1
|
Vilkaite A, Nguyen XP, Güzel CT, Gottschlich L, Bender U, Dietrich JE, Hinderhofer K, Strowitzki T, Rehnitz J. Beyond Repetition: The Role of Gray Zone Alleles in the Upregulation of FMR1-Binding miR-323a-3p and the Modification of BMP/SMAD-Pathway Gene Expression in Human Granulosa Cells. Int J Mol Sci 2025; 26:3192. [PMID: 40244008 PMCID: PMC11989689 DOI: 10.3390/ijms26073192] [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: 02/05/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The Fragile X mental retardation type 1 gene (FMR1) contains a CGG triplet cluster of varied length (30 repeats on average) located in its 5' UTR. In its premutated state (54-200 repeats), FMR1 contributes to the pathogenesis of premature ovarian insufficiency (POI). Its gray zone alleles (41-54 repeats) are supposed to impair the ovarian function as well. In the case of a CGG repeat length > 200, Fragile X syndrome occurs. Post-transcriptional expression of FMR1 is regulated by microRNAs. Although miR-323a-3p overexpression suppresses FMR1 in various tissues, this relationship has not been evaluated in the human ovary. Additionally, this microRNA targets SMADs, which are suggested regulators of ovarian cell proliferation, growth, and function. This study investigated how FMR1 allele lengths with CGG repeat numbers n < 55 (normal and gray zone genotypes) relate to miR-323a-3p expression and how they may impact associated SMAD expression in human granulosa cells. COV434 cells and patient-derived GCs were used to evaluate FMR1, miR-323a-3p, and BMP/SMAD-pathway member expression levels. Briefly, miR-323a-3p was significantly upregulated in GCs of the gray zone group compared to the normal allele group (p < 0.0001), while the FMR1 level did not vary. Furthermore, the gray zone group showed a significant upregulation of BMPR2, SMAD1, SMAD4, and SMAD9. In contrast, the miR-323a-3p transfection of COV434 cells significantly downregulated SMAD3, SMAD4, SMAD5, and SMAD9, while the FMR1 and SMAD1 levels remained stable. Our findings highlight a CGG repeat number-dependent upregulation of miR-323a-3p and an alteration of the BMP/SMAD pathway, suggesting that these changes happen and contribute to impaired ovarian function independently.
Collapse
Affiliation(s)
- Adriana Vilkaite
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Xuan Phuoc Nguyen
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Cansu Türkan Güzel
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Lucas Gottschlich
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Ulrike Bender
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Jens E. Dietrich
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Katrin Hinderhofer
- Institute of Human Genetics, University Heidelberg, 69120 Heidelberg, Germany;
| | - Thomas Strowitzki
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| | - Julia Rehnitz
- Department of Gynecological Endocrinology and Fertility Disorders, University Women’s Hospital, 69120 Heidelberg, Germany; (A.V.); (X.P.N.); (C.T.G.); (L.G.); (U.B.); (J.E.D.); (T.S.)
| |
Collapse
|
2
|
Yang X, He H, Wang P, Wang Y, Wang L, Yang F, Li J, Zhang H. Role of miRNAs in Bovine Oocyte Maturation and Reproductive Regulation. Int J Mol Sci 2025; 26:2828. [PMID: 40243418 PMCID: PMC11989158 DOI: 10.3390/ijms26072828] [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: 02/12/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 04/18/2025] Open
Abstract
MicroRNAs (miRNAs) are a class of endogenous small non-coding RNAs that regulate target gene expression in many eukaryotes. MiRNAs are essential for post-transcriptional regulation, influencing various biological functions, including oocyte growth and maturation, fertilization, early embryo development, and implantation. In recent decades, numerous studies have identified a substantial number of miRNAs associated with mammalian oocyte maturation and early embryo development, utilizing methods such as small RNA sequencing and modulating miRNA expression through overexpression or inhibition. In this review, we introduce the biosynthesis of miRNAs and their regulatory roles in germ cells, summarizing the expression patterns and post-transcriptional regulation of miRNAs during bovine oocyte maturation and early embryo development, as well as their potential application in bovine assisted reproductive technology (ART).
Collapse
Affiliation(s)
- Xiaogeng Yang
- Key Laboratory of Animal Medicine, Southwest Minzu University of Sichuan Province, Chengdu 610041, China; (X.Y.); (H.H.); (Y.W.); (F.Y.); (J.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Chengdu 610041, China
| | - Honghong He
- Key Laboratory of Animal Medicine, Southwest Minzu University of Sichuan Province, Chengdu 610041, China; (X.Y.); (H.H.); (Y.W.); (F.Y.); (J.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Chengdu 610041, China
| | - Peng Wang
- Animal Husbandry Science Institute of Ganzi Tibetan Autonomous Prefecture, Kangding 626000, China;
| | - Yaying Wang
- Key Laboratory of Animal Medicine, Southwest Minzu University of Sichuan Province, Chengdu 610041, China; (X.Y.); (H.H.); (Y.W.); (F.Y.); (J.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Chengdu 610041, China
| | - Linlin Wang
- College of Pharmacy and Food, Southwest Minzu University, Chengdu 610041, China;
| | - Falong Yang
- Key Laboratory of Animal Medicine, Southwest Minzu University of Sichuan Province, Chengdu 610041, China; (X.Y.); (H.H.); (Y.W.); (F.Y.); (J.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Chengdu 610041, China
| | - Jian Li
- Key Laboratory of Animal Medicine, Southwest Minzu University of Sichuan Province, Chengdu 610041, China; (X.Y.); (H.H.); (Y.W.); (F.Y.); (J.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Chengdu 610041, China
| | - Huizhu Zhang
- Key Laboratory of Animal Medicine, Southwest Minzu University of Sichuan Province, Chengdu 610041, China; (X.Y.); (H.H.); (Y.W.); (F.Y.); (J.L.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Chengdu 610041, China
| |
Collapse
|
3
|
Barancheshmeh M, Najafzadehvarzi H, Shokrzadeh N, Aram C. Comparative analysis of fennel essential oil and manganese in PCOS rat model via modulating miR-145 expression and structure-based virtual screening of IGF2R protein to address insulin resistance and obesity. OBESITY MEDICINE 2025; 53:100574. [DOI: 10.1016/j.obmed.2024.100574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
4
|
Myszczynski K, Szuszkiewicz J, Krawczynski K, Sikora M, Romaniewicz M, Guzewska MM, Zabielski P, Kaczmarek MM. In-Depth Analysis of miRNA Binding Sites Reveals the Complex Response of Uterine Epithelium to miR-26a-5p and miR-125b-5p During Early Pregnancy. Mol Cell Proteomics 2025; 24:100879. [PMID: 39536955 PMCID: PMC11758581 DOI: 10.1016/j.mcpro.2024.100879] [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: 03/17/2024] [Revised: 10/18/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Posttranscriptional regulation of gene expression by miRNAs likely makes significant contributions to mRNA abundance at the embryo-maternal interface. In this study, we investigated how miR-26a-5p and miR-125b-5p contribute to molecular changes occurring in the uterine luminal epithelium, which serves as the first site of signal exchange between the mother and the developing embryo. To measure de novo protein synthesis after miRNA delivery to primary uterine luminal epithelial cells, we used pulsed stable isotope labeling by amino acids (pSILACs). We found that both miRNAs alter the proteome of luminal epithelial cells, impacting numerous cellular functions, immune responses, as well as intracellular and second messenger signaling pathways. Additionally, we identified several features of miRNA-mRNA interactions that may influence the targeting efficiency of miR-26a-5p and miR-125b-5p. Overall, our study suggests a complex interaction of miR-26a-5p and miR-125b-5p with their respective targets. However, both appear to cooperatively function in modulating the cellular environment of the luminal epithelium, facilitating the morphological and molecular changes that occur during the intensive communication between the embryo and uterus at pregnancy.
Collapse
Affiliation(s)
- Kamil Myszczynski
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Joanna Szuszkiewicz
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Kamil Krawczynski
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Małgorzata Sikora
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marta Romaniewicz
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Maria M Guzewska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, Poland
| | - Monika M Kaczmarek
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland; Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland.
| |
Collapse
|
5
|
Nadri P, Nadri T, Gholami D, Zahmatkesh A, Hosseini Ghaffari M, Savvulidi Vargova K, Georgijevic Savvulidi F, LaMarre J. Role of miRNAs in assisted reproductive technology. Gene 2024; 927:148703. [PMID: 38885817 DOI: 10.1016/j.gene.2024.148703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Cellular proteins and the mRNAs that encode them are key factors in oocyte and sperm development, and the mechanisms that regulate their translation and degradation play an important role during early embryogenesis. There is abundant evidence that expression of microRNAs (miRNAs) is crucial for embryo development and are highly involved in regulating translation during oocyte and early embryo development. MiRNAs are a group of short (18-24 nucleotides) non-coding RNA molecules that regulate post-transcriptional gene silencing. The miRNAs are secreted outside the cell by embryos during preimplantation embryo development. Understanding regulatory mechanisms involving miRNAs during gametogenesis and embryogenesis will provide insights into molecular pathways active during gamete formation and early embryo development. This review summarizes recent findings regarding multiple roles of miRNAs in molecular signaling, plus their transport during gametogenesis and embryo preimplantation.
Collapse
Affiliation(s)
- Parisa Nadri
- Department of Animal Science, College of Agriculture, Isfahan University of Technology, Isfahan, Iran
| | - Touba Nadri
- Department of Animal Science, College of Agriculture, Urmia University, Urmia, Iran; Department of Animal Science, College of Agriculture, Tehran University, Karaj, Iran.
| | - Dariush Gholami
- Department of Microbial Biotechniligy, Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Azadeh Zahmatkesh
- Department of Anaerobic Vaccine Research and Production, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | | | - Karin Savvulidi Vargova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Filipp Georgijevic Savvulidi
- Department of Animal Science, Faculty of Agrobiology, Food and Natural Resources, Czech University, Prague, Kamýcká, Czech Republic
| | - Jonathan LaMarre
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Canada
| |
Collapse
|
6
|
Meewes C, Gupta K, Geisler WM. Role of microRNAs in immune regulation and pathogenesis of Chlamydia trachomatis and Chlamydia muridarum infections: a rapid review. Microbes Infect 2024; 26:105397. [PMID: 39025257 PMCID: PMC11609027 DOI: 10.1016/j.micinf.2024.105397] [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: 02/21/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
MicroRNAs in Chlamydia trachomatis (CT) and Chlamydia muridarum (CM) infections are an emerging topic of research that provide knowledge that could advance vaccine development and strategies for managing infection. This rapid review summarizes human and murine studies on miRNA expression in CT and CM infections in vivo and ex vivo.
Collapse
Affiliation(s)
- Chloe Meewes
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kanupriya Gupta
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - William M Geisler
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
7
|
Zhan X, Qi N, Toms D, Freiburger R, Fletcher L, Wang B, Li J. MiR-29b inhibits COC expansion and oocyte in vitro maturation via induction of ROS by targeting CYCS. Anim Reprod Sci 2024; 270:107598. [PMID: 39342691 DOI: 10.1016/j.anireprosci.2024.107598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024]
Abstract
Cumulus-oocyte complex (COC) expansion and oocyte maturation are crucial processes for embryo development and fertility across species. Although miR-29b has been detected in porcine ovarian granulosa cells, its specific role in regulating oocyte maturation remains largely unknown. In this study, using the pig as a model, we report that over-expression of miR-29b lead to a decrease of COC expansion area and inhibits oocyte maturation (P<0.05). This suppression correlated with a decrease expression of COC-expansion-associated genes, including SHAS2, ADAMTS1, ADAMTS2, ADAMTS17 and PTX 3 in both mural granulosa cells (mGCs) and cumulus granulosa cells (cGCs). Further investigation revealed that miR-29b over-expression induces reactive oxygen species (ROS) accumulation in both mGCs and cGCs, conversely, knock-down of miR-29b reverses all these effects. Treatment with the antioxidant β-mercaptoethanol alleviates ROS accumulation, rescues COC expansion and restores oocyte polar body formation impaired by miR-29b mimics. Computational analysis predicted CYCS, the gene encoding cytochrome C, as a potential target of miR-29b. Subsequent examination demonstrated that miR-29b downregulates CYCS at both mRNA and protein levels. Dual-luciferase reporter assays further confirmed that miR-29b interacts with the 3'-untranslated region (3'UTR) of CYCS. Over-expression of CYCS decreases ROS accumulation and promotes COC expansion (P<0.05). These results indicate that miR-29b regulates COC expansion and oocyte maturation in vitro by inducing ROS, likely through targeting of CYCS. This study sheds light on the role of miR-29b in oocyte maturation and provides insight into the regulatory function of miRNAs in ovarian physiology.
Collapse
Affiliation(s)
- Xiaoshu Zhan
- School of Animal Science and Technology, Foshan University, Foshan, Guangdong 528231, China; Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Nanshan Qi
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, China
| | - Derek Toms
- Myo palate Corporation, Toronto, ON M6R2B2, Canada
| | - Renee Freiburger
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Lauren Fletcher
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Bingyun Wang
- School of Animal Science and Technology, Foshan University, Foshan, Guangdong 528231, China.
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
8
|
Yao X, Wang W, He Y. Clinicopathological analysis of 22 Müllerian adenosarcomas and the sequencing of DICER1 mutation. Diagn Pathol 2024; 19:56. [PMID: 38570882 PMCID: PMC10988924 DOI: 10.1186/s13000-024-01477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Müllerian adenosarcoma, a rare malignancy, presents diagnostic and therapeutic challenges. In this study, we conducted an analysis of the clinicopathological characteristics of 22 adenosarcomas, with a particular focus on screening for DICER1 hot mutations. METHODS The cohort consisted of patients with adenosarcoma who were registered at the West China Second Hospital between the years 2020 and June 2022. Sanger sequencing was employed to screen for somatic Hotspot mutations in the RNase IIIb domain of DICER1 in the 22 adenosarcomas. RESULTS Only one patient exhibited a DICER1 mutation that was not a DICER1 Hotspot mutation. Among the 22 patients, all underwent total hysterectomy with bilateral salpingo-oophorectomy, and 14 out of these 22 patients received adjuvant treatment. CONCLUSION In summary, our study of 22 Müllerian adenosarcomas focused on the clinicopathological features and the presence of DICER1 Hotspot mutations. Although our findings did not reveal any DICER1 mutations in the studied samples, this negative result provides valuable information for the field by narrowing down the genetic landscape of adenosarcomas and highlighting the need for further research into alternative molecular pathways driving this malignancy.
Collapse
Affiliation(s)
- Xiaohong Yao
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Wang
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China
| | - Ying He
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Yu C, Qiu M, Yin H, Zhang Z, Hu C, Jiang X, Du H, Li Q, Li J, Xiong X, Yang C, Liu Y. miR-138-5p promotes chicken granulosa cell apoptosis via targeting SIRT1. Anim Biotechnol 2023; 34:2449-2458. [PMID: 35792779 DOI: 10.1080/10495398.2022.2095642] [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] [Indexed: 11/01/2022]
Abstract
Granulosa cell (GC) apoptosis is the main trigger of follicular atresia. MicroRNAs (miRNAs) are 18-22 nt RNAs whose function is primarily determined by their extended seed region and are considered to be involved in the biological functions of follicular development, including follicular atresia, folliculogenesis, and oogenesis. MiR-138-5p is known to act on chicken GCs. In this study, we found that miR-138-5p was enriched in reproductive organs, such as the uterus and ovaries. To examine whether miR-138-5p could regulate the biological process of GCs, miR-138-5p was examined by transfection of cells with a mimic or inhibitor of miR-138-5p. Expression levels of caspase-3 and caspase-9 mRNA and protein were markedly increased or decreased after transfection of the mimic or inhibitor, respectively. Furthermore, following miR-138-5p inhibition, SIRT1, one of the target genes of miR-138-5p, was found to increase the mRNA, which is correlated with the increased levels of BCL2 expression, an anti-apoptotic gene in the chicken GCs. These results suggest that miR-138-5p promotes apoptosis in chicken GCs by targeting SIRT1.
Collapse
Affiliation(s)
- Chunlin Yu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Mohan Qiu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zengrong Zhang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Chenming Hu
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Xiaosong Jiang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Huarui Du
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Qingyun Li
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Jingjing Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xia Xiong
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, Sichuan, China
| | - Yiping Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Chen R, Wu X, Qiu H, Yang B, Chen Y, Chen X, Li Y, Yuan S, Liu D, Xiao L, Yu Y. Obesity-induced inflammatory miR-133a mediates apoptosis of granulosa cells and causes abnormal folliculogenesis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1234-1246. [PMID: 37337633 PMCID: PMC10448043 DOI: 10.3724/abbs.2023089] [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: 11/18/2022] [Accepted: 02/15/2023] [Indexed: 06/21/2023] Open
Abstract
Obesity has been reported to promote disordered folliculogenesis, but the exact molecular mechanisms are still not fully understood. In this study, we find that miR-133a is involved in obesity-induced follicular development disorder. After feeding with a high-fat diet (HFD) and fructose water for nine weeks, the mouse body weight is significantly increased, accompanied by an inflammatory state and increased expression of miR-133a in the adipose tissues and ovaries as well as accelerated follicle depletion. Although miR-133a is increased in the fat and ovaries of HFD mice, the increased miR-133a in the HFD ovaries is not derived from exosome transferred from obese adipose tissues but is synthesized by ovarian follicular cells in response to HFD-induced inflammation. In vivo experiments show that intrabursal injection of miR-133a agomir induces a decrease in primordial follicles and an increase in antral follicles and atretic follicles, which is similar to HFD-induced abnormal folliculogenesis. Overexpression of miR-133a modestly promotes granulosa cell apoptosis by balancing the expression of anti-apoptotic proteins such as C1QL1 and XIAP and pro-apoptotic proteins such as PTEN. Overall, this study reveals the function of miR-133a in obesity-induced ovarian folliculogenesis dysfunction and sheds light on the etiology of female reproductive disorders.
Collapse
Affiliation(s)
- Ruizhi Chen
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Xueqing Wu
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Han Qiu
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Baiming Yang
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Yao Chen
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Xiang Chen
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Yingshan Li
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Shaochun Yuan
- Guangdong Province Key Laboratory of Pharmaceutical Functional GenesCollege of Life SciencesSun Yat-Sen UniversityGuangzhou510275China
| | - Dan Liu
- Department of Women’s HealthCareAffiliated Foshan Women and Children’s HospitalSouthern Medical UniversityFoshan528000China
| | - Luanjuan Xiao
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| | - Yanhong Yu
- Key Laboratory of Regenerative Medicine (JNU-CUHK)Ministry of EducationDepartment of Developmental and Regenerative BiologyCollege of Life Science and TechnologyJinan UniversityGuangzhou510632China
| |
Collapse
|
11
|
Nouri N, Shareghi-Oskoue O, Aghebati-Maleki L, Danaii S, Ahmadian Heris J, Soltani-Zangbar MS, Kamrani A, Yousefi M. Role of miRNAs interference on ovarian functions and premature ovarian failure. Cell Commun Signal 2022; 20:198. [PMID: 36564840 PMCID: PMC9783981 DOI: 10.1186/s12964-022-00992-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/22/2022] [Indexed: 12/24/2022] Open
Abstract
Premature ovarian failure is a to some extent unknown and intricate problem with diverse causes and clinical manifestations. The lack of ovarian sex hormones presumably is effective in the occurrence of ovarian failure. Our progress in this field has been very little despite undertaken scientific research endeavors; scholars still are trying to understand the explanation of this dilemmatic medical condition. In contrast, the practice of clinical medicine has made meaningful strides in providing assurance to the women with premature ovarian insufficiency that their quality of life as well as long-term health can be optimized through timely intervention. Very recently Scientists have investigated the regulating effects of small RNA molecules on steroidogenesis apoptosis, ovulation, gonadal, and corpus luteum development of ovaries. In this literature review, we tried to talk over the mechanisms of miRNAs in regulating gene expression after transcription in the ovary. Video abstract.
Collapse
Affiliation(s)
- Narjes Nouri
- grid.412888.f0000 0001 2174 8913Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, PO Box: 516-6615573, Tabriz, Iran
| | - Olduz Shareghi-Oskoue
- grid.412888.f0000 0001 2174 8913Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, PO Box: 516-6615573, Tabriz, Iran
| | - Leili Aghebati-Maleki
- grid.412888.f0000 0001 2174 8913Immunology Research Center, Tabriz University of Medical Sciences, PO Box: 516-6615573, Tabriz, Iran
| | - Shahla Danaii
- Gynecology Department, Eastern Azerbaijan ACECR ART Center, Eastern Azerbaijan Branch of ACECR, Tabriz, Iran
| | - Javad Ahmadian Heris
- grid.412888.f0000 0001 2174 8913Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sadegh Soltani-Zangbar
- grid.412888.f0000 0001 2174 8913Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, PO Box: 516-6615573, Tabriz, Iran
| | - Amin Kamrani
- grid.412888.f0000 0001 2174 8913Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, PO Box: 516-6615573, Tabriz, Iran
| | - Mehdi Yousefi
- grid.412888.f0000 0001 2174 8913Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, PO Box: 516-6615573, Tabriz, Iran ,grid.412888.f0000 0001 2174 8913Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Vitale SG, Fulghesu AM, Mikuš M, Watrowski R, D’Alterio MN, Lin LT, Shah M, Reyes-Muñoz E, Sathyapalan T, Angioni S. The Translational Role of miRNA in Polycystic Ovary Syndrome: From Bench to Bedside—A Systematic Literature Review. Biomedicines 2022; 10:biomedicines10081816. [PMID: 36009364 PMCID: PMC9405312 DOI: 10.3390/biomedicines10081816] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNAs that are essential for the regulation of post-transcriptional gene expression during tissue development and differentiation. They are involved in the regulation of manifold metabolic and hormonal processes and, within the female reproductive tract, in oocyte maturation and folliculogenesis. Altered miRNA levels have been observed in oncological and inflammatory diseases, diabetes or polycystic ovary syndrome (PCOS). Therefore, miRNAs are proving to be promising potential biomarkers. In women with PCOS, circulating miRNAs can be obtained from whole blood, serum, plasma, urine, and follicular fluid. Our systematic review summarizes data from 2010–2021 on miRNA expression in granulosa and theca cells; the relationship between miRNAs, hormonal changes, glucose and lipid metabolism in women with PCOS; and the potential role of altered miRNAs in fertility (oocyte quality) in PCOS. Furthermore, we discuss miRNAs as a potential therapeutic target in PCOS and as a diagnostic marker for PCOS.
Collapse
Affiliation(s)
- Salvatore Giovanni Vitale
- Obstetrics and Gynecology Unit, Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy;
| | - Anna Maria Fulghesu
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy; (A.M.F.); (M.N.D.)
| | - Mislav Mikuš
- Department of Obstetrics and Gynecology, University Hospital Centre Zagreb, 10 000 Zagreb, Croatia;
| | - Rafał Watrowski
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
| | - Maurizio Nicola D’Alterio
- Division of Gynecology and Obstetrics, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy; (A.M.F.); (M.N.D.)
| | - Li-Te Lin
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City 81362, Taiwan;
- Department of Obstetrics and Gynecology, School of Medicine, National Yang-Ming University, Pei-Tou, Taipei 112, Taiwan
- Department of Biological Science, National Sun Yat-sen University, 70 Lienhai Rd., Kaohsiung City 80424, Taiwan
| | - Mohsin Shah
- Department of Physiology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan;
| | - Enrique Reyes-Muñoz
- Department of Gynecological and Perinatal Endocrinology, Instituto Nacional de Perinatología, Mexico City 11000, Mexico;
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Kingston upon Hull HU6 7RX, UK;
| | - Stefano Angioni
- Obstetrics and Gynecology Unit, Department of General Surgery and Medical Surgical Specialties, University of Catania, 95124 Catania, Italy;
- Correspondence:
| |
Collapse
|
13
|
Fu TY, Wang SH, Lin TY, Shen PC, Chang SC, Lin YH, Chou CJ, Yu YH, Yang KT, Huang CW, Shaw SW, Peng SY. The Exploration of miRNAs From Porcine Fallopian Tube Stem Cells on Porcine Oocytes. Front Vet Sci 2022; 9:869217. [PMID: 35615247 PMCID: PMC9125035 DOI: 10.3389/fvets.2022.869217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022] Open
Abstract
Fallopian tube is essential to fertilization and embryonic development. Extracellular vesicles (EVs) from Fallopian tube containing biological regulatory factors, such as lipids, proteins and microRNAs (miRNAs) serve as the key role. At present, studies on oocytes from porcine oviduct and components from EVs remain limited. We aim to explore the effect of EVs secreted by porcine fallopian tube stem cells (PFTSCs) on oocyte. When the fifth-generation PFTSCs reached 80-90% of confluency, the pig in vitro maturation medium was utilized, and the conditioned medium collected for oocyte incubations. To realize the functions of EVs, several proteins were used to determine whether extracted EVs were cell-free. Field emission scanning electron microscope and nanoparticle tracking analyzer were used to observe the morphology. By next generation sequencing, 267 miRNAs were identified, and those with higher expression were selected to analyze the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment maps. The selected miR-152-3p, miR-148a-3p, miR-320a-3p, let-7f-5p, and miR-22-3p, were predicted to target Cepb1 gene affecting MAPK pathway. Of the five miRNAs, miR-320a-3p showed significant difference in maturation rate in vitro maturation. The blastocyst rate of pig embryos was also significantly enhanced by adding 50 nM miR-320a-3p. In vitro culture with miR-320a-3p, the blastocyst rate was significantly higher, but the cleavage rate and cell numbers were not. The CM of PFTSCs effectively improves porcine oocyte development. The miRNAs in EVs are sequenced and identified. miR-320a-3p not only helps the maturation, but also increases the blastocyst rates.
Collapse
Affiliation(s)
- Tzu-Yen Fu
- Department of Animal Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Shu-Hsuan Wang
- Department of Animal Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Tzu-Yi Lin
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Perng-Chih Shen
- Department of Animal Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Shen-Chang Chang
- Kaohsiung Animal Propagation Station, Livestock Research Institute, Council of Agriculture, Executive Yuan, Pingtung, Taiwan
| | - Yu-Han Lin
- Department of Animal Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Chih-Jen Chou
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsiang Yu
- Department of Biotechnology and Animal Science, National Ilan University, Yilan, Taiwan
| | - Kuo-Tai Yang
- Department of Animal Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Chao-Wei Huang
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Steven W. Shaw
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan
- Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Shao-Yu Peng
- Department of Animal Science, National Pingtung University of Science and Technology, Pingtung, Taiwan
| |
Collapse
|
14
|
Załęcka J, Pankiewicz K, Issat T, Laudański P. Molecular Mechanisms Underlying the Association between Endometriosis and Ectopic Pregnancy. Int J Mol Sci 2022; 23:ijms23073490. [PMID: 35408850 PMCID: PMC8998627 DOI: 10.3390/ijms23073490] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
Endometriosis is a common inflammatory disease characterized by the presence of endometrial cells outside the uterine cavity. It is estimated that it affects 10% of women of reproductive age. Its pathogenesis covers a wide range of abnormalities, including adhesion, proliferation, and cell signaling disturbances. It is associated with a significant deterioration in quality of life as a result of chronic pelvic pain and may also lead to infertility. One of the most serious complications of endometriosis is an ectopic pregnancy (EP). Currently, the exact mechanism explaining this phenomenon is unknown; therefore, there are no effective methods of prevention. It is assumed that the pathogenesis of EP is influenced by abnormalities in the contraction of the fallopian tube muscles, the mobility of the cilia, and in the fallopian microenvironment. Endometriosis can disrupt function on all three levels and thus contribute to the implantation of the embryo beyond the physiological site. This review takes into account aspects of the molecular mechanisms involved in the pathophysiology of endometriosis and EP, with particular emphasis on the similarities between them.
Collapse
Affiliation(s)
- Julia Załęcka
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland;
| | - Katarzyna Pankiewicz
- Department of Obstetrics and Gynecology, Institute of Mother and Child in Warsaw, Kasprzaka 17a, 01-211 Warsaw, Poland; (K.P.); (T.I.)
| | - Tadeusz Issat
- Department of Obstetrics and Gynecology, Institute of Mother and Child in Warsaw, Kasprzaka 17a, 01-211 Warsaw, Poland; (K.P.); (T.I.)
| | - Piotr Laudański
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1/3, 02-015 Warsaw, Poland;
- OVIklinika Infertility Center, Połczyńska 31, 01-377 Warsaw, Poland
- Correspondence:
| |
Collapse
|
15
|
Chemerinski A, Liu C, Morelli SS, Babwah AV, Douglas NC. Mouse Cre drivers: tools for studying disorders of the human female neuroendocrine-reproductive axis†. Biol Reprod 2022; 106:835-853. [PMID: 35084017 PMCID: PMC9113446 DOI: 10.1093/biolre/ioac012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 12/14/2021] [Accepted: 01/17/2022] [Indexed: 01/29/2023] Open
Abstract
Benign disorders of the human female reproductive system, such primary ovarian insufficiency and polycystic ovary syndrome are associated with infertility and recurrent miscarriage, as well as increased risk of adverse health outcomes, including cardiovascular disease and type 2 diabetes. For many of these conditions, the contributing molecular and cellular processes are poorly understood. The overarching similarities between mice and humans have rendered mouse models irreplaceable in understanding normal physiology and elucidating pathological processes that underlie disorders of the female reproductive system. The utilization of Cre-LoxP recombination technology, which allows for spatial and temporal control of gene expression, has identified the role of numerous genes in development of the female reproductive system and in processes, such as ovulation and endometrial decidualization, that are required for the establishment and maintenance of pregnancy in mammals. In this comprehensive review, we provide a detailed overview of Cre drivers with activity in the neuroendocrine-reproductive axis that have been used to study disruptions in key intracellular signaling pathways. We first summarize normal development of the hypothalamus, pituitary, ovary, and uterus, highlighting similarities and differences between mice and humans. We then describe human conditions resulting from abnormal development and/or function of the organ. Finally, we describe loss-of-function models for each Cre driver that elegantly recapitulate some key features of the human condition and are associated with impaired fertility. The examples we provide illustrate use of each Cre driver as a tool for elucidating genetic and molecular underpinnings of reproductive dysfunction.
Collapse
Affiliation(s)
- Anat Chemerinski
- Correspondence: Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB E561, Newark, NJ 07103, USA. Tel: 301-910-6800; Fax: 973-972-4574. E-mail:
| | | | - Sara S Morelli
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | | | | |
Collapse
|
16
|
McIlwraith EK, Zhang N, Belsham DD. The Regulation of Phoenixin: A Fascinating Multidimensional Peptide. J Endocr Soc 2021; 6:bvab192. [PMID: 35059547 PMCID: PMC8763610 DOI: 10.1210/jendso/bvab192] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Indexed: 02/07/2023] Open
Abstract
The phoenixin (PNX) peptide is linked to the control of reproduction, food intake, stress, and inflammation. However, little is known about what regulates its gene and protein expression, information that is critical to understand the physiological role of PNX. In this review, we summarize what is known about the transcriptional control of Pnx and its receptor Gpr173. A main function of PNX is as a positive regulator of the hypothalamic-pituitary-gonadal axis, but there is a lack of research on its control by reproductive hormones and peptides. PNX is also associated with food intake, and its expression is linked to feeding status, fatty acids, and glucose. It is influenced by environmental and hormonal-induced stress. The regulation of Pnx in most contexts remains an enigma, in part due to conflicting and negative results. An extensive analysis of the response of the Pnx gene to factors related to reproduction, metabolism, stress, and inflammation is required. Analysis of the Pnx promoter and epigenetic regulation must be considered to understand how this level of control contributes to its pleiotropic effects. PNX is now linked to a broad range of functions, but more research on its gene regulation is required to understand its place in overall physiology and therapeutic potential.
Collapse
Affiliation(s)
| | - Ningtong Zhang
- Department of Physiology, University of Toronto, ON, Canada
| | - Denise D Belsham
- Department of Physiology, University of Toronto, ON, Canada
- Department of Medicine, University of Toronto, ON, Canada
- Department of Obstetrics and Gynaecology, University of Toronto, ON, Canada
| |
Collapse
|
17
|
Ramakrishna NB, Murison K, Miska EA, Leitch HG. Epigenetic Regulation during Primordial Germ Cell Development and Differentiation. Sex Dev 2021; 15:411-431. [PMID: 34847550 DOI: 10.1159/000520412] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/10/2021] [Indexed: 11/19/2022] Open
Abstract
Germline development varies significantly across metazoans. However, mammalian primordial germ cell (PGC) development has key conserved landmarks, including a critical period of epigenetic reprogramming that precedes sex-specific differentiation and gametogenesis. Epigenetic alterations in the germline are of unique importance due to their potential to impact the next generation. Therefore, regulation of, and by, the non-coding genome is of utmost importance during these epigenomic events. Here, we detail the key chromatin changes that occur during mammalian PGC development and how these interact with the expression of non-coding RNAs alongside broader epitranscriptomic changes. We identify gaps in our current knowledge, in particular regarding epigenetic regulation in the human germline, and we highlight important areas of future research.
Collapse
Affiliation(s)
- Navin B Ramakrishna
- Wellcome/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Genome Institute of Singapore, A*STAR, Biopolis, Singapore, Singapore
| | - Keir Murison
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
| | - Eric A Miska
- Wellcome/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Harry G Leitch
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Burgos M, Hurtado A, Jiménez R, Barrionuevo FJ. Non-Coding RNAs: lncRNAs, miRNAs, and piRNAs in Sexual Development. Sex Dev 2021; 15:335-350. [PMID: 34614501 DOI: 10.1159/000519237] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/09/2021] [Indexed: 11/19/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are a group of RNAs that do not encode functional proteins, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), PIWI-interacting RNAs (piRNAs), and short interfering RNAs (siRNAs). In the last 2 decades an effort has been made to uncover the role of ncRNAs during development and disease, and nowadays it is clear that these molecules have a regulatory function in many of the developmental and physiological processes where they have been studied. In this review, we provide an overview of the role of ncRNAs during gonad determination and development, focusing mainly on mammals, although we also provide information from other species, in particular when there is not much information on the function of particular types of ncRNAs during mammalian sexual development.
Collapse
Affiliation(s)
- Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Alicia Hurtado
- Epigenetics and Sex Development Group, Berlin Institute for Medical Systems Biology, Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Rafael Jiménez
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Francisco J Barrionuevo
- Departamento de Genética e Instituto de Biotecnología, Lab. 127, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| |
Collapse
|
19
|
Ben Maamar M, Nilsson EE, Skinner MK. Epigenetic transgenerational inheritance, gametogenesis and germline development†. Biol Reprod 2021; 105:570-592. [PMID: 33929020 PMCID: PMC8444706 DOI: 10.1093/biolre/ioab085] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
One of the most important developing cell types in any biological system is the gamete (sperm and egg). The transmission of phenotypes and optimally adapted physiology to subsequent generations is in large part controlled by gametogenesis. In contrast to genetics, the environment actively regulates epigenetics to impact the physiology and phenotype of cellular and biological systems. The integration of epigenetics and genetics is critical for all developmental biology systems at the cellular and organism level. The current review is focused on the role of epigenetics during gametogenesis for both the spermatogenesis system in the male and oogenesis system in the female. The developmental stages from the initial primordial germ cell through gametogenesis to the mature sperm and egg are presented. How environmental factors can influence the epigenetics of gametogenesis to impact the epigenetic transgenerational inheritance of phenotypic and physiological change in subsequent generations is reviewed.
Collapse
Affiliation(s)
- Millissia Ben Maamar
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Eric E Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
20
|
Stener-Victorin E, Deng Q. Epigenetic inheritance of polycystic ovary syndrome - challenges and opportunities for treatment. Nat Rev Endocrinol 2021; 17:521-533. [PMID: 34234312 DOI: 10.1038/s41574-021-00517-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is the main cause of female infertility worldwide and is associated with a substantially increased lifetime risk of comorbidities, including type 2 diabetes mellitus, psychiatric disorders and gynaecological cancers. Despite its high prevalence (~15%) and substantial economic burden, the aetiology of PCOS remains elusive. The genetic loci linked to PCOS so far account for only ~10% of its heritability, which is estimated at 70%. However, growing evidence suggests that altered epigenetic and developmental programming resulting from hormonal dysregulation of the maternal uterine environment contributes to the pathogenesis of PCOS. Male as well as female relatives of women with PCOS are also at an increased risk of developing PCOS-associated reproductive and metabolic disorders. Although PCOS phenotypes are highly heterogenous, hyperandrogenism is thought to be the principal driver of this condition. Current treatments for PCOS are suboptimal as they can only alleviate some of the symptoms; preventative and targeted treatments are sorely needed. This Review presents an overview of the current understanding of the aetiology of PCOS and focuses on the developmental origin and epigenetic inheritance of this syndrome.
Collapse
Affiliation(s)
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
21
|
Huo S, Qi H, Si Y, Li C, Du W. MicroRNA 26a targets Ezh2 to regulate apoptosis in mouse ovarian granulosa cells. Syst Biol Reprod Med 2021; 67:221-229. [PMID: 34058933 DOI: 10.1080/19396368.2021.1895362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In the mammalian ovary, <1% of the follicles ovulate, with most undergoing degenerative atresia during ovarian follicular development. Follicular atresia is caused by the apoptosis of granulosa cells (GCs), although the precise underpinning mechanism remains unidentified. MiR-26a regulates various cellular events, including cell division, apoptotic signaling, and cell differentiation, migration, and autophagy. Here, we demonstrated that miR-26a regulated apoptosis in GCs in the mouse ovary through Ezh2, a key regulator of GC viability. We also found that transcription of miR-26a changed in response to an LH antagonist and a GnRH agonist. In addition, miR-26a transcription was downregulated following LH-induced transition of GCs to granulosa-lutein cells (GLCs). Dual-luciferase reporter assays confirmed Ezh2 as a miR-26a target. Exogenous expression in GCs of miR-26a mimics resulted in decreased Ezh2 expression, while miR-26a inhibition in GCs induced the opposite phenotype. Ezh2 silencing additionally reduced the anti-apoptotic effect of miR-26a inhibition in GCs. These data highlight the critical role of miR-26a in targeting Ezh2 and regulating apoptosis in mouse ovarian GCs.Abbreviations: GC: Granulosa cell; GLCs: Granulosa-lutein cells; LH: Luteinizing hormone; miRNA: MicroRNA; NC: Negative control; Cyt-c: Cytochrome c; GnRH: Gonadotropin releasing hormone; i.p.: intraperitoneal injection; cKO: conditional knock-out; WB: Western blotting; hCG: Human chorionic gonadotropin; NPC: nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Shiwei Huo
- Department of Reproductive Medicine, The Central Hospital of Taian, Taian, China
| | - Hongrong Qi
- Department of Reproductive Medicine, The Central Hospital of Taian, Taian, China
| | - Yuexiu Si
- Department of Respiration, The Fifth Hospital of Jinan, Jinan, China
| | - Changzhou Li
- Department of Reproductive Medicine, The Central Hospital of Taian, Taian, China
| | - Wenyan Du
- Department of Outpatient, The Branch of Taian Central Hospital, Taian, China
| |
Collapse
|
22
|
Santana Gonzalez L, Rota IA, Artibani M, Morotti M, Hu Z, Wietek N, Alsaadi A, Albukhari A, Sauka-Spengler T, Ahmed AA. Mechanistic Drivers of Müllerian Duct Development and Differentiation Into the Oviduct. Front Cell Dev Biol 2021; 9:605301. [PMID: 33763415 PMCID: PMC7982813 DOI: 10.3389/fcell.2021.605301] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
The conduits of life; the animal oviducts and human fallopian tubes are of paramount importance for reproduction in amniotes. They connect the ovary with the uterus and are essential for fertility. They provide the appropriate environment for gamete maintenance, fertilization and preimplantation embryonic development. However, serious pathologies, such as ectopic pregnancy, malignancy and severe infections, occur in the oviducts. They can have drastic effects on fertility, and some are life-threatening. Despite the crucial importance of the oviducts in life, relatively little is known about the molecular drivers underpinning the embryonic development of their precursor structures, the Müllerian ducts, and their successive differentiation and maturation. The Müllerian ducts are simple rudimentary tubes comprised of an epithelial lumen surrounded by a mesenchymal layer. They differentiate into most of the adult female reproductive tract (FRT). The earliest sign of Müllerian duct formation is the thickening of the anterior mesonephric coelomic epithelium to form a placode of two distinct progenitor cells. It is proposed that one subset of progenitor cells undergoes partial epithelial-mesenchymal transition (pEMT), differentiating into immature Müllerian luminal cells, and another subset undergoes complete EMT to become Müllerian mesenchymal cells. These cells invaginate and proliferate forming the Müllerian ducts. Subsequently, pEMT would be reversed to generate differentiated epithelial cells lining the fully formed Müllerian lumen. The anterior Müllerian epithelial cells further specialize into the oviduct epithelial subtypes. This review highlights the key established molecular and genetic determinants of the processes involved in Müllerian duct development and the differentiation of its upper segment into oviducts. Furthermore, an extensive genome-wide survey of mouse knockout lines displaying Müllerian or oviduct phenotypes was undertaken. In addition to widely established genetic determinants of Müllerian duct development, our search has identified surprising associations between loss-of-function of several genes and high-penetrance abnormalities in the Müllerian duct and/or oviducts. Remarkably, these associations have not been investigated in any detail. Finally, we discuss future directions for research on Müllerian duct development and oviducts.
Collapse
Affiliation(s)
- Laura Santana Gonzalez
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ioanna A Rota
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Developmental Immunology Research Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Mara Artibani
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matteo Morotti
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Zhiyuan Hu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Nina Wietek
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Abdulkhaliq Alsaadi
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Ashwag Albukhari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Tatjana Sauka-Spengler
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Ahmed A Ahmed
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Kim YS, Yang SC, Park M, Choi Y, DeMayo FJ, Lydon JP, Kim H, Lim HJ, Song H. Different Cre systems induce differential microRNA landscapes and abnormalities in the female reproductive tracts of Dgcr8 conditional knockout mice. Cell Prolif 2021; 54:e12996. [PMID: 33496365 PMCID: PMC7941225 DOI: 10.1111/cpr.12996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES The female reproductive tract comprises several different cell types. Using three representative Cre systems, we comparatively analysed the phenotypes of Dgcr8 conditional knockout (cKO) mice to understand the function of Dgcr8, involved in canonical microRNA biogenesis, in the female reproductive tract. MATERIALS AND METHODS Dgcr8f/f mice were crossed with Ltficre/+ , Amhr2cre/+ or PRcre/+ mice to produce mice deficient in Dgcr8 in epithelial (Dgcr8ed/ed ), mesenchymal (Dgcr8md/md ) and all the compartments (Dgcr8td/td ) in the female reproductive tract. Reproductive phenotypes were evaluated in Dgcr8 cKO mice. Uteri and/or oviducts were used for small RNA-seq, mRNA-seq, real-time RT-PCR, and/or morphologic and histological analyses. RESULT Dgcr8ed/ed mice did not exhibit any distinct defects, whereas Dgcr8md/md mice showed sub-fertility and oviductal smooth muscle deformities. Dgcr8td/td mice were infertile due to anovulation and acute inflammation in the female reproductive tract and suffered from an atrophic uterus with myometrial defects. The microRNAs and mRNAs related to immune modulation and/or smooth muscle growth were systemically altered in the Dgcr8td/td uterus. Expression profiles of dysregulated microRNAs and mRNAs in the Dgcr8td/td uterus were different from those in other genotypes in a Cre-dependent manner. CONCLUSIONS Dgcr8 deficiency with different Cre systems induces overlapping but distinct phenotypes as well as the profiles of microRNAs and their target mRNAs in the female reproductive tract, suggesting the importance of selecting the appropriate Cre driver to investigate the genes of interest.
Collapse
Affiliation(s)
- Yeon Sun Kim
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
- Present address:
Division of reproductive sciencesDepartment of PediatricsCincinnati Children’s HospitalOHUSA
| | | | - Mira Park
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
| | - Youngsok Choi
- Department of Stem Cell and Regenerative BiotechnologyKonkuk UniversitySeoulKorea
| | - Francesco J. DeMayo
- Department of Reproductive and Developmental Biology LaboratoryNational Institute of Environmental Health SciencesResearch Triangle ParkNCUSA
| | - John P. Lydon
- Department of Molecular and Cellular Biology and Center for Reproductive MedicineBaylor College of MedicineHoustonTXUSA
| | - Hye‐Ryun Kim
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
| | - Hyunjung Jade Lim
- Department of Veterinary Medicine, School of Veterinary MedicineKonkuk UniversitySeoulKorea
| | - Haengseok Song
- Department of Biomedical ScienceCHA UniversitySeongnamKorea
| |
Collapse
|
24
|
Mhatre P, Dighe V, Das DK, Pawar A. Novel Technique of Vaginoplasty Developing Normal Vagina, Role of Stemness Markers and Translational Genes. J Hum Reprod Sci 2021; 13:303-309. [PMID: 33627980 PMCID: PMC7879834 DOI: 10.4103/jhrs.jhrs_68_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/10/2020] [Accepted: 09/17/2020] [Indexed: 12/04/2022] Open
Abstract
Aims and Objectives: To study development of neo-vagina by metaplastic conversion of peritoneum, To identify translational Stemness markers using NANOG/OCT4/SOX2 from serial neo-vaginal mRNA, cDNA and to study role of WNT and HOXA genes in patients undergoing vaginoplasty. Material and Methods: 75 MRKH Syndrome women underwent laparoscopic peritoneal vaginoplasty (LPV). Two patients underwent serial neo-vaginal biopsies on day 0, 7-9, 12-14, 21 and 33. Fifteen MRKHS and twelve controls were subjected for neo-vaginal biopsy to detect genes upregulation. Remaining patients were evaluated anatomically and functionally. Results: The translational stemness markers NANOG, OCT4 and SOX2 responsible for neo-vaginal formation were identified. Their appearance, concentration at different stages of conversion were demonstrated. The neo-vagina has shown up-regulation of these translational stemness markers. The study demonstrates expression of the specific genes (WNT4, WNT5A and WNT7A) and their role in formation of the neo-vagina. In the subjects stemness markers (NANOG, OCT4 and SOX2) appeared from day 9 to 14 of the neo-vaginal biopsies and after achieving the peak declined later. Genetic analysis showed low values in HOXA 9,10,11,13 and up-regulation of WNT 4A,5A,7 genes in neo-vagina. Conclusions: Study shows peritoneal metaplastic conversion to normal vagina, identified the translational stemness markers and genes responsible. The neo-vagina has shown up-regulation of these genes. The study demonstrates expression of the specific genes (WNT4, WNT5A and WNT7A) and their role in formation of the neo-vagina. Furthering this research, activating these genes may lead to treatment of developmental defects of Mullerian duct, obviating the need of transplant.
Collapse
Affiliation(s)
- Pravin Mhatre
- Department of Obstetrics and Gynecology, Seth G S Medical College, KEM Hospital, N. Wadia Hospital, Parel, Mumbai, Maharashtra, India.,Department of Genetic, Kedar Hospital, Parel, Mumbai, Maharashtra, India
| | - Vikas Dighe
- Department of Reproductive and Genetic Toxicology, National Institute for Research in Reproductive Health, Parel, Mumbai, Maharashtra, India
| | - Dhanjit Kumar Das
- Department of Genetic Research Centre, National Institute for Research in Reproductive Health, Mumbai, Maharashtra, India
| | - Amol Pawar
- Department of Obstetrics and Gynecology, Seth G S Medical College, KEM Hospital, N. Wadia Hospital, Parel, Mumbai, Maharashtra, India
| |
Collapse
|
25
|
Gonella-Diaza AM, Lopes E, Ribeiro da Silva K, Perecin Nociti R, Mamede Andrade G, Atuesta-Bustos JE, Coelho da Silveira J, Vieira Meirelles F, Binelli M. Steroidal Regulation of Oviductal microRNAs Is Associated with microRNA-Processing in Beef Cows. Int J Mol Sci 2021; 22:953. [PMID: 33477993 PMCID: PMC7835783 DOI: 10.3390/ijms22020953] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/21/2022] Open
Abstract
Information on molecular mechanisms through which sex-steroids regulate oviductal function to support early embryo development is lacking. Here, we hypothesized that the periovulatory endocrine milieu affects the miRNA processing machinery and miRNA expression in bovine oviductal tissues. Growth of the preovulatory follicle was controlled to obtain cows that ovulated a small follicle (SF) and subsequently bore a small corpus luteum (CL; SF-SCL) or a large follicle (LF) and large CL (LF-LCL). These groups differed in the periovulatory plasmatic sex-steroid's concentrations. Ampulla and isthmus samples were collected on day four of the estrous cycle. Abundance of DROSHA, DICER1, and AGO4 transcripts was greater in the ampulla than the isthmus. In the ampulla, transcription of these genes was greater for the SF-SCL group, while the opposite was observed in the isthmus. The expression of the 88 most abundant miRNAs and 14 miRNAs in the ampulla and 34 miRNAs in isthmus were differentially expressed between LF-LCL and SF-SCL groups. Integration of transcriptomic and miRNA data and molecular pathways enrichment showed that important pathways were inhibited in the SF-SCL group due to miRNA control. In conclusion, the endocrine milieu affects the miRNA expression in the bovine oviduct in a region-specific manner.
Collapse
Affiliation(s)
- Angela Maria Gonella-Diaza
- North Florida Research and Education Center, Institute of Food and Agricultural Sciences, University of Florida, Marianna, FL 32446, USA;
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, 225, Avenida Duque de Caxias, Norte, Jardim, Elite, Pirassununga, SP 13635-900, Brazil; (E.L.); (K.R.d.S.)
| | - Everton Lopes
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, 225, Avenida Duque de Caxias, Norte, Jardim, Elite, Pirassununga, SP 13635-900, Brazil; (E.L.); (K.R.d.S.)
- Unianchieta, Av. Doutor Adoniro Ladeira, 94, (Km 55, 5 Rodovia Anhanguera), Jundiaí, SP 13210-795, Brazil
| | - Kauê Ribeiro da Silva
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, 225, Avenida Duque de Caxias, Norte, Jardim, Elite, Pirassununga, SP 13635-900, Brazil; (E.L.); (K.R.d.S.)
| | - Ricardo Perecin Nociti
- Department of Veterinary Medicine, College of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP 13635-900, Brazil; (R.P.N.); (G.M.A.); (J.C.d.S.); (F.V.M.)
| | - Gabriella Mamede Andrade
- Department of Veterinary Medicine, College of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP 13635-900, Brazil; (R.P.N.); (G.M.A.); (J.C.d.S.); (F.V.M.)
| | - Jorge Eduardo Atuesta-Bustos
- College of Agricultural Science—Agrarian University Foundation of Colombia-UNIAGRARIA, Calle 170 No 54a-10, Bogotá 111166, Colombia;
| | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, College of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP 13635-900, Brazil; (R.P.N.); (G.M.A.); (J.C.d.S.); (F.V.M.)
| | - Flávio Vieira Meirelles
- Department of Veterinary Medicine, College of Animal Sciences and Food Engineering, University of São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga, SP 13635-900, Brazil; (R.P.N.); (G.M.A.); (J.C.d.S.); (F.V.M.)
| | - Mario Binelli
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, 225, Avenida Duque de Caxias, Norte, Jardim, Elite, Pirassununga, SP 13635-900, Brazil; (E.L.); (K.R.d.S.)
- Department of Animal Sciences, University of Florida, PO Box 110910, Gainesville, FL 32611, USA
| |
Collapse
|
26
|
Kim O, Park EY, Kwon SY, Shin S, Emerson RE, Shin YH, DeMayo FJ, Lydon JP, Coffey DM, Hawkins SM, Quilliam LA, Cheon DJ, Fernández FM, Nephew KP, Karpf AR, Widschwendter M, Sood AK, Bast RC, Godwin AK, Miller KD, Cho CH, Kim J. Targeting progesterone signaling prevents metastatic ovarian cancer. Proc Natl Acad Sci U S A 2020; 117:31993-32004. [PMID: 33262282 PMCID: PMC7749341 DOI: 10.1073/pnas.2013595117] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (∼18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene.
Collapse
MESH Headings
- Adult
- Animals
- BRCA1 Protein/genetics
- Breast/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/prevention & control
- Cystadenocarcinoma, Serous/chemistry
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/prevention & control
- Disease Models, Animal
- Estradiol/administration & dosage
- Female
- Humans
- Mice
- Middle Aged
- Mifepristone/pharmacology
- Mifepristone/therapeutic use
- Mutation
- Neoplasms, Experimental/chemically induced
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
- Ovarian Neoplasms/chemically induced
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/prevention & control
- Ovary/pathology
- Ovary/surgery
- Progesterone/administration & dosage
- Progesterone/antagonists & inhibitors
- Progesterone/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Salpingo-oophorectomy
- Signal Transduction/drug effects
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Olga Kim
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Eun Young Park
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sun Young Kwon
- Department of Pathology, School of Medicine, Keimyung University, 41931 Daegu, Republic of Korea
| | - Sojin Shin
- Department of Obstetrics and Gynecology, School of Medicine, Keimyung University, 41931 Daegu, Republic of Korea
| | - Robert E Emerson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Yong-Hyun Shin
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
| | - John P Lydon
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030
| | - Donna M Coffey
- Department of Pathology and Genomic Medicine, Houston Methodist and Weill Cornell Medical College, Houston, TX 77030
| | - Shannon M Hawkins
- Department of Obstetrics and Gynecology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Lawrence A Quilliam
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Dong-Joo Cheon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332
| | - Kenneth P Nephew
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405
| | - Adam R Karpf
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Martin Widschwendter
- Department of Women's Cancer, Institute for Women's Health, University College London, WC1E 6AU London, United Kingdom
- Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020 Innsbruck, Austria
- European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060 Hall in Tirol, Austria
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- Department of Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Robert C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Kathy D Miller
- Department of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Chi-Heum Cho
- Department of Obstetrics and Gynecology, School of Medicine, Keimyung University, 41931 Daegu, Republic of Korea;
| | - Jaeyeon Kim
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202;
| |
Collapse
|
27
|
Alexandri C, Daniel A, Bruylants G, Demeestere I. The role of microRNAs in ovarian function and the transition toward novel therapeutic strategies in fertility preservation: from bench to future clinical application. Hum Reprod Update 2020; 26:174-196. [PMID: 32074269 DOI: 10.1093/humupd/dmz039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/02/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND New therapeutic approaches in oncology have converted cancer from a certain death sentence to a chronic disease. However, there are still challenges to be overcome regarding the off-target toxicity of many of these treatments. Oncological therapies can lead to future infertility in women. Given this negative impact on long-term quality of life, fertility preservation is highly recommended. While gamete and ovarian tissue cryopreservation are the usual methods offered, new pharmacological-based options aiming to reduce ovarian damage during oncological treatment are very attractive. In this vein, advances in the field of transcriptomics and epigenomics have brought small noncoding RNAs, called microRNAs (miRNAs), into the spotlight in oncology. MicroRNAs also play a key role in follicle development as regulators of follicular growth, atresia and steroidogenesis. They are also involved in DNA damage repair responses and they can themselves be modulated during chemotherapy. For these reasons, miRNAs may be an interesting target to develop new protective therapies during oncological treatment. This review summarizes the physiological role of miRNAs in reproduction. Considering recently developed strategies based on miRNA therapy in oncology, we highlight their potential interest as a target in fertility preservation and propose future strategies to make the transition from bench to clinic. OBJECTIVE AND RATIONALE How can miRNA therapeutic approaches be used to develop new adjuvant protective therapies to reduce the ovarian damage caused by cytotoxic oncological treatments? SEARCH METHODS A systematic search of English language literature using PubMed and Google Scholar databases was performed through to 2019 describing the role of miRNAs in the ovary and their use for diagnosis and targeted therapy in oncology. Personal data illustrate miRNA therapeutic strategies to target the gonads and reduce chemotherapy-induced follicular damage. OUTCOMES This review outlines the importance of miRNAs as gene regulators and emphasizes the fact that insights in oncology can inspire new adjuvant strategies in the field of onco-fertility. Recent improvements in nanotechnology offer the opportunity for drug development using next-generation miRNA-nanocarriers. WIDER IMPLICATIONS Although there are still some barriers regarding the immunogenicity and toxicity of these treatments and there is still room for improvement concerning the specific delivery of miRNAs into the ovaries, we believe that, in the future, miRNAs can be developed as powerful and non-invasive tools for fertility preservation.
Collapse
Affiliation(s)
- C Alexandri
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - A Daniel
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.,Université de Tours, Faculty of Science and Technology, 37200 Tours, France
| | - G Bruylants
- Engineering of Molecular NanoSystems, Ecole Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - I Demeestere
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.,Fertility Clinic, CUB-Erasme, 1070 Brussels, Belgium
| |
Collapse
|
28
|
Apellaniz-Ruiz M, McCluggage WG, Foulkes WD. DICER1-associated embryonal rhabdomyosarcoma and adenosarcoma of the gynecologic tract: Pathology, molecular genetics, and indications for molecular testing. Genes Chromosomes Cancer 2020; 60:217-233. [PMID: 33135284 DOI: 10.1002/gcc.22913] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
Gynecologic sarcomas are uncommon neoplasms, the majority occurring in the uterus. Due to the diverse nature of these, the description of "new" morphological types and the rarity of some of them, pathological diagnosis and treatment is often challenging. Finding genetic alterations specific to, and frequently occurring, in a certain type can aid in the diagnosis. DICER1 is a highly conserved ribonuclease crucial in the biogenesis of microRNAs and mutations in DICER1 (either somatic or germline) have been detected in a wide range of sarcomas including genitourinary embryonal rhabdomyosarcomas (ERMS) and adenosarcomas. Importantly, DICER1-associated sarcomas share morphological features irrespective of the site of origin such that the pathologist can strongly suspect a DICER1 association. A review of the literature shows that almost all gynecologic ERMS reported (outside of the vagina) harbor DICER1 alterations, while approximately 20% of adenosarcomas also do so. These two tumor types exhibit significant morphological overlap and DICER1 tumor testing may be helpful in distinguishing between them, because a negative result makes ERMS unlikely. Given that germline pathogenic DICER1 variants are frequent in uterine (corpus and cervix) ERMS and pathogenic germline variants in this gene cause a hereditary cancer predisposition syndrome (DICER1 syndrome), patients diagnosed with these neoplasms should be referred to medical genetic services. Cooperation between pathologists and geneticists is crucial and will help in improving the diagnosis and management of these uncommon sarcomas.
Collapse
Affiliation(s)
| | - W Glenn McCluggage
- Department of Pathology, Belfast Health and Social Care Trust, Belfast, UK
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
| |
Collapse
|
29
|
Dicing the Disease with Dicer: The Implications of Dicer Ribonuclease in Human Pathologies. Int J Mol Sci 2020; 21:ijms21197223. [PMID: 33007856 PMCID: PMC7583940 DOI: 10.3390/ijms21197223] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/27/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022] Open
Abstract
Gene expression dictates fundamental cellular processes and its de-regulation leads to pathological conditions. A key contributor to the fine-tuning of gene expression is Dicer, an RNA-binding protein (RBPs) that forms complexes and affects transcription by acting at the post-transcriptional level via the targeting of mRNAs by Dicer-produced small non-coding RNAs. This review aims to present the contribution of Dicer protein in a wide spectrum of human pathological conditions, including cancer, neurological, autoimmune, reproductive and cardiovascular diseases, as well as viral infections. Germline mutations of Dicer have been linked to Dicer1 syndrome, a rare genetic disorder that predisposes to the development of both benign and malignant tumors, but the exact correlation of Dicer protein expression within the different cancer types is unclear, and there are contradictions in the data. Downregulation of Dicer is related to Geographic atrophy (GA), a severe eye-disease that is a leading cause of blindness in industrialized countries, as well as to psychiatric and neurological diseases such as depression and Parkinson's disease, respectively. Both loss and upregulation of Dicer protein expression is implicated in severe autoimmune disorders, including psoriasis, ankylosing spondylitis, rheumatoid arthritis, multiple sclerosis and autoimmune thyroid diseases. Loss of Dicer contributes to cardiovascular diseases and causes defective germ cell differentiation and reproductive system abnormalities in both sexes. Dicer can also act as a strong antiviral with a crucial role in RNA-based antiviral immunity. In conclusion, Dicer is an essential enzyme for the maintenance of physiology due to its pivotal role in several cellular processes, and its loss or aberrant expression contributes to the development of severe human diseases. Further exploitation is required for the development of novel, more effective Dicer-based diagnostic and therapeutic strategies, with the goal of new clinical benefits and better quality of life for patients.
Collapse
|
30
|
He R, Zhao Z, Yang Y, Liang X. Using bioinformatics and metabolomics to identify altered granulosa cells in patients with diminished ovarian reserve. PeerJ 2020; 8:e9812. [PMID: 32923184 PMCID: PMC7457930 DOI: 10.7717/peerj.9812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 08/04/2020] [Indexed: 11/20/2022] Open
Abstract
Background During fertility treatment, diminished ovarian reserve (DOR) is a challenge that can seriously affect a patient's reproductive potential. However, the pathogenesis of DOR is still unclear and its treatment options are limited. This study aimed to explore DOR's molecular mechanisms. Methods We used R software to analyze the mRNA microarray dataset E-MTAB-391 downloaded from ArrayExpress, screen for differentially expressed genes (DEGs), and perform functional enrichment analyses. We also constructed the protein-protein interaction (PPI) and miRNA-mRNA networks. Ovarian granulosa cells (GCs) from women with DOR and the control group were collected to perform untargeted metabolomics analyses. Additionally, small molecule drugs were identified using the Connectivity Map database. Results We ultimately identified 138 DEGs. Our gene ontology (GO) analysis indicated that DEGs were mainly enriched in cytokine and steroid biosynthetic processes. According to the Kyoto Encyclopedia of Genes and Genomes (KEGG), the DEGs were mainly enriched in the AGE (advanced glycation end-product)-RAGE (receptor for AGE) signaling pathway in diabetic complications and steroid biosynthesis. In the PPI network, we determined that JUN, EGR1, HMGCR, ATF3, and SQLE were hub genes that may be involved in steroid biosynthesis and inflammation. miRNAs also played a role in DOR development by regulating target genes. We validated the differences in steroid metabolism across GCs using liquid chromatography-tandem mass spectrometry (LC-MS/MS). We selected 31 small molecules with potentially positive or negative influences on DOR development. Conclusion We found that steroidogenesis and inflammation played critical roles in DOR development, and our results provide promising insights for predicting and treating DOR.
Collapse
Affiliation(s)
- Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Zhongying Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| |
Collapse
|
31
|
Li Y, Xiang Y, Song Y, Wan L, Yu G, Tan L. Dysregulated miR-142, -33b and -423 in granulosa cells target TGFBR1 and SMAD7: a possible role in polycystic ovary syndrome. Mol Hum Reprod 2020; 25:638-646. [PMID: 30865275 DOI: 10.1093/molehr/gaz014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/10/2019] [Accepted: 03/03/2019] [Indexed: 12/15/2022] Open
Abstract
It is well established that microRNA (miRNA) expression profiles are altered in patients with polycystic ovary syndrome (PCOS). In addition, abnormal transforming growth factor beta (TGFB) signaling in granulosa cells is related to the pathological conditions of PCOS. However, the function of dysregulated miRNAs in PCOS is still unclear. In this study, we aimed to elucidate the roles of specific miRNAs in PCOS. We collected follicular fluid from 46 patients with PCOS and 32 healthy controls. Granulosa cells (GCs) were separated and the levels of six candidate miRNAs were determined by quantitative RT-PCR. The direct targets of three dysregulated miRNAs were predicted using bioinformatic tools and confirmed using a dual luciferase assay and immunoblotting. The biological function of three dysregulated miRNAs in primary GCs was determined using a cell proliferation assay and flow cytometry. We found that miR-423 expression was downregulated (P = 0.038), and the levels of miR-33b (P = 0.032) and miR-142 (P = 0.021) were upregulated in GCs from patients with PCOS, compared to controls. miR-423 directly repressed SMAD family member 7 (SMAD7) expression, while transforming growth factor beta receptor 1 (TGFBR1) was a direct target of both miR-33b and miR-142. An RNA oligonucleotide mixture containing miR-423 inhibitor, miR-33b mimic, and miR-142 mimic repressed TGFB signaling, promoted cell proliferation (P = 0.0098), repressed apoptosis (P = 0.027), and increased S phase cell numbers (P = 0.0036) in primary cultures of GCs, compared to the cells treated with a sequence scrambled control RNA oligonucleotide. This study unveiled the possible roles of three miRNAs in PCOS and might provide candidate biomarkers for PCOS diagnosis while in vivo functional studies, using transgenic or knockout mouse models, are expected to confirm the roles of dysregulated miRNAs in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Yan Li
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yungai Xiang
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuxia Song
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lijing Wan
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guo Yu
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Tan
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
32
|
Aboutalebi H, Bahrami A, Soleimani A, Saeedi N, Rahmani F, Khazaei M, Fiuji H, Shafiee M, Ferns GA, Avan A, Hassanian SM. The diagnostic, prognostic and therapeutic potential of circulating microRNAs in ovarian cancer. Int J Biochem Cell Biol 2020; 124:105765. [PMID: 32428568 DOI: 10.1016/j.biocel.2020.105765] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/06/2020] [Accepted: 05/03/2020] [Indexed: 12/11/2022]
Abstract
Ovarian cancer (OC) is often diagnosed at an advanced stage because of the late onset of symptoms, and this together with the lack of effective treatments, has meant it is associated with a very high mortality. The aberrant expression of MicroRNA (miRNA) contributes to the initiation and development of human tumors including OC. Several miRNAs are secreted by tumor cells and can be identified in body fluids. Serum miRNAs levels are associated with several clinical conditions, and may be used to predict prognosis and response to treatments in some cancers including OC. This review summarizes the current progresses regarding the potential applications of circulating miRNA as innovative biomarkers in OC.
Collapse
Affiliation(s)
- Hamideh Aboutalebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Atena Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nikoo Saeedi
- Student Research Committee, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Farzad Rahmani
- Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Fiuji
- Department of Biochemistry, Payame-Noor University, Mashhad, Iran
| | - Mojtaba Shafiee
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK.
| |
Collapse
|
33
|
Xie S, Zhang Q, Zhao J, Hao J, Fu J, Li Y. MiR-423-5p may regulate ovarian response to ovulation induction via CSF1. Reprod Biol Endocrinol 2020; 18:26. [PMID: 32264887 PMCID: PMC7137414 DOI: 10.1186/s12958-020-00585-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 03/27/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND We have previously shown that hsa-miR-423-5p expression in ovarian granulosa cells is decreased in high ovarian response populations. The objective of the present study was to find the target gene and mechanism for miR-423-5p involved in ovarian response regulation. METHODS (a) TargetScan was used to predict the target gene of hsa-miR-423-5p. (b) A model for hsa-miR-423-5p overexpression or inhibition was constructed by transfecting KGN cells with lentivirus. CSF1 mRNA and protein expression and luciferase activity were measured. (c) The cell cycles of control and lentivirus treated KGN cells were analyzed. Western blot was used to measure the expression of CDKN1A in KGN cells. (d) The concentration of E2 in KGN cell culture medium were measured. RESULTS (a) TargetScan revealed that the 3' un-translated region of CSF1 matched 11 bases at the 5' end of miR-423-5p, making it a likely target gene. (b) Overexpression or inhibition of miR-423-5p were associated with respective decreases or increases in CSF1 expression (both mRNA and protein) (p < 0.05) and luciferase activity (p < 0.05). (c) When miR-423-5p expression increased, the number of G0/G1 phase cells and the expression of CDKN1A protein increased while estradiol concentrations in the cell culture solution decreased (p < 0.05). However, when miR-423-5p expression decreased, the number of S phase cells increased and E2 concentrations increased while the expression of CDKN1A protein decreased (p < 0.05). CONCLUSIONS Colony stimulating factor 1 is a target gene of miR-423-5p and that it may regulate ovarian response to ovulation induction by affecting granulosa cells proliferation and estrogen secretion.
Collapse
Affiliation(s)
- Shi Xie
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Clinical Research Center For Women's Reproductive Health In Human Province, Changsha, Hunan, China
| | - Qiong Zhang
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Clinical Research Center For Women's Reproductive Health In Human Province, Changsha, Hunan, China
| | - Jing Zhao
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Clinical Research Center For Women's Reproductive Health In Human Province, Changsha, Hunan, China
| | - Jie Hao
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Clinical Research Center For Women's Reproductive Health In Human Province, Changsha, Hunan, China
| | - Jing Fu
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China
- Clinical Research Center For Women's Reproductive Health In Human Province, Changsha, Hunan, China
| | - Yanping Li
- Reproductive Medicine Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, China.
- Clinical Research Center For Women's Reproductive Health In Human Province, Changsha, Hunan, China.
| |
Collapse
|
34
|
Apellaniz‐Ruiz M, Cullinan N, Grant R, Marrano P, Priest JR, Thorner PS, Goudie C, Foulkes WD. DICER1 screening in 15 paediatric paratesticular sarcomas unveils an unusual DICER1-associated sarcoma. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2020; 6:185-194. [PMID: 32222066 PMCID: PMC7339209 DOI: 10.1002/cjp2.164] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
Individuals with DICER1 syndrome, a genetic disorder caused by pathogenic germline variants in DICER1, are at increased risk of developing a wide array of predominantly childhood onset conditions, including genitourinary sarcomas. However, data on DICER1 involvement in paratesticular sarcomas have not been published. Herein, we analyse a series of 15 paediatric paratesticular sarcomas and describe in detail the case of a male infant with a paratesticular myxoid tumour, considered to be a low‐grade sarcoma, who also manifested a cystic nephroma, a classic DICER1 syndrome phenotype. He harboured a pathogenic germline DICER1 variant and different somatic hot‐spot mutations in each tumour. The paratesticular tumour showed strong and diffuse expression for WT1 and CD10, an unusual immunophenotype in paediatric sarcomas, but typical of tumours of Müllerian origin. The tumour was postulated to arise from the appendix testis, a Müllerian remnant located in the paratestis. Such an origin would be analogous to other DICER1‐associated non‐epithelial gynaecological tumours, thought to arise from Müllerian derivatives. These findings point towards a key role of DICER1 in Müllerian‐derived structures. Supporting this hypothesis is the fact that the other paratesticular sarcomas from the series were either negative or focally positive for WT1 and for CD10, and none had any DICER1 mutations. In summary, we present the first case of a paratesticular sarcoma associated with DICER1 syndrome, emphasising that paratesticular tumours with an unusual histological appearance may suggest an underlying DICER1 mutation, especially in the presence of a personal or family history of DICER1‐associated disease. In this context, DICER1 mutation testing could lead to changes in clinical care including implementation of cancer care surveillance strategies.
Collapse
Affiliation(s)
- Maria Apellaniz‐Ruiz
- Department of Human GeneticsMcGill UniversityMontréalCanada
- Lady Davis Institute, Segal Cancer CentreJewish General Hospital, McGill UniversityMontréalCanada
| | - Noelle Cullinan
- Division of Hematology‐Oncology, The Hospital for Sick Children, and Department of PediatricsUniversity of TorontoTorontoCanada
| | - Ronald Grant
- Division of Hematology‐Oncology, The Hospital for Sick Children, and Department of PediatricsUniversity of TorontoTorontoCanada
| | - Paula Marrano
- Division of PathologyThe Hospital for Sick ChildrenTorontoCanada
| | | | - Paul S Thorner
- Division of PathologyThe Hospital for Sick ChildrenTorontoCanada
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoCanada
| | - Catherine Goudie
- Division of Hematology‐Oncology, Montréal Children's Hospital, Department of PediatricsMcGill UniversityMontréalCanada
| | - William D Foulkes
- Department of Human GeneticsMcGill UniversityMontréalCanada
- Lady Davis Institute, Segal Cancer CentreJewish General Hospital, McGill UniversityMontréalCanada
- Program in Cancer Genetics, Department of Oncology and Human GeneticsMcGill UniversityMontréalCanada
- Department of Medical GeneticsResearch Institute of the McGill University Health CentreMontréalCanada
| |
Collapse
|
35
|
Casarini L, Crépieux P, Reiter E, Lazzaretti C, Paradiso E, Rochira V, Brigante G, Santi D, Simoni M. FSH for the Treatment of Male Infertility. Int J Mol Sci 2020; 21:ijms21072270. [PMID: 32218314 PMCID: PMC7177393 DOI: 10.3390/ijms21072270] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022] Open
Abstract
Follicle-stimulating hormone (FSH) supports spermatogenesis acting via its receptor (FSHR), which activates trophic effects in gonadal Sertoli cells. These pathways are targeted by hormonal drugs used for clinical treatment of infertile men, mainly belonging to sub-groups defined as hypogonadotropic hypogonadism or idiopathic infertility. While, in the first case, fertility may be efficiently restored by specific treatments, such as pulsatile gonadotropin releasing hormone (GnRH) or choriogonadotropin (hCG) alone or in combination with FSH, less is known about the efficacy of FSH in supporting the treatment of male idiopathic infertility. This review focuses on the role of FSH in the clinical approach to male reproduction, addressing the state-of-the-art from the little data available and discussing the pharmacological evidence. New compounds, such as allosteric ligands, dually active, chimeric gonadotropins and immunoglobulins, may represent interesting avenues for future personalized, pharmacological approaches to male infertility.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-0593961705; Fax: +39-0593962018
| | - Pascale Crépieux
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
| | - Eric Reiter
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
| | - Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Vincenzo Rochira
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Giulia Brigante
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
36
|
Kaczmarek MM, Najmula J, Guzewska MM, Przygrodzka E. MiRNAs in the Peri-Implantation Period: Contribution to Embryo-Maternal Communication in Pigs. Int J Mol Sci 2020; 21:ijms21062229. [PMID: 32210170 PMCID: PMC7139304 DOI: 10.3390/ijms21062229] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) constitute a large family of noncoding RNAs, approximately 22 nucleotides long, which function as guide molecules in RNA silencing. Targeting most protein-coding transcripts, miRNAs are involved in nearly all developmental and pathophysiological processes in animals. To date, the regulatory roles of miRNAs in reproduction, such as fertilization, embryo development, implantation, and placenta formation, among others, have been demonstrated in numerous mammalian species, including domestic livestock such as pigs. Over the past years, it appeared that understanding the functions of miRNAs in mammalian reproduction can substantially improve our understanding of the biological challenges of successful reproductive performance. This review describes the current knowledge on miRNAs, specifically in relation to the peri-implantation period when the majority of embryonic mortality occurs in pigs. To present a broader picture of crucial peri-implantation events, we focus on the role of miRNA-processing machinery and miRNA–mRNA infarctions during the maternal recognition of pregnancy, leading to maintenance of the corpus luteum function and further embryo implantation. Furthermore, we summarize the current knowledge on cell-to-cell communication involving extracellular vesicles at the embryo–maternal interface in pigs. Finally, we discuss the potential of circulating miRNAs to serve as indicators of ongoing embryo–maternal crosstalk.
Collapse
Affiliation(s)
- Monika M. Kaczmarek
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (J.N.); (M.M.G.)
- Correspondence:
| | - Joanna Najmula
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (J.N.); (M.M.G.)
| | - Maria M. Guzewska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland; (J.N.); (M.M.G.)
| | | |
Collapse
|
37
|
Target prediction and validation of microRNAs expressed from FSHR and aromatase genes in human ovarian granulosa cells. Sci Rep 2020; 10:2300. [PMID: 32042028 PMCID: PMC7010774 DOI: 10.1038/s41598-020-59186-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are known post-transcriptional regulators of various biological processes including ovarian follicle development. We have previously identified miRNAs from human pre-ovulatory ovarian granulosa cells that are expressed from the intronic regions of two key genes in normal follicular development: FSH receptor (FSHR) and CYP19A1, the latter encoding the aromatase enzyme. The present study aims to identify the target genes regulated by these miRNAs: hsa-miR-548ba and hsa-miR-7973, respectively. The miRNAs of interest were transfected into KGN cell line and the gene expression changes were analyzed by Affymetrix microarray. Potential miRNA-regulated genes were further filtered by bioinformatic target prediction algorithms and validated for direct miRNA:mRNA binding by luciferase reporter assay. LIFR, PTEN, NEO1 and SP110 were confirmed as targets for hsa-miR-548ba. Hsa-miR-7973 target genes ADAM19, PXDN and FMNL3 also passed all verification steps. Additionally, the expression pattern of the miRNAs was studied in human primary cumulus granulosa cell culture in relation to the expression of their host genes and FSH stimulation. Based on our findings we propose the involvement of hsa-miR-548ba in the regulation of follicle growth and activation via LIFR and PTEN. Hsa-miR-7973 may be implicated in the modulation of extracellular matrix and cell-cell interactions by regulating the expression of its identified targets.
Collapse
|
38
|
Zhou X, He Y, Jiang Y, He B, Deng X, Zhang Z, Yuan X, Li J. MiR-126-3p inhibits apoptosis and promotes proliferation by targeting phosphatidylinositol 3-kinase regulatory subunit 2 in porcine ovarian granulosa cells. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 33:879-887. [PMID: 31480138 PMCID: PMC7206374 DOI: 10.5713/ajas.19.0290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022]
Abstract
Objective Numerous studies have indicated that the apoptosis and proliferation of granulosa cells (GCs) are closely related to the normal growth and development of follicles and ovaries. Previous evidence has suggested that miR-126-3p might get involved in the apoptosis and proliferation of GCs, and phosphatidylinositol 3-kinase regulatory subunit 2 (PIK3R2) gene has been predicted as one target of miR-126-3p. However, the molecular regulation of miR-126-3p on PIK3R2 and the effects of PIK3R2 on porcine GCs apoptosis and proliferation remain virtually unexplored. Methods In this study, using porcine GCs as a cellular model, luciferase report assay, mutation and deletion were applied to verify the targeting relationship between miR-126-3p and PIK3R2. Annexin-V/PI staining and 5-ethynyl-2′-deoxyuridine assay were applied to explore the effect of PIK3R2 on GCs apoptosis and proliferation, respectively. Real-time quantitative polymerase chain reaction and Western Blot were applied to explore the regulation of miR-126-3p on PIK3R2 expression. Results We found that miR-126-3p targeted at PIK3R2 and inhibited its mRNA and protein expression. Knockdown of PIK3R2 significantly inhibited the apoptosis and promoted the proliferation of porcine GCs, and significantly down-regulated the mRNA expression of several key genes of PI3K pathway such as insulin-like growth factor 1 receptor (IGF1R), insulin receptor (INSR), pyruvate dehydrogenase kinase 1 (PDK1), and serine/threonine kinase 1 (AKT1). Conclusion MiR-126-3p might target and inhibit the mRNA and protein expressions of PIK3R2, thereby inhibiting GC apoptosis and promoting GC proliferation by down-regulating several key genes of the PI3K pathway, IGF1R, INSR, PDK1, and AKT1. These findings would provide great insight into further exploring the molecular regulation of miR-126-3p and PIK3R2 on the functions of GCs during the folliculogenesis in female mammals.
Collapse
Affiliation(s)
- Xiaofeng Zhou
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yingting He
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yao Jiang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Bo He
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xi Deng
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhe Zhang
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiaolong Yuan
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jiaqi Li
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Centre for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| |
Collapse
|
39
|
Loke H, Rainczuk K, Dimitriadis E. MicroRNA Biogenesis Machinery Is Dysregulated in the Endometrium of Infertile Women Suggesting a Role in Receptivity and Infertility. J Histochem Cytochem 2019; 67:589-599. [PMID: 31145039 DOI: 10.1369/0022155419854064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRs) regulate endometrial function and their dysregulation could underlie unexplained infertility in women. Ribonucleases including DICER and DROSHA, and the proteins, ARGONAUTE 1 (AGO 1) and 2 (AGO 2) regulate the biogenesis/maturation of miRs. We aimed to elucidate the expression and localization of miR biogenesis machinery components during the human menstrual cycle and compare their levels in endometrium from women with normal fertility and primary unexplained infertility. miR biogenesis components were measured by quantitative-RT-PCR and immunohistochemistry. In the endometrium of women with normal fertility, DROSHA immunolocalized maximally to the epithelium during the early and mid-secretory phases compared with the proliferative and late-secretory phases. Stromal DICER immunostaining intensity was higher in the late-secretory phase compared with all other phases in fertile women. DROSHA mRNA was reduced in the mid-secretory-infertile whole endometrial tissue (has all cells of the tissue), and primary epithelial and stromal cells while no differences were found in DICER, AGO1, and AGO2 mRNA. In the luminal epithelium, DROSHA staining intensity was reduced in early and mid-secretory-infertile while DICER staining was reduced in the early secretory-infertile compared with their respective fertile groups. DICER and DROSHA were dynamically regulated across the menstrual cycle and reduced levels during receptivity phase could underlie implantation failure/infertility.
Collapse
Affiliation(s)
- Hannah Loke
- Embryo Implantation Laboratory, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Medicine, Monash University, Clayton, Victoria, Australia
| | - Kate Rainczuk
- Embryo Implantation Laboratory, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Medicine, Monash University, Clayton, Victoria, Australia
| | - Evdokia Dimitriadis
- Embryo Implantation Laboratory, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Medicine, Monash University, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, The University of Melbourne, The Royal Women's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
40
|
Robles V, Valcarce DG, Riesco MF. Non-coding RNA regulation in reproduction: Their potential use as biomarkers. Noncoding RNA Res 2019; 4:54-62. [PMID: 31193491 PMCID: PMC6531869 DOI: 10.1016/j.ncrna.2019.04.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are crucial regulatory elements in most biological processes and reproduction is also controlled by them. The different types of ncRNAs, as well as the high complexity of these regulatory pathways, present a complex scenario; however, recent studies have shed some light on these questions, discovering the regulatory function of specific ncRNAs on concrete reproductive biology processes. This mini review will focus on the role of ncRNAs in spermatogenesis and oogenesis, and their potential use as biomarkers for reproductive diseases or for reproduction success.
Collapse
Affiliation(s)
- Vanesa Robles
- Spanish Institute of Oceanography (IEO) Santander, Spain
- MODCELL GROUP, Department of Molecular Biology, Universidad de León, 24071, León, Spain
- Corresponding author. Planta de Cultivos el Bocal, IEO, Barrio Corbanera, Monte, Santander, 39012, Spain.
| | | | | |
Collapse
|
41
|
Lite C, Ahmed SSSJ, Santosh W, Seetharaman B. Prenatal exposure to bisphenol-A altered miRNA-224 and protein expression of aromatase in ovarian granulosa cells concomitant with elevated serum estradiol levels in F 1 adult offspring. J Biochem Mol Toxicol 2019; 33:e22317. [PMID: 30817060 DOI: 10.1002/jbt.22317] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/31/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022]
Abstract
This study was aimed to predict bisphenol-A (BPA)-responsive miRNA's using an in silico approach and to study their expression in granulosa cells of animals exposed prenatally to BPA. Pregnant Wistar rats were exposed to BPA through water (25 μg/L, 250 μg/L, and 2.5 mg/L) during gestation. The expression of miRNA-133b, miRNA-378 and miRNA-224 were analyzed in ovarian granulosa cells. BPA affected the postnatal developmental landmarks such as weight of the pups at birth and reduced anogenital distance. BPA exposed animals showed elevated serum estradiol (E2) levels, while follicle-stimulating hormone levels were reduced. The expression of miRNA-224 and aromatase protein levels were found to be increased. This preliminary finding reveals the impact of early life exposure to BPA on the long-term ovarian functions that may be mediated through miRNA-based granulosa cell response. Besides, it is also a compelling indicator for the subclinical response that could have important consequences on female fertility.
Collapse
Affiliation(s)
- Christy Lite
- Endocrine Disruption and Reproductive Toxicology (EDART) Laboratory, SRM Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Sheik S S J Ahmed
- Department of Computational Biology, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | - Winkins Santosh
- Endocrine Disruption and Reproductive Toxicology (EDART) Laboratory, SRM Institute of Science and Technology, Chennai, Tamil Nadu, India.,P.G. Research Departments of Advanced Zoology & Biotechnology, Government College for Men, Chennai, Tamil Nadu, India
| | - Barathi Seetharaman
- Endocrine Disruption and Reproductive Toxicology (EDART) Laboratory, SRM Institute of Science and Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
42
|
Sohel MMH, Akyuz B, Konca Y, Arslan K, Sariozkan S, Cinar MU. Oxidative stress modulates the expression of apoptosis-associated microRNAs in bovine granulosa cells in vitro. Cell Tissue Res 2019; 376:295-308. [PMID: 30666538 DOI: 10.1007/s00441-019-02990-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
Despite its essential role in ovulation, oxidative stress (OS) has been found to be cytotoxic to cells, while microRNAs (miRNAs) are known as a major regulator of genes involved in cellular defense against cytotoxicity. However, a functional link between OS and miRNA expression changes in granulosa cells (GCs) remains to be investigated. Here, we investigate the OS modulation of apoptosis-associated miRNAs and their biological relevance in bovine GCs. Following the evaluation of cell viability, accumulation of reactive oxygen species (ROS), cytotoxicity and mitochondrial activity, we used a ready-to-use miRNA PCR array to identify differentially regulated miRNAs. The results showed that exposure to 150 μM H2O2 for 4 h creates remarkable signs of OS in GCs characterized by more than 50% loss of cell viability, higher nuclear factor erythroid 2-related factor 2 (NRF2) nuclear translocation, significantly (p < 0.05) higher abundance of antioxidant genes, significantly (p < 0.001) higher accumulation of ROS, lower mitochondrial activity and a higher (p < 0.001) number of apoptotic nuclei compared to that of the control group. miRNA expression analysis revealed that a total of 69 miRNAs were differentially regulated in which 47 and 22 miRNAs were up- and downregulated, respectively, in stressed GCs. By applying the 2-fold and p < 0.05 criteria, we found 16 miRNAs were upregulated and 10 miRNAs were downregulated. Target prediction revealed that up- and downregulated miRNAs potentially targeted a total of 6210 and 3575 genes, respectively. Pathway analysis showed that upregulated miRNAs are targeting the genes involved mostly in cell survival, intracellular communication and homeostasis, cellular migration and growth control and disease pathways. Our results showed that OS modulates the expression of apoptosis-associated miRNAs that might have effects on cellular or molecular damages.
Collapse
Affiliation(s)
- Md Mahmodul Hasan Sohel
- Genome and Stem Cell Centre, Erciyes University, 38039, Kayseri, Turkey.
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039, Kayseri, Turkey.
| | - Bilal Akyuz
- Department of Genetics, Faculty of Veterinary Science, Erciyes University, 38039, Kayseri, Turkey
| | - Yusuf Konca
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039, Kayseri, Turkey
| | - Korhan Arslan
- Department of Genetics, Faculty of Veterinary Science, Erciyes University, 38039, Kayseri, Turkey
| | - Serpil Sariozkan
- Department of Fertility and Artificial Insemination, Faculty of Veterinary Science, Erciyes University, 38039, Kayseri, Turkey
| | - Mehmet Ulas Cinar
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039, Kayseri, Turkey
| |
Collapse
|
43
|
Tu J, Cheung AHH, Chan CLK, Chan WY. The Role of microRNAs in Ovarian Granulosa Cells in Health and Disease. Front Endocrinol (Lausanne) 2019; 10:174. [PMID: 30949134 PMCID: PMC6437095 DOI: 10.3389/fendo.2019.00174] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/01/2019] [Indexed: 02/02/2023] Open
Abstract
The granulosa cell (GC) is a critical somatic component of the ovary. It is essential for follicle development by supporting the developing oocyte, proliferating and producing sex steroids and disparate growth factors. Knowledge of the GC's function in normal ovarian development and function, and reproductive disorders, such as polycystic ovary syndrome (PCOS) and premature ovarian failure (POF), is largely acquired through clinical studies and preclinical animal models. Recently, microRNAs have been recognized to play important regulatory roles in GC pathophysiology. Here, we examine the recent findings on the role of miRNAs in the GC, including four related signaling pathways (Transforming growth factor-β pathway, Follicle-stimulating hormones pathway, hormone-related miRNAs, Apoptosis-related pathways) and relevant diseases. Therefore, miRNAs appear to be important regulators of GC function in both physiological and pathological conditions. We suggest that targeting specific microRNAs is a potential therapeutic option for treating ovary-related diseases, such as PCOS, POF, and GCT.
Collapse
Affiliation(s)
- Jiajie Tu
- Institute of Clinical Pharmacology, Anhui Medical University, Anhui, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Albert Hoi-Hung Cheung
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | | | - Wai-Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- *Correspondence: Wai-Yee Chan
| |
Collapse
|
44
|
Ziecik AJ, Przygrodzka E, Jalali BM, Kaczmarek MM. Regulation of the porcine corpus luteum during pregnancy. Reproduction 2018; 156:R57-R67. [PMID: 29794023 DOI: 10.1530/rep-17-0662] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/22/2018] [Indexed: 12/31/2022]
Abstract
The new corpora lutea (CLs) in pigs are formed from the preovulatory follicles after the luteinizing hormone (LH) surge. However, total autonomy and independence of CLs from LH up to Day 12 of cycle has recently been questioned. Transformation of estrous cycle CL to CL of pregnancy initiated by embryonic signals requires not only the cessation of prostaglandin F2 (PGF2α) supply to the luteal tissue but also needs the CL to overcome luteolytic acquisition and/or changing its sensitivity to PGF2α during Days 12-14 of pregnancy. The luteolytic cascade is prevented by inhibition of lymphocyte infiltration and leucocyte recruitment, limitation of cell apoptosis, upregulation of pregnancy-associated genes and an enhanced antiluteolytic role of PGE2 Our 'two-signal switch hypothesis' highlights the importance of post PGF2α and PGE2 receptor signaling pathways activation in CLs during luteolysis and rescue. The 'luteolytic switch' involves increased expression of many regression mediators and activation of the post PTGFR signaling pathway. The 'rescue switch' initiated by embryonic signals - estradiol 17β and PGE2 - induces post PTGER2/4 pathway, turning the 'luteolytic switch' off and triggering activity of genes responsible for CL maintenance. In mid and late pregnancy, CLs are maintained by LH and the synergistic action of metabolic hormones. This paper provides an outline of recent views on CL regression, rescue and maintenance during pregnancy in pigs that conflict with previous paradigms and highlights new findings regarding the actions of prostaglandins, role of microRNAs (miRNA) and immune system and signaling pathways governing the life cycle of porcine CL.
Collapse
Affiliation(s)
- Adam J Ziecik
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research PAS, Olsztyn, Poland
| | - Emilia Przygrodzka
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research PAS, Olsztyn, Poland
| | - Beenu M Jalali
- Department of Immunology and Pathology of Reproduction, Institute of Animal Reproduction and Food Research PAS, Olsztyn, Poland
| | - Monika M Kaczmarek
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research PAS, Olsztyn, Poland
| |
Collapse
|
45
|
Eskandari F, Teimoori B, Rezaei M, Mohammadpour‐Gharehbagh A, Narooei‐Nejad M, Mehrabani M, Salimi S. Relationships between Dicer 1 polymorphism and expression levels in the etiopathogenesis of preeclampsia. J Cell Biochem 2018; 119:5563-5570. [DOI: 10.1002/jcb.26725] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/24/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Fatemeh Eskandari
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| | - Batool Teimoori
- Department of Obstetrics and GynecologySchool of MedicineZahedan University of Medical SciencesZahedanIran
| | - Mahnaz Rezaei
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| | - Abbas Mohammadpour‐Gharehbagh
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| | - Mehrnaz Narooei‐Nejad
- Department of Medical GeneticsSchool of MedicineZahedan University of Medical SciencesZahedanIran
| | - Mehrnaz Mehrabani
- Physiology Research CenterInstitute of Basic and Clinical Physiology SciencesKerman University of Medical SciencesKermanIran
| | - Saeedeh Salimi
- Department of Clinical BiochemistrySchool of MedicineZahedan University of Medical SciencesZahedanIran
- Cellular and Molecular Research CenterZahedan University of Medical SciencesZahedanIran
| |
Collapse
|
46
|
Fernández-Pérez D, Brieño-Enríquez MA, Isoler-Alcaraz J, Larriba E, Del Mazo J. MicroRNA dynamics at the onset of primordial germ and somatic cell sex differentiation during mouse embryonic gonad development. RNA (NEW YORK, N.Y.) 2018; 24:287-303. [PMID: 29187591 PMCID: PMC5824349 DOI: 10.1261/rna.062869.117] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/27/2017] [Indexed: 06/07/2023]
Abstract
In mammals, commitment and specification of germ cell lines involves complex programs that include sex differentiation, control of proliferation, and meiotic initiation. Regulation of these processes is genetically controlled by fine-tuned mechanisms of gene regulation in which microRNAs (miRNAs) are involved. We have characterized, by small-RNA-seq and bioinformatics analyses, the miRNA expression patterns of male and female mouse primordial germ cells (PGCs) and gonadal somatic cells at embryonic stages E11.5, E12.5, and E13.5. Differential expression analyses revealed differences in the regulation of key miRNA clusters such as miR-199-214, miR-182-183-96, and miR-34c-5p, whose targets have defined roles during gonadal sexual determination in both germ and somatic cells. Extensive analyses of miRNA sequences revealed an increase in noncanonical isoforms on PGCs at E12.5 and dramatic changes of 3' isomiR expression and 3' nontemplate nucleotide additions in female PGCs at E13.5. Additionally, RT-qPCR analyses of genes encoding proteins involved in miRNA biogenesis and 3' nucleotide addition uncovered sexually and developmentally specific expression, characterized by the decay of Drosha, Dgcr8, and Xpo5 expression along gonadal development. These results demonstrate that miRNAs, their isomiRs, and miRNA machinery are differentially regulated and participate actively in gonadal sexual differentiation in both PGCs and gonadal somatic cells.
Collapse
Affiliation(s)
- Daniel Fernández-Pérez
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Miguel A Brieño-Enríquez
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Javier Isoler-Alcaraz
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Eduardo Larriba
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| | - Jesús Del Mazo
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Madrid 28040, Spain
| |
Collapse
|
47
|
Zhu X, Chen S, Jiang Y, Xu Y, Zhao Y, Chen L, Li C, Zhou X. Analysis of miRNA expression profiles in melatonin-exposed GC-1 spg cell line. Gene 2018; 642:513-521. [DOI: 10.1016/j.gene.2017.11.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/24/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022]
|
48
|
Da Silveira J, Andrade GM, Perecin F, Meireles FV, Winger QA, Bouma GJ. Isolation and Analysis of Exosomal MicroRNAs from Ovarian Follicular Fluid. Methods Mol Biol 2018; 1733:53-63. [PMID: 29435922 DOI: 10.1007/978-1-4939-7601-0_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Mammalian ovarian follicular growth is characterized by development of a large fluid filled antrum that separates mural granulosa cells and cumulus cells. Extensive communication between the different cell types is necessary for maturation of a developmentally competent oocyte. Here, we describe an approach for the isolation of cell-secreted exosomes from ovarian follicular fluid, identification of small RNAs (i.e., microRNAs) in exosomes, labeling of exosomes, and examining cell uptake of exosomes by follicular cells.
Collapse
Affiliation(s)
- Juliano Da Silveira
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of Sao Paulo, Pirassununga, SP, Brazil
| | - Gabriella M Andrade
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of Sao Paulo, Pirassununga, SP, Brazil
| | - Felipe Perecin
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of Sao Paulo, Pirassununga, SP, Brazil
| | - Flávio Vieira Meireles
- Faculty of Animal Sciences and Food Engineering, Department of Veterinary Medicine, University of Sao Paulo, Pirassununga, SP, Brazil
| | - Quinton A Winger
- College of Veterinary and Biomedical Sciences, Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, CO, USA
| | - Gerrit J Bouma
- College of Veterinary and Biomedical Sciences, Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
49
|
Follicle-Stimulating Hormone Receptor: Advances and Remaining Challenges. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:1-58. [DOI: 10.1016/bs.ircmb.2018.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
50
|
Molecular Pathogenesis of Chlamydia Disease Complications: Epithelial-Mesenchymal Transition and Fibrosis. Infect Immun 2017; 86:IAI.00585-17. [PMID: 29084894 DOI: 10.1128/iai.00585-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022] Open
Abstract
The reproductive system complications of genital chlamydial infection include fallopian tube fibrosis and tubal factor infertility. However, the molecular pathogenesis of these complications remains poorly understood. The induction of pathogenic epithelial-mesenchymal transition (EMT) through microRNA (miRNA) dysregulation was recently proposed as the pathogenic basis of chlamydial complications. Focusing on fibrogenesis, we investigated the hypothesis that chlamydia-induced fibrosis is caused by EMT-driven generation of myofibroblasts, the effector cells of fibrosis that produce excessive extracellular matrix (ECM) proteins. The results revealed that the targets of a major category of altered miRNAs during chlamydial infection are key components of the pathophysiological process of fibrogenesis; these target molecules include collagen types I, III, and IV, transforming growth factor β (TGF-β), TGF-β receptor 1 (TGF-βR1), connective tissue growth factor (CTGF), E-cadherin, SRY-box 7 (SOX7), and NFAT (nuclear factor of activated T cells) kinase dual-specificity tyrosine (Y) phosphorylation-regulated kinase 1a (Dyrk1a). Chlamydial induction of EMT resulted in the generation of α-smooth muscle actin (α-SMA)-positive myofibroblasts that produced ECM proteins, including collagen types I and III and fibronectin. Furthermore, the inhibition of EMT prevented the generation of myofibroblasts and production of ECM proteins during chlamydial infection. These findings may provide useful avenues for targeting EMT or specific components of the EMT pathways as a therapeutic intervention strategy to prevent chlamydia-related complications.
Collapse
|