Inhibition of GGPPS suppressed TGF-β1 induced F-actin reorganization and HSCs activation in LX-2 cells. Further, we found that GGPPS regulated HSCs activation and liver fibrosis possibly by enhancing RhoA/Rock kinase signaling. So its concluded that GGPPS promotes liver fibrosis by activating HSCs, which may represent a potential target for anti-fibrosis therapies.
Collapse
Affiliation(s)
- Shan-Shan Lai
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal UniversityNanjing 210023, China
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Xiao Fu
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing UniversityNanjing 210093, China
| | - Qi Cheng
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Zi-Han Yu
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - En-Ze Jiang
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Dan-Dan Zhao
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- Research Center, Shanghai Chest Hospital, Shanghai Jiao Tong UniversityShanghai 200030, China
| | - De-Cai Yu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing UniversityNanjing 210093, China
| | - Yu-Dong Qiu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Medical School of Nanjing UniversityNanjing 210093, China
| | - Xiang Gao
- Key Laboratory of Model Animal for Disease Study of Ministry of Education, Model Animal Research Center, Nanjing UniversityNanjing 210093, China
| | - Huang-Xian Ju
- MOE Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing UniversityNanjing 210093, China
| | - Wei Wang
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing UniversityNanjing 210093, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing UniversityNanjing 210008, China
- Joint Research Center for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing UniversityNanjing 210093, China
| | - Min-Sheng Zhu
- Key Laboratory of Model Animal for Disease Study of Ministry of Education, Model Animal Research Center, Nanjing UniversityNanjing 210093, China
| | - Chao-Jun Li
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical UniversityNanjing 211166, China
- Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing UniversityNanjing 210093, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical UniversityNanjing 210009, China
| |
Collapse
|
28
|
Shen M, Li T, Zhang G, Wu P, Chen F, Lou Q, Chen L, Yin X, Zhang T, Wang J. Dynamic expression and functional analysis of circRNA in granulosa cells during follicular development in chicken. BMC Genomics 2019; 20:96. [PMID: 30700247 PMCID: PMC6354403 DOI: 10.1186/s12864-019-5462-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 01/17/2019] [Indexed: 01/17/2023] Open
Abstract
Background Circular RNA (circRNA) is a type of noncoding RNA involved in a variety of biological processes, especially in post-transcriptional regulation. The granulosa cells of follicles play a determining role in ovarian development. However, the function of circRNA in chicken follicles is unclear. To better understand the molecular mechanism underlying follicular development and granulosa cell function, we performed a strategy of second-generation sequencing and linear RNA depletion for granulosa cells from small yellow follicles (SYF, 5–8 mm), the smallest hierarchal follicles (F6, 9–12 mm), and the largest hierarchal follicles (F1, ~ 40 mm). Results We predicted a total of 11,642 circRNAs that distributed on almost all chromosomes. The majority of the splice lengths of circRNAs were 200–500 nt and mainly produced from intron and CDS regions. During follicle growth, differentially expressed (DE) circRNAs showed dynamic changes which were tissue- and stage-specific. The host genes of DE circRNAs were functionally enriched in GTPase activity and several pathways involved in reproduction. Moreover, bioinformatic prediction analysis for circRalGPS2 demonstrated that circRNAs from the same genes may share common miRNA to act as a sponge. The predicted target genes were enriched in various biological processes including cognition, cell communication, and regulation of signaling, and several pathways related to reproduction such as tight junction, oocyte meiosis, progesterone-mediated oocyte maturation, and GnRH signaling. Conclusions This study provides a starting point for further experimental investigations into chicken circRNAs and casts a light on the understanding of follicle development. Electronic supplementary material The online version of this article (10.1186/s12864-019-5462-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manman Shen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, 225216, China
| | - Tingting Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Genxi Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| | - Pengfei Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Fuxiang Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Qiuhong Lou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Lan Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Xuemei Yin
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Tao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Jinyu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
29
|
Xu R, Qin N, Xu X, Sun X, Chen X, Zhao J. Inhibitory effect of SLIT2 on granulosa cell proliferation mediated by the CDC42-PAKs-ERK1/2 MAPK pathway in the prehierarchical follicles of the chicken ovary. Sci Rep 2018; 8:9168. [PMID: 29907785 PMCID: PMC6003946 DOI: 10.1038/s41598-018-27601-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/06/2018] [Indexed: 01/09/2023] Open
Abstract
The SLIT2 ligand and ROBO receptors of the SLIT/ROBO pathway are expressed in hen ovarian follicles and have been shown to play critical roles in ovary development, cell proliferation and apoptosis in mammals. However, the exact roles of SLIT2 and the molecular mechanisms of chicken follicle development remain poorly understood. Here, we discovered that high levels of SLIT2 suppress FSHR, GDF9, STAR and CYP11A1 mRNA and protein expression in granulosa cells (GCs) and cell proliferation (p < 0.01). However, these inhibitory effects can be abolished by the siRNA-mediated knockdown of the ROBO1 and ROBO2 receptors. Furthermore, the activity of CDC42, which is a key Rho GTPase in the SLIT/ROBO pathway, is regulated by the ligand SLIT2 because the intrinsic GTPase activation activity of CDC42 is activated or repressed by regulating SRGAP1 expression (p < 0.01). The effects of the SLIT2 overexpression on GC proliferation and phosphorylation of the B-RAF, RAF1 and ERK1/2 kinases were completely abrogated by knocking down endogenous PAK1 and partially abrogated by the knockdown of PAK2 and PAK3 in the GCs. Collectively, our findings indicate that SLIT2 suppresses GC proliferation, differentiation and follicle selection mainly by a mechanism involving ROBO1 and ROBO2 and that this suppression is mediated by the CDC42-PAKs-ERK1/2 MAPK signaling cascade in the prehierarchical follicles of the chicken ovary.
Collapse
Affiliation(s)
- Rifu Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China. .,Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Changchun, 130118, People's Republic of China.
| | - Ning Qin
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China.,Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Changchun, 130118, People's Republic of China
| | - Xiaoxing Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Xue Sun
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Xiaoxia Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Jinghua Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| |
Collapse
|
30
|
Yao W, Pan Z, Du X, Zhang J, Li Q. miR-181b-induced SMAD7 downregulation controls granulosa cell apoptosis through TGF-β signaling by interacting with the TGFBR1 promoter. J Cell Physiol 2018; 233:6807-6821. [PMID: 29319157 DOI: 10.1002/jcp.26431] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
SMAD7 disrupts the TGF-β signaling pathway by influencing TGFBR1 stability and by blocking the binding of TGFBR1 to SMAD2/3. In this study, we showed that SMAD7 attenuated the TGF-β signaling pathway in ovarian granulosa cells (GCs) by regulating TGFBR1 transcriptional activity. To function as a transcription factor, SMAD7 downregulated the mRNA levels of TGFBR1 via direct binding to the SMAD-binding elements (SBEs) within the promoter region of pig TGFBR1. We also showed that SMAD7 enhanced porcine GC apoptosis by interrupting TGFBR1 and the TGF-β signaling pathway. Interestingly, miR-181b, a microRNA that is downregulated during porcine follicular atresia, was identified to be directly targeting SMAD7 at its 3'-UTR. By inhibiting SMAD7, miR-181b could inhibit GC apoptosis by activating the TGF-β signaling pathway. Our findings provide new insights into the mechanisms underlying the regulation of the TGF-β signaling pathway by SMAD7 and miR-181b.
Collapse
Affiliation(s)
- Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zengxiang Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jinbi Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|