1
|
Giarolla J, Holdaway KA, Nazari M, Aiad L, Sarkar B, Georg GI. Targeting cyclin-dependent kinase 2 (CDK2) interactions with cyclins and Speedy 1 (Spy1) for cancer and male contraception. Future Med Chem 2025; 17:607-627. [PMID: 40034037 PMCID: PMC11901406 DOI: 10.1080/17568919.2025.2463868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
The review discusses progress in discovering cyclin-dependent kinase 2 (CDK2) inhibitors for cancer treatment and their potential for male contraception. It summarizes first-, second-, and third-generation CDK inhibitors and selective CDK2 inhibitors currently in clinical trials for cancer. Novel strategies to discover allosteric inhibitors, covalent inhibitors, and degraders are also discussed.
Collapse
Affiliation(s)
- Jeanine Giarolla
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
- Departamento de Farmacia, School of Pharmaceutical Sciences, University of São Paulo—USP, São Paulo, SP, Brazil
| | - Kelsey A. Holdaway
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Maryam Nazari
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Laila Aiad
- Department of Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Bidisha Sarkar
- Department of Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - Gunda I. Georg
- Medicinal Chemistry, University of Minnesota Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
2
|
Bradley RA, Wolff ID, Cohen PE, Gray S. Dynamic regulatory phosphorylation of mouse CDK2 occurs during meiotic prophase I. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.24.550435. [PMID: 37546989 PMCID: PMC10402020 DOI: 10.1101/2023.07.24.550435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
During prophase I of meiosis, DNA double-strand breaks form throughout the genome, with a subset repairing as crossover events, enabling the accurate segregation of homologous chromosomes during the first meiotic division. The mechanism by which DSBs become selected to repair as crossovers is unknown, although the crossover positioning and levels in each cell indicate it is a highly regulated process. One of the proteins that localises to crossover sites is the serine/threonine cyclin-dependent kinase CDK2. Regulation of CDK2 occurs via phosphorylation at tyrosine 15 (Y15) and threonine 160 (T160) inhibiting and activating the kinase, respectively. In this study we use a combination of immunofluorescence staining on spread spermatocytes and fixed testis sections, and STA-PUT gravitational sedimentation to isolate cells at different developmental stages to further investigate the temporal phospho regulation of CDK2 during prophase I. Western blotting reveals differential levels of the two CDK2 isoforms (CDK233kDa and CDK239kDa) throughout prophase I, with inhibitory phosphorylation of CDK2 at Y15 occurring early in prophase I, localising to telomeres and diminishing as cells enter pachynema. Conversely, the activatory phosphorylation on T160 occurs later, specifically the CDK233kDa isoform, and T160 signal is detected in spermatogonia and pachytene spermatocytes, where it co-localises with the Class I crossover protein MLH3. Taken together, our data reveals intricate control of CDK2 both with regards to levels of the two CDK2 isoforms, and differential regulation via inhibitory and activatory phosphorylation.
Collapse
Affiliation(s)
- Rachel A. Bradley
- Department of Biomedical Sciences and Cornell Reproductive Sciences Center (CoRe), Cornell University, Ithaca, NY, 14853, United States of America
| | - Ian D. Wolff
- Department of Biomedical Sciences and Cornell Reproductive Sciences Center (CoRe), Cornell University, Ithaca, NY, 14853, United States of America
| | - Paula E. Cohen
- Department of Biomedical Sciences and Cornell Reproductive Sciences Center (CoRe), Cornell University, Ithaca, NY, 14853, United States of America
| | - Stephen Gray
- Queen’s Medical Centre, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, United Kingdom
| |
Collapse
|
3
|
Understanding the Underlying Molecular Mechanisms of Meiotic Arrest during In Vitro Spermatogenesis in Rat Prepubertal Testicular Tissue. Int J Mol Sci 2022; 23:ijms23115893. [PMID: 35682573 PMCID: PMC9180380 DOI: 10.3390/ijms23115893] [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: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 12/10/2022] Open
Abstract
In vitro spermatogenesis appears to be a promising approach to restore the fertility of childhood cancer survivors. The rat model has proven to be challenging, since germ cell maturation is arrested in organotypic cultures. Here, we report that, despite a meiotic entry, abnormal synaptonemal complexes were found in spermatocytes, and in vitro matured rat prepubertal testicular tissues displayed an immature phenotype. RNA-sequencing analyses highlighted up to 600 differentially expressed genes between in vitro and in vivo conditions, including genes involved in blood-testis barrier (BTB) formation and steroidogenesis. BTB integrity, the expression of two steroidogenic enzymes, and androgen receptors were indeed altered in vitro. Moreover, most of the top 10 predicted upstream regulators of deregulated genes were involved in inflammatory processes or immune cell recruitment. However, none of the three anti-inflammatory molecules tested in this study promoted meiotic progression. By analysing for the first time in vitro matured rat prepubertal testicular tissues at the molecular level, we uncovered the deregulation of several genes and revealed that defective BTB function, altered steroidogenic pathway, and probably inflammation, could be at the origin of meiotic arrest.
Collapse
|
4
|
Amevor FK, Cui Z, Ning Z, Shu G, Du X, Jin N, Deng X, Xu D, Tian Y, Zhang Y, Li D, Wang Y, Du X, Zhao X. Dietary quercetin and vitamin E supplementation modulates the reproductive performance and antioxidant capacity of aged male breeder chickens. Poult Sci 2022; 101:101851. [PMID: 35472738 PMCID: PMC9061638 DOI: 10.1016/j.psj.2022.101851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 11/25/2022] Open
Abstract
Aged male chickens experience rapid declines in spermatogenesis, antioxidant capacity, immunity, and hormone synthesis. Vitamin E plays a significant role in reproduction, nervous system function, and disease resistance in animals. Quercetin also exerts many biological effects, such as antioxidant ability, immunostimulation, and protection of spermatozoal plasma membranes. This study evaluated the effects of combining dietary quercetin (Q) and vitamin E (VE) on sperm quality, antioxidant capacity, immunity, and expression of genes related to spermatogenesis, immunity, apoptosis, and inflammation in aged male chickens. A total of 120 Tianfu breeder male chickens (65 wk old) were randomly allotted to 4 treatments with 3 replicates (10 birds each). The birds were fed diets containing Q (0.4g/kg), VE (0.2g/kg), Q+VE (0.4g/kg + 0.2g/kg), and a basal diet for 11 wk. At the end of the experimental period, blood, semen, liver, testes, and spleen samples were collected from 2 birds per replicate. Serum hormones, antioxidant parameters, cytokines, and immunoglobulins were evaluated; and the mRNA expression of genes related to spermatogenesis, apoptosis, and inflammation are determined in the testes and liver tissues. The results showed that the combination quercetin and vitamin E significantly promoted the sperm count and motility, as well as elevated the levels of testosterone, follicle-stimulating hormone, and luteinizing hormone, antioxidant enzymes (Superoxide dismutase, Glutathione, and Total antioxidant capacity), and serum immunoglobulins (IgA and IgM) in the aged male chickens; also Q+VE showed protective effects on the liver against injury. In addition, Q+VE significantly increased the expression of genes related to spermatogenesis (AR, pgk2, Cyclin A1, and Cyclin A2), immunity (IFN-γ and IL-2), and anti-inflammatory cytokines (IL-10) (P < 0.05), whereas the expression of proinflammatory cytokines (IL-1β and IL-6) was decreased (P < 0.05). Taken together, these data indicate that the combination of quercetin and vitamin E improved reproductive characteristics such as spermatogenesis, sperm quality, and hormone regulation, as well as promoted antioxidant defense, hepatoprotective capacity, and immune response in aged male chickens without any detrimental effects.
Collapse
|
5
|
Faber EB, Wang N, Georg GI. Review of rationale and progress toward targeting cyclin-dependent kinase 2 (CDK2) for male contraception†. Biol Reprod 2021; 103:357-367. [PMID: 32543655 PMCID: PMC7523694 DOI: 10.1093/biolre/ioaa107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/03/2020] [Accepted: 04/19/2020] [Indexed: 12/30/2022] Open
Abstract
Cyclin-dependent kinase 2 (CDK2) is a member of the larger cell cycle regulating CDK family of kinases, activated by binding partner cyclins as its name suggests. Despite its canonical role in mitosis, CDK2 knockout mice are viable but sterile, suggesting compensatory mechanisms for loss of CDK2 in mitosis but not meiosis. Here, we review the literature surrounding the role of CDK2 in meiosis, particularly a cyclin-independent role in complex with another activator, Speedy 1 (SPY1). From this evidence, we suggest that CDK2 could be a viable nonhormonal male contraceptive target. Finally, we review the literature of pertinent CDK2 inhibitors from the preclinical to clinical stages, mostly developed to treat various cancers. To date, there is no potent yet selective CDK2 inhibitor that could be repurposed as a contraceptive without appreciable off-target toxicity. To achieve selectivity for CDK2 over closely related kinases, developing compounds that bind outside the conserved adenosine triphosphate-binding site may be necessary.
Collapse
Affiliation(s)
- Erik B Faber
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN, USA.,Medical-Scientist Training Program, University of Minnesota Medical School, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Nan Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Gunda I Georg
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
6
|
Cai YT, Xiong CL, Liu TS, Shen SL, Rao JP, Qiu F. Secretions released from mesenchymal stem cells improve spermatogenesis restoration of cytotoxic treatment with busulfan in azoospermia mice. Andrologia 2021; 53:e14144. [PMID: 34143903 DOI: 10.1111/and.14144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 12/25/2022] Open
Abstract
This study aimed at the efficacy of sequential treatment of bone marrow-derived mesenchymal stem cell secretion for busulfan-treated azoospermia in mice. The conditioned media (CM) was obtained from bone marrow mesenchymal stem cells (MSCs) or 293 cells. Chemically induced azoospermia mice received 200 μl MSC-CM or 293-CM twice a week intravenously for three consecutive weeks. The histological assessment of spermatogenic recovery quantifying the expression of meiosis-associated genes, and Sertoli cell barrier functional factors were assessed. The characteristics of TM4 cells (Sertoli cell line) after pre-incubation of MSC-CM in vitro were also obtained. The MSC-CM group had the most spermatogenic colonies among the three groups (p < .05), but no spermatids were seen. Expressions of the meiosis-associated genes Dazl, Vasa, Miwi, Stra8, CyclinA1, Pgk2 and Scp3 in MSC-CM testis were remarkably higher compared with 293-CM and busulfan groups respectively (p < .05). The levels of Sertoli cell barrier functional factors, for example ICAM-1 and N-cadherin, were significantly increased during MSC-CM treatment (p < .05). Moreover, pre-incubation of MSC-CM particularly accelerated the CD54 (ICAM-1) and CD44 expressions of TM4 cells and promoted cell inherent adhesion. MSC-CM treatment can significantly improve the short-term restoration of spermatogonial structures of chemically induced azoospermia related to facilitating Sertoli cell adhesion integrity.
Collapse
Affiliation(s)
- Yi-Ting Cai
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng-Liang Xiong
- Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian-Shu Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shi-Liang Shen
- Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Peng Rao
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Qiu
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
7
|
Function analysis and molecular characterization of cyclin A in ovary development of oriental river prawn, Macrobrachium nipponense. Gene 2021; 788:145583. [PMID: 33753150 DOI: 10.1016/j.gene.2021.145583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/26/2020] [Accepted: 03/12/2021] [Indexed: 11/24/2022]
Abstract
Macrobrachium nipponense has the characteristics of fast ovarian development cycle, which leads to the coexistence of multiple generations, the reduction of commodity specifications and the low economic benefit. Therefore, the study on the mechanism of ovarian development is of great significance to the development of industry. Cyclin A (CycA)is a key gene regulating ovarian development in vertebrates, but little information was available for its function in crustaceans. In this study, the full-length cDNA of Mn-CycA was obtained from the ovary. The full-length cDNA (2033 bp) with an open reading frame of 1368 bp, encoded a 456-amino acid protein. qRT-PCR revealed tissue-specific expression pattern of Mn-CycA, with abundant expression in the ovary. Results in different developmental stages of ovary indicated that Mn-CycA expression is positively correlated with ovarian maturation. qRT-PCR In different developmental stages, the expression of Mn-CycA mRNA gradually increased during the embryonic stage and decreased significantly on the first day of the hatching stage. At the 25th day of the metamorphosis stage, the expression level of Mn-CycAmRNA in female shrimp was 3.5 times higher than that in male shrimp, which may be related to the proliferation of oogonia and the formation of oocytes. In situ hybridization (ISH) of ovary showed Mn-CycA was examined in all stages and was mainly located in oogonia and oocytes. Compared with the control group, the obvious change of gonad somatic index (GSI) proved that injection of Mn-CycA dsRNA could delay the ovarian development cycle, which provided strong evidence for the involvement of Mn-CycA in ovarian maturation and oogenesis, and expanded a new perspective for studying the fast ovarian development cycle in M. nipponense.
Collapse
|
8
|
Kobayashi Y, Tanaka T, Mulati M, Ochi H, Sato S, Kaldis P, Yoshii T, Okawa A, Inose H. Cyclin-Dependent Kinase 1 Is Essential for Muscle Regeneration and Overload Muscle Fiber Hypertrophy. Front Cell Dev Biol 2020; 8:564581. [PMID: 33163487 PMCID: PMC7591635 DOI: 10.3389/fcell.2020.564581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/18/2020] [Indexed: 12/27/2022] Open
Abstract
Satellite cell proliferation is an essential step in proper skeletal muscle development and muscle regeneration. However, the mechanisms regulating satellite cell proliferation are relatively unknown compared to the knowledge associated with the differentiation of satellite cells. Moreover, it is still unclear whether overload muscle fiber hypertrophy is dependent on satellite cell proliferation. In general, cell proliferation is regulated by the activity of cell cycle regulators, such as cyclins and cyclin-dependent kinases (CDKs). Despite recent reports on the function of CDKs and CDK inhibitors in satellite cells, the physiological role of Cdk1 in satellite cell proliferation remains unknown. Herein, we demonstrate that Cdk1 regulates satellite cell proliferation, muscle regeneration, and muscle fiber hypertrophy. Cdk1 is highly expressed in myoblasts and is downregulated upon myoblast differentiation. Inhibition of CDK1 activity inhibits myoblast proliferation. Deletion of Cdk1 in satellite cells leads to inhibition of muscle recovery after muscle injury due to reduced satellite cell proliferation in vivo. Finally, we provide direct evidence that Cdk1 expression in satellite cells is essential for overload muscle fiber hypertrophy in vivo. Collectively, our results demonstrate that Cdk1 is essential for myoblast proliferation, muscle regeneration, and muscle fiber hypertrophy. These findings could help to develop treatments for refractory muscle injuries and muscle atrophy, such as sarcopenia.
Collapse
Affiliation(s)
- Yutaka Kobayashi
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoyuki Tanaka
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mieradilli Mulati
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons with Disabilities, Research Institute, Tokorozawa, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Toshitaka Yoshii
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Okawa
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Inose
- Department of Orthopedic and Trauma Research, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
9
|
Chotiner JY, Wolgemuth DJ, Wang PJ. Functions of cyclins and CDKs in mammalian gametogenesis†. Biol Reprod 2020; 101:591-601. [PMID: 31078132 DOI: 10.1093/biolre/ioz070] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/10/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
Cyclins and cyclin-dependent kinases (CDKs) are key regulators of the cell cycle. Most of our understanding of their functions has been obtained from studies in single-cell organisms and mitotically proliferating cultured cells. In mammals, there are more than 20 cyclins and 20 CDKs. Although genetic ablation studies in mice have shown that most of these factors are dispensable for viability and fertility, uncovering their functional redundancy, CCNA2, CCNB1, and CDK1 are essential for embryonic development. Cyclin/CDK complexes are known to regulate both mitotic and meiotic cell cycles. While some mechanisms are common to both types of cell divisions, meiosis has unique characteristics and requirements. During meiosis, DNA replication is followed by two successive rounds of cell division. In addition, mammalian germ cells experience a prolonged prophase I in males or a long period of arrest in prophase I in females. Therefore, cyclins and CDKs may have functions in meiosis distinct from their mitotic functions and indeed, meiosis-specific cyclins, CCNA1 and CCNB3, have been identified. Here, we describe recent advances in the field of cyclins and CDKs with a focus on meiosis and early embryogenesis.
Collapse
Affiliation(s)
- Jessica Y Chotiner
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
- Cell and Molecular Biology Graduate Program, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Debra J Wolgemuth
- Department of Genetics & Development, Columbia University Medical Center, New York, New York, USA
| | - P Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
- Cell and Molecular Biology Graduate Program, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Ma F, Wang X, Chung SSW, Sicinski P, Shang E, Wolgemuth DJ. Cyclin A2 is essential for mouse gonocyte maturation. Cell Cycle 2020; 19:1654-1664. [PMID: 32420805 DOI: 10.1080/15384101.2020.1762314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
In mammals, male gonocytes are derived from primordial germ cells during embryogenesis, enter a period of mitotic proliferation, and then become quiescent until birth. After birth, the gonocytes proliferate and migrate from the center of testicular cord toward the basement membrane to form the pool of spermatogonial stem cells (SSCs) and establish the SSC niche architecture. However, the molecular mechanisms underlying gonocyte proliferation, migration and differentiation are largely unknown. Cyclin A2 is a key component of the cell cycle and required for cell proliferation. Here, we show that cyclin A2 is required in mouse male gonocyte development and the establishment of spermatogenesis in the neonatal testis. Loss of cyclin A2 function in embryonic gonocytes by targeted gene disruption affected the regulation of the male gonocytes to SSC transition, resulting in the disruption of SSC pool formation, imbalance between SSC self-renewal and differentiation, and severely abnormal spermatogenesis in the adult testis.
Collapse
Affiliation(s)
- Fanhua Ma
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction, College of Animal Science, Huazhong Agricultural University , Wuhan, Hubei, China.,Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA
| | - Xiangyuan Wang
- Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA
| | - Sanny S W Chung
- Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute , Boston, MA, USA
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York , New York, NY, USA
| | - Debra J Wolgemuth
- Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA.,Institute of Human Nutrition, Columbia University Medical Center , New York, NY, USA
| |
Collapse
|
11
|
Cyclin A1 in Oocytes Prevents Chromosome Segregation And Anaphase Entry. Sci Rep 2020; 10:7455. [PMID: 32366979 PMCID: PMC7198627 DOI: 10.1038/s41598-020-64418-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/13/2020] [Indexed: 11/08/2022] Open
Abstract
In several species, including Xenopus, mouse and human, two members of cyclin A family were identified. Cyclin A2, which is ubiquitously expressed in dividing cells and plays role in DNA replication, entry into mitosis and spindle assembly, and cyclin A1, whose function is less clear and which is expressed in spermatocytes, leukemia cells and in postmitotic multiciliated cells. Deletion of the gene showed that cyclin A1 is essential for male meiosis, but nonessential for female meiosis. Our results revealed, that the cyclin A1 is not only dispensable in oocytes, we show here that its expression is in fact undesirable in these cells. Our data demonstrate that the APC/C and proteasome in oocytes are unable to target sufficiently cyclin A1 before anaphase, which leads into anaphase arrest and direct inhibition of separase. The cyclin A1-induced cell cycle arrest is oocyte-specific and the presence of cyclin A1 in early embryos has no effect on cell cycle progression or chromosome division. Cyclin A1 is therefore not only an important cell cycle regulator with biased expression in germline, being essential for male and damaging for female meiosis, its persistent expression during anaphase in oocytes shows fundamental differences between APC/C function in oocytes and in early embryos.
Collapse
|
12
|
Wang C, Zeng YT, Chen XY, Wu QY, Yang LQ, Xu L, Zhang Y, Qazi IH, Zhou GB, Zeng CJ, Zuo ZZ, Song TZ, Zhu Q, Zhang M. Improvac induces immunocastration by affecting testosterone levels and disrupting spermatogenesis in male broiler chickens. Poult Sci 2020; 98:6034-6045. [PMID: 31041439 DOI: 10.3382/ps/pez228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/26/2019] [Indexed: 02/03/2023] Open
Abstract
Immunocastration (vaccination against Gonadotropin-releasing hormone (GnRH)) has been regarded as a friendly substitution to physical castration in animals. To date, a few studies have reported the use of Improvac for immunocastration in boar and one study in broiler chickens; however, there is an apparent dearth of scientific evidence regarding the application of Improvac for immunocastration in birds. In the present study, we evaluated the effects of Improvac-based immunocastration on testosterone levels and spermatogenesis in broiler chickens and the effects of Improvac on the expression of genes related to testosterone biosynthesis and metabolism as well as spermatogenesis. The birds were randomly divided into 4 groups (n = 30 each): Control group (non-immunized), Early group (immunized with Improvac at week 3), Late group (immunized with Improvac at week 6), and Early + Late group (immunized with Improvac at weeks 3 and 6). Immunization with Improvac significantly improved the average daily gain compared to the Control group. Of note, following Improvac vaccination, the reproductive efficiency was significantly decreased in male broiler chickens. Furthermore, parameters such as the serum testosterone concentration, spermatogenesis, and the expression levels of genes related to testosterone metabolism (Cyp17A1, Cyp19, HSD3B1, and HSD17B3) and spermatogenesis (Cyclin A1 and Cyclin A2) were significantly reduced in the immunized groups compared to the Control group. Taken together, these findings reveal that immunization against GnRH can be achieved, at least partially, in male broiler chickens. The results of our study also support the hypothesis of using Improvac as an alternative solution to caponization, with considerably improved animal welfare.
Collapse
Affiliation(s)
- C Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - Y T Zeng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - X Y Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - Q Y Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - L Q Yang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - L Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - Y Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - Izhar Hyder Qazi
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu-611130, P.R. China.,Department of Veterinary Anatomy & Histology, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand-67210, Sindh, Pakistan
| | - G B Zhou
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China.,College of Animal Science and Technology, Sichuan Agricultural University, Chengdu-611130, P.R. China
| | - C J Zeng
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China.,College of Animal Science and Technology, Sichuan Agricultural University, Chengdu-611130, P.R. China
| | - Z Z Zuo
- College of Animal Veterinary Medicine, Sichuan Agricultural University, Chengdu-611130, P.R. China
| | - T Z Song
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, Tibet 850009, China
| | - Q Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| | - M Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, China
| |
Collapse
|
13
|
Singh P, Patel RK, Palmer N, Grenier JK, Paduch D, Kaldis P, Grimson A, Schimenti JC. CDK2 kinase activity is a regulator of male germ cell fate. Development 2019; 146:dev180273. [PMID: 31582414 PMCID: PMC6857589 DOI: 10.1242/dev.180273] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/21/2019] [Indexed: 12/27/2022]
Abstract
The ability of men to remain fertile throughout their lives depends upon establishment of a spermatogonial stem cell (SSC) pool from gonocyte progenitors, and thereafter balancing SSC renewal versus terminal differentiation. Here, we report that precise regulation of the cell cycle is crucial for this balance. Whereas cyclin-dependent kinase 2 (Cdk2) is not necessary for mouse viability or gametogenesis stages prior to meiotic prophase I, mice bearing a deregulated allele (Cdk2Y15S ) are severely deficient in spermatogonial differentiation. This allele disrupts an inhibitory phosphorylation site (Tyr15) for the kinase WEE1. Remarkably, Cdk2Y15S/Y15S mice possess abnormal clusters of mitotically active SSC-like cells, but these are eventually removed by apoptosis after failing to differentiate properly. Analyses of lineage markers, germ cell proliferation over time, and single cell RNA-seq data revealed delayed and defective differentiation of gonocytes into SSCs. Biochemical and genetic data demonstrated that Cdk2Y15S is a gain-of-function allele causing elevated kinase activity, which underlies these differentiation defects. Our results demonstrate that precise regulation of CDK2 kinase activity in male germ cell development is crucial for the gonocyte-to-spermatogonia transition and long-term spermatogenic homeostasis.
Collapse
Affiliation(s)
- Priti Singh
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853, USA
| | - Ravi K Patel
- Cornell University, Department of Molecular Biology and Genetics, Ithaca, NY 14853, USA
| | - Nathan Palmer
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore 138673
- Department of Biochemistry, National University of Singapore, Singapore 117599, Republic of Singapore
| | - Jennifer K Grenier
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853, USA
| | - Darius Paduch
- Cornell University, Weill Cornell Medicine, Department of Urology, New York, NY 10065, USA
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (A*STAR), Singapore 138673
- Department of Biochemistry, National University of Singapore, Singapore 117599, Republic of Singapore
| | - Andrew Grimson
- Cornell University, Department of Molecular Biology and Genetics, Ithaca, NY 14853, USA
| | - John C Schimenti
- Cornell University, College of Veterinary Medicine, Department of Biomedical Sciences, Ithaca, NY 14853, USA
| |
Collapse
|
14
|
Palmer N, Talib SZA, Kaldis P. Diverse roles for CDK-associated activity during spermatogenesis. FEBS Lett 2019; 593:2925-2949. [PMID: 31566717 PMCID: PMC6900092 DOI: 10.1002/1873-3468.13627] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/20/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022]
Abstract
The primary function of cyclin-dependent kinases (CDKs) in complex with their activating cyclin partners is to promote mitotic division in somatic cells. This canonical cell cycle-associated activity is also crucial for fertility as it allows the proliferation and differentiation of stem cells within the reproductive organs to generate meiotically competent cells. Intriguingly, several CDKs exhibit meiosis-specific functions and are essential for the completion of the two reductional meiotic divisions required to generate haploid gametes. These meiosis-specific functions are mediated by both known CDK/cyclin complexes and meiosis-specific CDK-regulators and are important for a variety of processes during meiotic prophase. The majority of meiotic defects observed upon deletion of these proteins occur during the extended prophase I of the first meiotic division. Importantly a lack of redundancy is seen within the meiotic arrest phenotypes described for many of these proteins, suggesting intricate layers of cell cycle control are required for normal meiotic progression. Using the process of male germ cell development (spermatogenesis) as a reference, this review seeks to highlight the diverse roles of selected CDKs their activators, and their regulators during gametogenesis.
Collapse
Affiliation(s)
- Nathan Palmer
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biochemistry, National University of Singapore (NUS), Singapore, Singapore
| | - S Zakiah A Talib
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biochemistry, National University of Singapore (NUS), Singapore, Singapore.,Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Malmö, Sweden
| |
Collapse
|
15
|
La Y, Liu Q, Zhang L, Chu M. Single Nucleotide Polymorphisms in SLC5A1, CCNA1, and ABCC1 and the Association with Litter Size in Small-Tail Han Sheep. Animals (Basel) 2019; 9:ani9070432. [PMID: 31323962 PMCID: PMC6681111 DOI: 10.3390/ani9070432] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/01/2019] [Accepted: 07/01/2019] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Litter size is one of the most important reproductive traits in sheep. Four single nucleotide polymorphisms (SNPs), g.70067210 T > C in SLC5A1, g.25350431 C > T and g.25360220 T > C in CCNA1, and g.14413132 C > T in ABCC1, were identified by mass spectrometry and may be associated with litter size in sheep. Four SNPs were genotyped in Small-Tail Han, Hu, Cele Black, Suffolk, Sunite, Prairie Tibetan, and Tan sheep, and the expression patterns of SLC5A1, CCNA1, and ABCC1 were determined in Small-Tail Han sheep with different fecundities. Furthermore, we also studied the FecB mutation’s association with litter size in Small-Tail Han sheep. The results indicated that all genes included in this study were differentially expressed in the ovary and uterus of polytocous and monotocous Small-Tail Han sheep. Furthermore, association analysis indicated that both g.70067210 T > C in SLC5A1 and the FecB mutation in BMPR-IB were significantly associated with litter size in Small-Tail Han sheep. Linear regression analysis of the association of multiple markers (FecB and g.70067210 T > C in SCL5A1) with litter size indicated that homozygous ewes carrying the BB/TT genotype had a larger litter size than any other genotype. Abstract SLC5A1, CCNA1, and ABCC1 have been extensively studied as candidate genes because of their great influence on the reproductive traits of animals. However, little is known about the association between polymorphisms of the SLC5A1, CCNA1, and ABCC1 genes and litter size in Small-Tail Han sheep. In this study, the expression levels of SLC5A1, CCNA1, and ABCC1 in HPG (hypothalamic–pituitary–gonadal) axis tissues of polytocous and monotocous Small-Tail Han sheep were analyzed by qPCR. To better understand the effects of four single nucleotide polymorphisms (SNPs) comprising of g.70067210 T > C in SLC5A1, g.25350431 C > T and g.25360220 T > C in CCNA1, and g.14413132 C > T in ABCC1, a population genetic analysis was conducted using data obtained from genotyping in 728 sheep from seven breeds. The results indicated that all genes included in this study were differentially expressed in the pituitary and uterus of polytocous and monotocous Small-Tail Han sheep (p < 0.05). The associations of these four SNPs and the FecB mutation with litter size in 384 Small-Tail Han sheep were analyzed, therefore, and it was found that both g.70067210T > C and the FecB mutation were significantly associated with litter size (p < 0.05). The linear regression analysis of the association of multiple markers (FecB and g.70067210 T > C in SCL5A1) with litter size indicated that homozygous ewes carrying the BB/TT genotype had larger litter size than any ewes with any other genotype. In conclusion, the SLC5A1 SNPs significantly affect litter size in sheep and are useful as genetic marker for litter size.
Collapse
Affiliation(s)
- Yongfu La
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Qiuyue Liu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Liping Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Mingxing Chu
- Key Laboratory of Animal Genetics and Breeding and Reproduction of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
16
|
Malla S, Melguizo-Sanchis D, Aguilo F. Steering pluripotency and differentiation with N 6-methyladenosine RNA modification. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:394-402. [PMID: 30412796 DOI: 10.1016/j.bbagrm.2018.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/21/2018] [Accepted: 10/27/2018] [Indexed: 11/15/2022]
Abstract
Chemical modifications of RNA provide a direct and rapid way to modulate the existing transcriptome, allowing the cells to adapt rapidly to the changing environment. Among these modifications, N6-methyladenosine (m6A) has recently emerged as a widely prevalent mark of messenger RNA in eukaryotes, linking external stimuli to an intricate network of transcriptional, post-transcriptional and translational processes. m6A modification modulates a broad spectrum of biochemical processes, including mRNA decay, translation and splicing. Both m6A modification and the enzymes that control m6A metabolism are essential for normal development. In this review, we summarized the most recent findings on the role of m6A modification in maintenance of the pluripotency of embryonic stem cells (ESCs), cell fate specification, the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs), and differentiation of stem and progenitor cells. This article is part of a Special Issue entitled: mRNA modifications in gene expression control edited by Dr. Soller Matthias and Dr. Fray Rupert.
Collapse
Affiliation(s)
- Sandhya Malla
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umeå, Sweden; Department of Medical Biosciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Dario Melguizo-Sanchis
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umeå, Sweden; Department of Medical Biosciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Francesca Aguilo
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umeå, Sweden; Department of Medical Biosciences, Umeå University, SE-901 85 Umeå, Sweden.
| |
Collapse
|
17
|
Jain D, Puno MR, Meydan C, Lailler N, Mason CE, Lima CD, Anderson KV, Keeney S. ketu mutant mice uncover an essential meiotic function for the ancient RNA helicase YTHDC2. eLife 2018; 7:30919. [PMID: 29360036 PMCID: PMC5832417 DOI: 10.7554/elife.30919] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
Mechanisms regulating mammalian meiotic progression are poorly understood. Here we identify mouse YTHDC2 as a critical component. A screen yielded a sterile mutant, ‘ketu’, caused by a Ythdc2 missense mutation. Mutant germ cells enter meiosis but proceed prematurely to aberrant metaphase and apoptosis, and display defects in transitioning from spermatogonial to meiotic gene expression programs. ketu phenocopies mutants lacking MEIOC, a YTHDC2 partner. Consistent with roles in post-transcriptional regulation, YTHDC2 is cytoplasmic, has 3′→5′ RNA helicase activity in vitro, and has similarity within its YTH domain to an N6-methyladenosine recognition pocket. Orthologs are present throughout metazoans, but are diverged in nematodes and, more dramatically, Drosophilidae, where Bgcn is descended from a Ythdc2 gene duplication. We also uncover similarity between MEIOC and Bam, a Bgcn partner unique to schizophoran flies. We propose that regulation of gene expression by YTHDC2-MEIOC is an evolutionarily ancient strategy for controlling the germline transition into meiosis.
Collapse
Affiliation(s)
- Devanshi Jain
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - M Rhyan Puno
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States.,Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, United States
| | - Nathalie Lailler
- Integrated Genomics Operation, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, United States.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, United States.,The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
| | - Christopher D Lima
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States.,Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Kathryn V Anderson
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Scott Keeney
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States.,Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
18
|
Soh YQS, Mikedis MM, Kojima M, Godfrey AK, de Rooij DG, Page DC. Meioc maintains an extended meiotic prophase I in mice. PLoS Genet 2017; 13:e1006704. [PMID: 28380054 PMCID: PMC5397071 DOI: 10.1371/journal.pgen.1006704] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/19/2017] [Accepted: 03/20/2017] [Indexed: 01/13/2023] Open
Abstract
The meiosis-specific chromosomal events of homolog pairing, synapsis, and recombination occur over an extended meiotic prophase I that is many times longer than prophase of mitosis. Here we show that, in mice, maintenance of an extended meiotic prophase I requires the gene Meioc, a germ-cell specific factor conserved in most metazoans. In mice, Meioc is expressed in male and female germ cells upon initiation of and throughout meiotic prophase I. Mouse germ cells lacking Meioc initiate meiosis: they undergo pre-meiotic DNA replication, they express proteins involved in synapsis and recombination, and a subset of cells progress as far as the zygotene stage of prophase I. However, cells in early meiotic prophase—as early as the preleptotene stage—proceed to condense their chromosomes and assemble a spindle, as if having progressed to metaphase. Meioc-deficient spermatocytes that have initiated synapsis mis-express CYCLIN A2, which is normally expressed in mitotic spermatogonia, suggesting a failure to properly transition to a meiotic cell cycle program. MEIOC interacts with YTHDC2, and the two proteins pull-down an overlapping set of mitosis-associated transcripts. We conclude that when the meiotic chromosomal program is initiated, Meioc is simultaneously induced so as to extend meiotic prophase. Specifically, MEIOC, together with YTHDC2, promotes a meiotic (as opposed to mitotic) cell cycle program via post-transcriptional control of their target transcripts. Meiosis is the specialized cell division that halves the genetic content of germ cells to produce haploid gametes. This reductive division is preceded by a preparative phase of the cell cycle, meiotic prophase I, during which several meiosis-specific chromosomal events occur. Across sexually reproducing organisms, prophase of meiosis I is dramatically longer than mitotic prophase. However, it was not known in mammals how and why meiotic prophase I is extended. We have identified a mouse mutant in which this extended prophase I is disrupted: germ cells lacking Meioc initiate meiosis, but prematurely proceed to metaphase. Mutant male meiotic germ cells mis-express a cell cycle regulator that is normally expressed in mitotic male germ cells, suggesting that Meioc is required for germ cells to properly transition to a meiotic cell cycle program. Biochemical analyses of proteins and transcripts that associate with MEIOC protein suggest that MEIOC may promote the transition from a mitotic to meiotic cell cycle program by post-transcriptionally regulating target transcripts. Our studies indicate that in mammals, as in other sexually reproducing organisms, meiotic prophase I must be extended to allow time for meiotic chromosomal events to reach completion.
Collapse
Affiliation(s)
- Y. Q. Shirleen Soh
- Whitehead Institute, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | | | - Mina Kojima
- Whitehead Institute, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Alexander K. Godfrey
- Whitehead Institute, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | | | - David C. Page
- Whitehead Institute, Cambridge, MA, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
19
|
The alpha-fetoprotein (AFP) third domain: a search for AFP interaction sites of cell cycle proteins. Tumour Biol 2016; 37:12697-12711. [DOI: 10.1007/s13277-016-5131-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/29/2016] [Indexed: 01/28/2023] Open
|
20
|
Clement TM, Inselman AL, Goulding EH, Willis WD, Eddy EM. Disrupting Cyclin Dependent Kinase 1 in Spermatocytes Causes Late Meiotic Arrest and Infertility in Mice. Biol Reprod 2015; 93:137. [PMID: 26490841 DOI: 10.1095/biolreprod.115.134940] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 10/15/2015] [Indexed: 01/22/2023] Open
Abstract
While cyclin dependent kinase 1 (CDK1) has a critical role in controlling resumption of meiosis in oocytes, its role has not been investigated directly in spermatocytes. Unique aspects of male meiosis led us to hypothesize that its role is different in male meiosis than in female meiosis. We generated a conditional knockout (cKO) of the Cdk1 gene in mouse spermatocytes to test this hypothesis. We found that CDK1-null spermatocytes undergo synapsis, chiasmata formation, and desynapsis as is seen in oocytes. Additionally, CDK1-null spermatocytes relocalize SYCP3 to centromeric foci, express H3pSer10, and initiate chromosome condensation. However, CDK1-null spermatocytes fail to form condensed bivalent chromosomes in prophase of meiosis I and instead are arrested at prometaphase. Thus, CDK1 has an essential role in male meiosis that is consistent with what is known about the role of CDK1 in female meiosis, where it is required for formation of condensed bivalent metaphase chromosomes and progression to the first meiotic division. We found that cKO spermatocytes formed fully condensed bivalent chromosomes in the presence of okadaic acid, suggesting that cKO chromosomes are competent to condense, although they do not do so in vivo. Additionally, arrested cKO spermatocytes exhibited irregular cell shape, irregular large nuclei, and large distinctive nucleoli. These cells persist in the seminiferous epithelium through the next seminiferous epithelial cycle with a lack of stage XII checkpoint-associated cell death. This indicates that CDK1 is required upstream of a checkpoint-associated cell death as well as meiotic metaphase progression in mouse spermatocytes.
Collapse
Affiliation(s)
- Tracy M Clement
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Amy L Inselman
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Eugenia H Goulding
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - William D Willis
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Edward M Eddy
- Gamete Biology Group, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
21
|
The genetics of human infertility by functional interrogation of SNPs in mice. Proc Natl Acad Sci U S A 2015; 112:10431-6. [PMID: 26240362 DOI: 10.1073/pnas.1506974112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Infertility is a prevalent health issue, affecting ∼15% of couples of childbearing age. Nearly one-half of idiopathic infertility cases are thought to have a genetic basis, but the underlying causes are largely unknown. Traditional methods for studying inheritance, such as genome-wide association studies and linkage analyses, have been confounded by the genetic and phenotypic complexity of reproductive processes. Here we describe an association- and linkage-free approach to identify segregating infertility alleles, in which CRISPR/Cas9 genome editing is used to model putatively deleterious nonsynonymous SNPs (nsSNPs) in the mouse orthologs of fertility genes. Mice bearing "humanized" alleles of four essential meiosis genes, each predicted to be deleterious by most of the commonly used algorithms for analyzing functional SNP consequences, were examined for fertility and reproductive defects. Only a Cdk2 allele mimicking SNP rs3087335, which alters an inhibitory WEE1 protein kinase phosphorylation site, caused infertility and revealed a novel function in regulating spermatogonial stem cell maintenance. Our data indicate that segregating infertility alleles exist in human populations. Furthermore, whereas computational prediction of SNP effects is useful for identifying candidate causal mutations for diverse diseases, this study underscores the need for in vivo functional evaluation of physiological consequences. This approach can revolutionize personalized reproductive genetics by establishing a permanent reference of benign vs. infertile alleles.
Collapse
|
22
|
Liu W, Wang L, Zhao W, Song G, Xu R, Wang G, Wang F, Li W, Lian J, Tian H, Wang X, Sun F. Phosphorylation of CDK2 at threonine 160 regulates meiotic pachytene and diplotene progression in mice. Dev Biol 2014; 392:108-16. [PMID: 24797635 DOI: 10.1016/j.ydbio.2014.04.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 03/31/2014] [Accepted: 04/26/2014] [Indexed: 12/20/2022]
Abstract
Telomere clustering is a widespread phenomenon among eukaryotes. However, the molecular mechanisms that regulate formation of telomere clustering in mammalian meiotic prophase I, are still largely unknown. Here, we show that CDK2, especially p39(cdk2), as a potential meiosis-specific connector interaction with SUN1 mediates formation of telomere clustering during mouse meiosis. The transition from CDK2 to p-CDK2 also regulates the progression from homologous recombination to desynapsis by interacting with MLH1. In addition, disappearance of CDK2 on the telomeres and of p-CDK2 on recombination sites, were observed in Sun1(-/-) mice and in pachytene-arrested hybrid sterile mice (pwk×C57BL/6 F1), respectively. These results suggest that transition from CDK2 to p-CDK2 plays a critical role for regulating meiosis progression.
Collapse
Affiliation(s)
- Wenjing Liu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China; College of Life Science and Technology, Southwest University of Science and Technology, Mianyang, Sichuan 621000, China
| | - Lu Wang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Weidong Zhao
- Engineering College of Animal Husbandry and Veterinary Science, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Gendi Song
- Engineering College of Animal Husbandry and Veterinary Science, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Rener Xu
- Institute of Developmental Biology and Molecular Medicine and School of Life Science, Fudan University, Shanghai 200433, China
| | - Guishuan Wang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Fei Wang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Wenqing Li
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jie Lian
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Hui Tian
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaorong Wang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Fei Sun
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|
23
|
Wei H, Li Y, Zhao C, Jiang X, Chen H, Lang MF, Sun J. Cyclin A1 is expressed in mouse ovary. Int J Med Sci 2014; 11:754-7. [PMID: 24904232 PMCID: PMC4045796 DOI: 10.7150/ijms.8059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 04/01/2014] [Indexed: 11/25/2022] Open
Abstract
Cyclin A1 belongs to the type-A cyclins and participates in cell cycle regulation. Since its discovery, cyclin A1 has been shown mostly in testis. It plays important roles in spermatogenesis. However, there were also reports on ovary expression of cyclin A1. Therefore, we intended to revisit the expression of cyclin A1 in mouse ovary. Our study showed that cyclin A1 was expressed at the mRNA level and the protein level in mouse ovary. Tissue staining revealed that cyclin A1 was expressed in maturating oocytes. With the recent data on the functions of cyclins in somatic and stem cells, we also discussed the possibilities of further studies of cyclin A1 in mouse oocytes and perhaps in the oogonial stem cells. Our findings not only add to the supportive evidence of cyclin A1 expression in oocytes, but also may promote more interest in exploring cyclin A1 functions in ovary.
Collapse
Affiliation(s)
- Hongquan Wei
- 1. Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yuanhong Li
- 2. Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chen Zhao
- 1. Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xuejun Jiang
- 1. Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Hongduo Chen
- 2. Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ming-Fei Lang
- 3. Department of Physiology, Dalian University Medical School, Dalian, Liaoning 116622, P.R. China
| | - Jing Sun
- 1. Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China; ; 4. Current address: Department of Medical Laboratory Science and Biotechnology, Dalian University Medical School, Dalian, Liaoning 116622, P.R. China
| |
Collapse
|
24
|
Chen J, Liu P, Li Z, Chen Y, Qiu GF. The cloning of the cdk2 transcript and the localization of its expression during gametogenesis in the freshwater giant prawn, Macrobrachium rosenbergii. Mol Biol Rep 2013; 40:4781-90. [PMID: 23653005 DOI: 10.1007/s11033-013-2574-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 04/29/2013] [Indexed: 11/28/2022]
Abstract
Cyclin-dependent kinases (cdks) are key regulators of the cell cycle. In mammals, cdk2 plays an essential role in the meiosis of spermatocytes and oocytes. To investigate the role of cdk2 kinase during gametogenesis in crustaceans, we cloned a complete cDNA sequence of cdk2 from the freshwater giant prawn, Macrobrachium rosenbergii, and examined its localization and expression in the developing gonads. The prawn cdk2 cDNA is 1,745 bp in length and encodes a putative protein of 305 amino acids. The deduced protein contains a conserved cyclin binding motif PSTAIRE and shares high homology with reported cdk2 kinases of other species. RT-PCR analysis showed a wide distribution of the cdk2 mRNA in all tested organs including the testis, ovary, heart, muscles, hepatopancreas and gills, and the highest level of expression in the ovary and testis. Localization by in situ hybridization of cdk2 mRNA in the ovary showed high expression in the ooplasm of previtellogenic and the nuclei of late vitellogenic oocytes. In testicular sections, cdk2 transcript is low in spermatogonia, high in spermatocytes, but reduced in spermatids and sperm. The high expression of the cdk2 transcripts in meiotic spermatocytes and oocytes indicated that the cdk2 gene has the conservative function in the germ cells meiosis during gametogenesis.
Collapse
Affiliation(s)
- Jie Chen
- Key Laboratory of Aquatic Genetic Resources and Utilization Certificated by the Ministry of Agriculture, College of Life Science, Shanghai Ocean University, 999 Hucheng Huan Road, Pudong New Area, Shanghai, 201306, People's Republic of China
| | | | | | | | | |
Collapse
|
25
|
Wolgemuth DJ, Manterola M, Vasileva A. Role of cyclins in controlling progression of mammalian spermatogenesis. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2013; 57:159-68. [PMID: 23784826 PMCID: PMC3982229 DOI: 10.1387/ijdb.130047av] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cyclins are key regulators of the mammalian cell cycle, functioning primarily in concert with their catalytic partners, the cyclin-dependent kinases (Cdks). While their function during mitosis in somatic cells has been extensively documented, their function during both mitosis and meiosis in the germ line is poorly understood. From the perspective of cell cycle regulation there are several aspects of mammalian spermatogenesis that suggest unique modes of regulation and hence, possible unique functions for the cyclins. This review will summarize our current understanding of cyclin expression and function in the male germ line, with particular focus on the A and E type cyclins in the mouse model. While the focus is on mammalian spermatogenesis, we note contrasts with similar functions in the female germ line when relevant and also draw upon observations in other model systems to provide further insight.
Collapse
Affiliation(s)
- Debra J Wolgemuth
- Departments of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA.
| | | | | |
Collapse
|
26
|
Boucheron-Houston C, Canterel-Thouennon L, Lee TL, Baxendale V, Nagrani S, Chan WY, Rennert OM. Long-term vitamin A deficiency induces alteration of adult mouse spermatogenesis and spermatogonial differentiation: direct effect on spermatogonial gene expression and indirect effects via somatic cells. J Nutr Biochem 2012; 24:1123-35. [PMID: 23253600 DOI: 10.1016/j.jnutbio.2012.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 08/03/2012] [Accepted: 08/20/2012] [Indexed: 10/27/2022]
Abstract
The objective of this study was to further understand the genetic mechanisms of vitamin A deficiency (VAD) induced arrest of spermatogonial stem-cell differentiation. Vitamin A and its derivatives (the retinoids) participate in many physiological processes including vision, cellular differentiation and reproduction. VAD affects spermatogenesis, the subject of our present study. Spermatogenesis is a highly regulated process of differentiation and complex morphologic alterations that leads to the formation of sperm in the seminiferous epithelium. VAD causes early cessation of spermatogenesis, characterized by degeneration of meiotic germ cells, leading to seminiferous tubules containing mostly type A spermatogonia and Sertoli cells. These observations led us to the hypothesis that VAD affects not only germ cells but also somatic cells. To investigate the effects of VAD on spermatogenesis in mice we used adult Balb/C mice fed with Control or VAD diet for an extended period of time (6-28 weeks). We first observed the chronology, then the extent of the effects of VAD on the testes. Using microarray analysis of isolated pure populations of spermatogonia, Leydig and Sertoli cells from control and VAD 18- and 25-week mice, we examined the effects of VAD on gene expression and identified target genes involved in the arrest of spermatogonial differentiation and spermatogenesis. Our results provide a more precise definition of the chronology and magnitude of the consequences of VAD on mouse testes than the previously available literature and highlight direct and indirect (via somatic cells) effects of VAD on germ cell differentiation.
Collapse
Affiliation(s)
- Catherine Boucheron-Houston
- Laboratory of Clinical Genomics, Section on Developmental Genomics, National Institutes of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-4429, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Panigrahi SK, Vasileva A, Wolgemuth DJ. Sp1 transcription factor and GATA1 cis-acting elements modulate testis-specific expression of mouse cyclin A1. PLoS One 2012; 7:e47862. [PMID: 23112860 PMCID: PMC3480434 DOI: 10.1371/journal.pone.0047862] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Accepted: 09/18/2012] [Indexed: 01/16/2023] Open
Abstract
Cyclin A1 is a male germ cell-specific cell cycle regulator that is essential for spermatogenesis. It is unique among the cyclins by virtue of its highly restricted expression in vivo, being present in pachytene and diplotene spermatocytes and not in earlier or later stages of spermatogenesis. To begin to understand the molecular mechanisms responsible for this narrow window of expression of the mouse cyclin A1 (Ccna1) gene, we carried out a detailed analysis of its promoter. We defined a 170-bp region within the promoter and showed that it is involved in repression of Ccna1 in cultured cells. Within this region we identified known cis-acting transcription factor binding sequences, including an Sp1-binding site and two GATA1-binding sites. Neither Sp1 nor GATA1 is expressed in pachytene spermatocytes and later stages of germ cell differentiation. Sp1 is readily detected at earlier stages of spermatogenesis. Site-directed mutagenesis demonstrated that neither factor alone was sufficient to significantly repress expression driven by the Ccna1 promoter, while concurrent binding of Sp1, and most likely GATA1 and possibly additional factors was inhibitory. Occupancy of Sp1 on the Ccna1 promoter and influence of GATA1-dependent cis-acting elements was confirmed by ChIP analysis in cell lines and most importantly, in spermatogonia. In contrast with many other testis-specific genes, the CpG island methylation status of the Ccna1 promoter was similar among various tissues examined, irrespective of whether Ccna1 was transcriptionally active, suggesting that this regulatory mechanism is not involved in the restricted expression of Ccna1.
Collapse
Affiliation(s)
- Sunil K. Panigrahi
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, United States of America
| | - Ana Vasileva
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, United States of America
- Center for Radiological Research, Columbia University Medical Center, New York, New York, United States of America
| | - Debra J. Wolgemuth
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, United States of America
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, New York, United States of America
- Institute of Human Nutrition, Columbia University Medical Center, New York, New York, United States of America
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
28
|
Greaves EA, Copeland NA, Coverley D, Ainscough JFX. Cancer associated variant expression and interaction of CIZ1 with cyclin A1 in differentiating male germ cells. J Cell Sci 2012; 125:2466-77. [DOI: 10.1242/jcs.101097] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
CIZ1 is a nuclear matrix associated DNA replication factor unique to higher eukaryotes, for which alternatively spliced isoforms have been associated with a range of disorders. In vitro the CIZ1 N-terminus interacts with cyclins E and A via distinct sites, enabling functional cooperation with cyclin A-Cdk2 to promote replication initiation. C-terminal sequences anchor CIZ1 to fixed sites on the nuclear matrix imposing spatial constraint on cyclin dependent kinase activity. Here we demonstrate that CIZ1 is predominantly expressed as predicted full-length product throughout mouse development, consistent with a ubiquitous role in cell and tissue renewal. CIZ1 is expressed in proliferating stem cells of the testis, but is notably down-regulated following commitment to differentiation. Significantly, CIZ1 is re-expressed at high levels in non-proliferative spermatocytes prior to meiotic division. Sequence analysis identifies at least seven alternatively spliced variants at this time, including a dominant cancer-associated form and a set of novel isoforms. Furthermore, we show that in these post-replicative cells CIZ1 interacts with the germ cell specific cyclin, A1, that has been implicated in DNA double-strand break repair. Consistent with this role, antibody depletion of CIZ1 reduces the capacity for testis extract to repair digested plasmid DNA in vitro. Together, the data imply novel post-replicative roles for CIZ1 in germ cell differentiation that may include meiotic recombination, a process intrinsic to genome stability and diversification.
Collapse
|
29
|
Abstract
The cyclins and their cyclin-dependent kinase partners, the Cdks, are the basic components of the machinery that regulates the passage of cells through the cell cycle. Among the cyclins, those known as the A-type cyclins are unique in that in somatic cells, they appear to function at two stages of the cell cycle, at the G1-S transition and again as the cells prepare to enter M-phase. Higher vertebrate organisms have two A-type cyclins, cyclin A1 and cyclin A2, both of which are expressed in the germ line and/or early embryo, following highly specialized patterns that suggest functions in both mitosis and meiosis. Insight into their in vivo functions has been obtained from gene targeting experiments in the mouse model. Loss of cyclin A1 results in disruption of spermatogenesis and male sterility due to cell arrest in the late diplotene stage of the meiotic cell cycle. In contrast, cyclin A2-deficiency is marked by early embryonic lethality; thus, understanding the function of cyclin A2 in the adult germ line awaits conditional mutagenesis or other approaches to knock down its expression.
Collapse
|
30
|
Abdou AG, Hammam MA, Farag AGA, Farouk S, Fawzy M. Immunohistochemical expression of cyclin A in testicular biopsies of fertile and infertile men: correlation with the morphometry of seminiferous tubules. Andrologia 2010; 43:57-64. [PMID: 21219384 DOI: 10.1111/j.1439-0272.2009.01018.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cyclin A is a member of the cyclin family of proteins, which are required for both the mitotic and meiotic divisions that characterise spermatogenesis in human and other mammalian species. The data on cyclin A expression in various human spermatogenic disorders and its relationship to the morphology of seminiferous tubules are not well clarified. This study aimed to evaluate the immunohistochemical expression of cyclin A in testicular biopsies of different spermatogenic disorders correlating with the morphology of seminiferous tubules using morphometry tools. Immunohistochemical evaluation of cyclin A was carried out on testicular biopsies obtained from 48 infertile males (nonobstructive azoospermia) and 15 normal subjects together with using semiautomatic morphometric analysis for evaluation of seminiferous tubules. Cyclin A is expressed in 100% of normal and hypospermatogenesis groups and in 80% of maturation arrest group, with complete absence in Sertoli cell only group. In positive cases, cyclin A stained the nuclei of spermatogonia and primary spermatocytes with a higher intensity of expression in normal cases compared with infertile group. Cyclin A expression was significantly associated with the different examined morphometric parameters. Cyclin A is involved in both mitosis and meiosis of human spermatogenesis as it is expressed in spermatogonia and primary spermatocytes. Morphometry of human testis is intimately correlated with the testicular histopathology.
Collapse
Affiliation(s)
- A G Abdou
- Pathology Department, Menofiya University, Shebein Elkom, Egypt.
| | | | | | | | | |
Collapse
|
31
|
Wolgemuth DJ, Roberts SS. Regulating mitosis and meiosis in the male germ line: critical functions for cyclins. Philos Trans R Soc Lond B Biol Sci 2010; 365:1653-62. [PMID: 20403876 DOI: 10.1098/rstb.2009.0254] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Key components of the cell cycle machinery are the regulatory subunits, the cyclins, and their catalytic partners the cyclin-dependent kinases. Regulating the cell cycle in the male germ line cells represents unique challenges for this machinery given the constant renewal of gametes throughout the reproductive lifespan and the induction of the unique process of meiosis, a highly specialized kind of cell division. With challenges come opportunities to the critical eye, recognizing that understanding these specialized modes of regulation will provide considerable insight into both normal differentiation as well as disease conditions, including infertility and oncogenesis.
Collapse
Affiliation(s)
- Debra J Wolgemuth
- Department of Genetics and Development, Columbia University Medical Center, 1150 St Nicholas Avenue, Room 608, New York, NY 10032, USA.
| | | |
Collapse
|
32
|
d'Erfurth I, Cromer L, Jolivet S, Girard C, Horlow C, Sun Y, To JPC, Berchowitz LE, Copenhaver GP, Mercier R. The cyclin-A CYCA1;2/TAM is required for the meiosis I to meiosis II transition and cooperates with OSD1 for the prophase to first meiotic division transition. PLoS Genet 2010; 6:e1000989. [PMID: 20585549 PMCID: PMC2887465 DOI: 10.1371/journal.pgen.1000989] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Accepted: 05/14/2010] [Indexed: 11/25/2022] Open
Abstract
Meiosis halves the chromosome number because its two divisions follow a single round of DNA replication. This process involves two cell transitions, the transition from prophase to the first meiotic division (meiosis I) and the unique meiosis I to meiosis II transition. We show here that the A-type cyclin CYCA1;2/TAM plays a major role in both transitions in Arabidopsis. A series of tam mutants failed to enter meiosis II and thus produced diploid spores and functional diploid gametes. These diploid gametes had a recombined genotype produced through the single meiosis I division. In addition, by combining the tam-2 mutation with AtSpo11-1 and Atrec8, we obtained plants producing diploid gametes through a mitotic-like division that were genetically identical to their parents. Thus tam alleles displayed phenotypes very similar to that of the previously described osd1 mutant. Combining tam and osd1 mutations leads to a failure in the prophase to meiosis I transition during male meiosis and to the production of tetraploid spores and gametes. This suggests that TAM and OSD1 are involved in the control of both meiotic transitions.
Collapse
Affiliation(s)
- Isabelle d'Erfurth
- Institut Jean-Pierre Bourgin, UMR1318 INRA-AgroParisTech, Versailles, France
| | - Laurence Cromer
- Institut Jean-Pierre Bourgin, UMR1318 INRA-AgroParisTech, Versailles, France
| | - Sylvie Jolivet
- Institut Jean-Pierre Bourgin, UMR1318 INRA-AgroParisTech, Versailles, France
| | - Chloé Girard
- Institut Jean-Pierre Bourgin, UMR1318 INRA-AgroParisTech, Versailles, France
| | - Christine Horlow
- Institut Jean-Pierre Bourgin, UMR1318 INRA-AgroParisTech, Versailles, France
| | - Yujin Sun
- Department of Biology and the Carolina Center for Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jennifer P. C. To
- Department of Biology and the Carolina Center for Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Luke E. Berchowitz
- Department of Biology and the Carolina Center for Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Gregory P. Copenhaver
- Department of Biology and the Carolina Center for Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Raphael Mercier
- Institut Jean-Pierre Bourgin, UMR1318 INRA-AgroParisTech, Versailles, France
| |
Collapse
|
33
|
FIGLA, a basic helix-loop-helix transcription factor, balances sexually dimorphic gene expression in postnatal oocytes. Mol Cell Biol 2010; 30:3661-71. [PMID: 20479125 DOI: 10.1128/mcb.00201-10] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Maintenance of sex-specific germ cells requires balanced activation and repression of genetic hierarchies to ensure gender-appropriate development in mammals. Figla (factor in the germ line, alpha) encodes a germ cell-specific basic helix-loop-helix transcription factor first identified as an activator of oocyte genes. In comparing the ovarian proteome of normal and Figla null newborn mice, 18 testis-specific or -enhanced proteins were identified that were more abundant in Figla null ovaries than in normal ovaries. Transgenic mice, ectopically expressing Figla in male germ cells, downregulated a subset of these genes and demonstrated age-related sterility associated with impaired meiosis and germ cell apoptosis. Testis-associated genes, including Tdrd1, Tdrd6, and Tdrd7, were suppressed in the transgenic males with a corresponding disruption of the sperm chromatoid body and mislocalization of MVH and MILI proteins, previously implicated in posttranscriptional processing of RNA. These data demonstrate that physiological expression of Figla plays a critical dual role in activation of oocyte-associated genes and repression of sperm-associated genes during normal postnatal oogenesis.
Collapse
|
34
|
Yousafzai FK, Al-Kaff N, Moore G. The molecular features of chromosome pairing at meiosis: the polyploid challenge using wheat as a reference. Funct Integr Genomics 2010; 10:147-56. [PMID: 20422242 DOI: 10.1007/s10142-010-0171-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 03/18/2010] [Accepted: 03/27/2010] [Indexed: 11/29/2022]
Abstract
During meiosis, chromosome numbers are halved, leading to haploid gametes, a process that is crucial for the maintenance of a stable genome through successive generations. The process for the accurate segregation of the homologues starts in pre-meiosis as each homologue is replicated and the respective products are held together as two sister chromatids via specific cohesion proteins. At the start of meiosis, each chromosome must recognise its homologue from amongst all the chromosomes present in the nucleus and then associate or pair with that homologue. This process of homologue recognition in meiosis is more complicated in polyploids because of the greater number of related chromosomes. Despite the presence of these related chromosomes, for polyploids such as wheat to produce viable gametes, they must behave as diploids during meiosis with only true homologues pairing. In this review, the relationship between the Ph1 cyclin-dependent kinase (CDK)-like genes in wheat and the CDK2 genes in mammals and their involvement in controlling this process at meiosis is examined.
Collapse
|
35
|
Kano K, Kitamura A, Matsuwaki T, Morimatsu M, Naito K. Discoidin domain receptor 2 (DDR2) is required for maintenance of spermatogenesis in male mice. Mol Reprod Dev 2010; 77:29-37. [PMID: 19681157 DOI: 10.1002/mrd.21093] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Discoidin domain receptor 2 (DDR2) is a receptor tyrosine kinase (RTK). We recently identified homozygous smallie mutant mice (BKS.HRS. Ddr2(slie/slie)/J, Ddr2(slie/slie) mutants), which lack a functional DDR2. Ddr2(slie/slie) mutant mice are dwarfed and infertile due to peripheral dysregulation of the endocrine system. To understand the role of DDR2 signaling in spermatogenesis, we studied the expression of several receptors, enzymes, and proteins related to spermatogenesis in wild-type and Ddr2(slie/slie) mutant mice at 10 weeks and 5 months of age. DDR2 were expressed in adult wild-type male mice in Leydig cells. The number of differentiated spermatozoa in the seminal fluid was significantly lower in the Ddr2(slie/slie) mutant mice than in the wild-type mice. The number of TUNEL-positive cells was significantly greater in 5-month-old Ddr2(slie/slie) mutants. Testosterone was significantly reduced at 5 months of age, but LH was similar in both types of mice at both 10 weeks and 5 months of age. The expression levels of LH receptors (Lhcgr), StAR, P450scc, and Hsd3beta6 were not significantly different between the two types of mice at 10 weeks of age, but they were significantly reduced in 5-month-old Ddr2(slie/slie) mutants compared to wild-type mice of the same age. DDR2 was expressed in the Leydig cells of adult wild-type male mice. In conclusion, our results indicated that DDR2 signaling plays a critical role in the maintenance of male spermatogenesis.
Collapse
Affiliation(s)
- Kiyoshi Kano
- Laboratory of Applied Genetics, Graduate School of Agricultural and Life Science, University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan.
| | | | | | | | | |
Collapse
|
36
|
Zhoucun A, Zhang S, Yang Y. Mutations of the cyclin A1 gene are not a common cause of male infertility. Syst Biol Reprod Med 2010; 55:125-8. [PMID: 19886767 DOI: 10.3109/19396360902839828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cyclin A1 is essential for meiosis as shown by its essential role in mouse spermatogenesis, suggesting that changes in the gene may also alter male fertility in humans. In the present study, we performed a mutation screening of the cyclin A1 gene in order to investigate the possible association between the mutations of the gene and human impaired spermatogenesis using denaturing high performance liquid chromatography (DHPLC) in 347 infertile patients with azoospermia or severe oligozoospermia and 210 fertile controls. Four point mutations, c.321T>C, IVS3 +32G>C, IVS5+38A>G and c.1158G>A, were identified, but no association of these with spermatogenesis impairment was detected, suggesting that these cyclin A1 gene mutations are unlikely a common genetic cause for impaired human spermatogenesis.
Collapse
Affiliation(s)
- A Zhoucun
- Department of Biology and Chemistry, Dali College, Dali, China.
| | | | | |
Collapse
|
37
|
Perrard MH, Chassaing E, Montillet G, Sabido O, Durand P. Cytostatic factor proteins are present in male meiotic cells and beta-nerve growth factor increases mos levels in rat late spermatocytes. PLoS One 2009; 4:e7237. [PMID: 19802389 PMCID: PMC2751818 DOI: 10.1371/journal.pone.0007237] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 08/27/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In co-cultures of pachytene spermatocytes with Sertoli cells, beta-NGF regulates the second meiotic division by blocking secondary spermatocytes in metaphase (metaphase II), and thereby lowers round spermatid formation. In vertebrates, mature oocytes are arrested at metaphase II until fertilization, because of the presence of cytostatic factor (CSF) in their cytoplasm. By analogy, we hypothesized the presence of CSF in male germ cells. METHODOLOGY/PRINCIPAL FINDINGS We show here, that Mos, Emi2, cyclin E and Cdk2, the four proteins of CSF, and their respective mRNAs, are present in male rat meiotic cells; this was assessed by using Western blotting, immunocytochemistry and reverse transcriptase PCR. We measured the relative cellular levels of Mos, Emi2, Cyclin E and Cdk2 in the meiotic cells by flow cytometry and found that the four proteins increased throughout the first meiotic prophase, reaching their highest levels in middle to late pachytene spermatocytes, then decreased following the meiotic divisions. In co-cultures of pachytene spermatocytes with Sertoli cells, beta-NGF increased the number of metaphases II, while enhancing Mos and Emi2 levels in middle to late pachytene spermatocytes, pachytene spermatocytes in division and secondary spermatocytes. CONCLUSION/SIGNIFICANCE Our results suggest that CSF is not restricted to the oocyte. In addition, they reinforce the view that NGF, by enhancing Mos in late spermatocytes, is one of the intra-testicular factors which adjusts the number of round spermatids that can be supported by Sertoli cells.
Collapse
Affiliation(s)
- Marie-Hélène Perrard
- Institut de génomique fonctionnelle de Lyon, Université de Lyon, INRA UMR 1288, CNRS UMR 5242, Université Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France.
| | | | | | | | | |
Collapse
|
38
|
Kim ST, Park NC, Yi LSH, Gye MC. EXPRESSION OF P57KIP2 IN GERM CELLS AND LEYDIG CELLS IN HUMAN TESTIS. ACTA ACUST UNITED AC 2009; 52:463-9. [PMID: 17050328 DOI: 10.1080/01485010600822903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
p57kip2, a KIP family cyclin-dependent kinase (Cdk) inhibitor, blocks the cell cycle by acting on multiple cyclin-Cdk complexes. To investigate the role of p57kip2 in human fertility, the expression of p57kip2 was investigated in testes from normal and obstructive azoospermic male patients who were positive for p57kip2 mRNA. In the seminiferous tubule, strong immunoreactivity of p57kip2 was found in nuclei of early spermatocytes, but not in the spermatogonia. The p57kip2 immunoreactivity in spermatocytes was markedly heterogeneous. Preleptotene spermatocytes showed strong p57kip2 immunoreactivity, but no visible signal was found in late pachytene spermatocytes. Nuclei of the elongating spermatids was also positive for p57kip2 immunoreactivity. Taken together, this suggests that p57kip2 may play a role in the regulation of meiotic progression of early spermatocytes and cell cycle arrest and differentiation of spermatids. p57kip2 immunoreactivity was found in the perinuclear region of the peritubular cells, but not in the Sertoli cells. In Leydig cells, moderate immunoreactivity of p57kip2 was largely found in the cytoplasm, suggesting the noble function of p57kip2 in the differentiation of adult Leydig cells.
Collapse
Affiliation(s)
- S T Kim
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
39
|
Satyanarayana A, Kaldis P. Mammalian cell-cycle regulation: several Cdks, numerous cyclins and diverse compensatory mechanisms. Oncogene 2009; 28:2925-39. [PMID: 19561645 DOI: 10.1038/onc.2009.170] [Citation(s) in RCA: 552] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
After a decade of extensive work on gene knockout mouse models of cell-cycle regulators, the classical model of cell-cycle regulation was seriously challenged. Several unexpected compensatory mechanisms were uncovered among cyclins and Cdks in these studies. The most astonishing observation is that Cdk2 is dispensable for the regulation of the mitotic cell cycle with both Cdk4 and Cdk1 covering for Cdk2's functions. Similar to yeast, it was recently discovered that Cdk1 alone can drive the mammalian cell cycle, indicating that the regulation of the mammalian cell cycle is highly conserved. Nevertheless, cell-cycle-independent functions of Cdks and cyclins such as in DNA damage repair are still under investigation. Here we review the compensatory mechanisms among major cyclins and Cdks in mammalian cell-cycle regulation.
Collapse
Affiliation(s)
- A Satyanarayana
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702-1201, USA.
| | | |
Collapse
|
40
|
Stoffel W, Holz B, Jenke B, Binczek E, Günter RH, Kiss C, Karakesisoglou I, Thevis M, Weber AA, Arnhold S, Addicks K. Delta6-desaturase (FADS2) deficiency unveils the role of omega3- and omega6-polyunsaturated fatty acids. EMBO J 2009; 27:2281-92. [PMID: 19172737 DOI: 10.1038/emboj.2008.156] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Mammalian cell viability is dependent on the supply of the essential fatty acids (EFAs) linoleic and alpha-linolenic acid. EFAs are converted into omega3- and omega6-polyunsaturated fatty acids (PUFAs), which are essential constituents of membrane phospholipids and precursors of eicosanoids, anandamide and docosanoids. Whether EFAs, PUFAs and eicosanoids are essential for cell viability has remained elusive. Here, we show that deletion of delta6-fatty acid desaturase (FADS2) gene expression in the mouse abolishes the initial step in the enzymatic cascade of PUFA synthesis. The lack of PUFAs and eicosanoids does not impair the normal viability and lifespan of male and female fads2 -/- mice, but causes sterility. We further provide the molecular evidence for a pivotal role of PUFA-substituted membrane phospholipids in Sertoli cell polarity and blood-testis barrier, and the gap junction network between granulosa cells of ovarian follicles. The fads2 -/- mouse is an auxotrophic mutant. It is anticipated that FADS2 will become a major focus in membrane, haemostasis, inflammation and atherosclerosis research.
Collapse
Affiliation(s)
- Wilhelm Stoffel
- Center of Molecular Medicine (CMMC), Laboratory of Molecular Neurosciences, Institute of Biochemistry, University of Cologne, Cologne, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Joshi AR, Jobanputra V, Lele KM, Wolgemuth DJ. Distinct properties of cyclin-dependent kinase complexes containing cyclin A1 and cyclin A2. Biochem Biophys Res Commun 2008; 378:595-9. [PMID: 19056339 DOI: 10.1016/j.bbrc.2008.11.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Accepted: 11/18/2008] [Indexed: 11/19/2022]
Abstract
The distinct expression patterns of the two A-type cyclins during spermatogenesis and the absolute requirement for cyclin A1 in this biological process in vivo suggest that they may confer distinct biochemical properties to their CDK partners. We therefore compared human cyclin A1- and cyclin A2-containing CDK complexes in vitro by determining kinetic constants and by examining the complexes for their ability to phosphorylate pRb and p53. Differences in biochemical activity were observed in CDK2 but not CDK1 when complexed with cyclin A1 versus cyclin A2. Further, CDK1/cyclin A1 is a better kinase complex for phosphorylating potentially physiologically relevant substrates pRb and p53 than CDK2/cyclin A2. The activity of CDKs can therefore be regulated depending upon which A-type cyclin they bind and CDK1/cyclin A1 might be preferred in vivo.
Collapse
Affiliation(s)
- Ayesha R Joshi
- Department of Gen. and Dev., Columbia University Medical Center, New York, NY 10032, USA
| | | | | | | |
Collapse
|
42
|
Chen X, Hu T, Liang G, Yang M, Zong S, Miao S, Koide SS, Wang L. A novel testis protein, RSB-66, interacting with INCA1 (inhibitor of Cdk interacting with cyclin A1). Biochem Cell Biol 2008; 86:345-51. [PMID: 18756329 DOI: 10.1139/o08-072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
rsb-66 is a novel gene from a suppression subtracted hybridization (SSH) library of round spermatid-specific cDNAs against those of primary spermatocytes. It was found to be specifically expressed in round spermatids. To explore the function of RSB-66, a yeast two-hybrid system was used to screen for potential interacting partners in a human testis cDNA library. HSD45, also known as INCA1 (inhibitor of Cdk interacting with cyclin A1), was identified as one of the positive clones. The interaction between RSB-66 and INCA1 was demonstrated to occur by GST pull down and coimmunoprecipitation. Using immunofluorescence, RSB-66 was found to be specifically expressed in round spermatids, mainly in the cytoplasm. When being transfected into HeLa cells, RSB-66 and INCA1 were found to be co-localized principally in the cytoplasm. The alpha helix in the RSB-66 C terminal and two amino acid residues (tyr117 and his119) appear to be crucial for its function.
Collapse
Affiliation(s)
- Xu Chen
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Wolgemuth DJ. Function of cyclins in regulating the mitotic and meiotic cell cycles in male germ cells. Cell Cycle 2008; 7:3509-13. [PMID: 19001847 PMCID: PMC4080918 DOI: 10.4161/cc.7.22.6978] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The specialized cell cycles that characterize various aspects of the differentiation of germ cells provide a unique opportunity to understand heretofore elusive aspects of the in vivo function of cell cycle regulators. Key components of the cell cycle machinery are the regulatory sub-units, the cyclins, and their catalytic partners, the cyclin-dependent kinases. Some of the cyclins exhibit unique patterns of expression in germ cells that suggest possible concomitant distinct functions, predictions that are being explored by targeted mutagenesis in mouse models. A novel, meiosis-specific function has been shown for one of the A-type cyclins, cyclin A1. Embryonic lethality has obviated understanding of the germline functions of cyclin A2 and cyclin B1, while yet other cyclins, although expressed at specific stages of germ cell development, may have less essential function in the male germline.
Collapse
Affiliation(s)
- Debra J Wolgemuth
- Departments of Genetics and Development and Obstetrics and Gynecology, The Institute of Human Nutrition, The Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York 10032, USA.
| |
Collapse
|
44
|
Satyanarayana A, Berthet C, Lopez-Molina J, Coppola V, Tessarollo L, Kaldis P. Genetic substitution of Cdk1 by Cdk2 leads to embryonic lethality and loss of meiotic function of Cdk2. Development 2008; 135:3389-400. [PMID: 18787066 PMCID: PMC2668819 DOI: 10.1242/dev.024919] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
It was believed that Cdk2-cyclin E complexes are essential to drive cells through the G1-S phase transition. However, it was discovered recently that the mitotic kinase Cdk1 (Cdc2a) compensates for the loss of Cdk2. In the present study, we tested whether Cdk2 can compensate for the loss of Cdk1. We generated a knockin mouse in which the Cdk2 cDNA was knocked into the Cdk1 locus (Cdk1Cdk2KI). Substitution of both copies of Cdk1 by Cdk2 led to early embryonic lethality, even though Cdk2 was expressed from the Cdk1 locus. In addition, we generated Cdk2-/- Cdk1+/Cdk2KI mice in which one copy of Cdk2 and one copy of Cdk1 were expressed from the Cdk1 locus and the Cdk2 gene was deleted from the endogenous Cdk2 locus. We found that both male and female Cdk2-/- Cdk1+/Cdk2KI mice were sterile, similar to Cdk2-/- mice, even though they expressed the Cdk2 protein from the Cdk1 locus in testes. The translocational and cell cycle properties of knockin Cdk2 in Cdk2-/- Cdk1+/Cdk2KI cells were comparable to those of endogenous Cdk2, but we detected premature transcriptional activation of Cdk1 during liver regeneration in the absence of Cdk2. This study provides evidence of the molecular differences between Cdk2 and Cdk1 and highlights that the timing of transcriptional activation and the genetic locus play important roles in determining the function of Cdk proteins in vivo.
Collapse
Affiliation(s)
- Ande Satyanarayana
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute-Frederick, Bldg. 560/22-56, 1050 Boyles Street, Frederick, MD 21702-1201, USA
| | | | | | | | | | | |
Collapse
|
45
|
Kehoe SM, Oka M, Hankowski KE, Reichert N, Garcia S, McCarrey JR, Gaubatz S, Terada N. A conserved E2F6-binding element in murine meiosis-specific gene promoters. Biol Reprod 2008; 79:921-30. [PMID: 18667754 DOI: 10.1095/biolreprod.108.067645] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
During gametogenesis, germ cells must undergo meiosis in order to become viable haploid gametes. Successful completion of this process is dependent upon the expression of genes whose protein products function specifically in meiosis. Failure to express these genes in meiotic cells often results in infertility, whereas aberrant expression in somatic cells may lead to mitotic catastrophe. The mechanisms responsible for regulating the timely expression of meiosis-specific genes have not been fully elucidated. Here we demonstrate that E2F6, a member of the E2F family of transcription factors, is essential for the repression of the newly identified meiosis-specific gene, Slc25a31 (also known as Ant4, Aac4), in somatic cells. This discovery, along with previous studies, prompted us to investigate the role of E2F6 in the regulation of meiosis-specific genes in general. Interestingly, the core E2F6-binding element (TCCCGC) was highly conserved in the proximal promoter regions of 19 out of 24 (79.2%) meiosis-specific genes. This was significantly higher than the frequency found in the promoters of all mouse genes (15.4%). In the absence of E2F6, only a portion of these meiosis-specific genes was derepressed in somatic cells. However, endogenous E2F6 bound to the promoters of these meiosis-specific genes regardless of whether they required E2F6 for their repression in somatic cells. Further, E2F6 overexpression was capable of reducing their transcription. These findings indicate that E2F6 possesses a broad ability to bind to and regulate the meiosis-specific gene population.
Collapse
Affiliation(s)
- Sarah M Kehoe
- Department of Pathology, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Karpurapu M, Wang D, Singh NK, Li Q, Rao GN. NFATc1 targets cyclin A in the regulation of vascular smooth muscle cell multiplication during restenosis. J Biol Chem 2008; 283:26577-90. [PMID: 18667424 DOI: 10.1074/jbc.m800423200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Platelet-derived growth factor BB (PDGF-BB) induced cyclin A expression and CDK2 activity in vascular smooth muscle cells (VSMC). Inhibition of nuclear factors of activated T cell (NFAT) activation by cyclosporin A (CsA) and VIVIT suppressed PDGF-BB-induced cyclin A expression and CDK2 activity, resulting in blockade of VSMC in the G(1) phase. In addition, CsA- and VIVIT-mediated inhibition of NFATs and small interfering RNA-targeted down-regulation of cyclin A levels suppressed PDGF-BB-induced VSMC DNA synthesis. PDGF-BB also induced cyclin A mRNA levels in VSMC in an NFAT-dependent manner. Cloning and bioinformatic analysis of rat cyclin A promoter revealed the presence of NFAT-binding elements, and PDGF-BB induced the binding of NFATs to these regulatory sequences in a CsA- and VIVIT-sensitive manner. Chromatin immunoprecipitation analysis showed that NFATc1 binds to the cyclin A promoter in response to PDGF-BB in a VIVIT-sensitive manner. Furthermore, PDGF-BB induced cyclin A promoter-luciferase reporter gene activity in VSMC, and it was inhibited by both CsA and VIVIT. Balloon injury induced cyclin A expression and CDK2 activity in rat carotid arteries, and these responses were also blocked by VIVIT. In addition, VIVIT attenuated balloon injury-induced SMC proliferation, resulting in reduced restenosis. Down-regulation of NFATc1 by its small interfering RNA inhibited PDGF-BB-induced cyclin A expression and DNA synthesis both in rat and human VSMC. Together, these findings demonstrate that the cyclin A-CDK2 complex may be a potential effector of NFATs, specifically NFATc1, in mediating SMC multiplication leading to neointima formation. Therefore, NFATs may be used as target molecules for the development of therapeutic agents against vascular diseases such as restenosis.
Collapse
Affiliation(s)
- Manjula Karpurapu
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | |
Collapse
|
47
|
Hofmann MC. Gdnf signaling pathways within the mammalian spermatogonial stem cell niche. Mol Cell Endocrinol 2008; 288:95-103. [PMID: 18485583 PMCID: PMC2491722 DOI: 10.1016/j.mce.2008.04.012] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 04/21/2008] [Accepted: 04/21/2008] [Indexed: 12/12/2022]
Abstract
Mammalian spermatogenesis is a complex process in which male germ-line stem cells develop to ultimately form spermatozoa. Spermatogonial stem cells, or SSCs, are found in the basal compartment of the seminiferous epithelium. They self-renew to maintain the pool of stem cells throughout life, or they differentiate to generate a large number of germ cells. A balance between SSC self-renewal and differentiation in the adult testis is therefore essential to maintain normal spermatogenesis and fertility. Maintenance and self-renewal are tightly regulated by extrinsic signals from the surrounding microenvironment, called the spermatogonial stem cell niche. By physically supporting the SSCs and providing them with growth factors, the Sertoli cell is the main component of the niche. In addition, adhesion molecules that connect the SSCs to the basement membrane and cellular components of the interstitium between the seminiferous tubules are important regulators of the niche function. This review mainly focuses on glial cell line-derived neurotrophic factor (Gdnf), which is produced by Sertoli cells to maintain SSCs self-renewal, and the downstream signaling pathways induced by this crucial growth factor. Interactions between Gdnf and other signaling pathways that maintain self-renewal, as well as the role of novel SSC- and Sertoli cell-specific transcription factors, are also discussed.
Collapse
Affiliation(s)
- Marie-Claude Hofmann
- Department of Veterinary Biosciences, College of Veterinary Medicine, and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States.
| |
Collapse
|
48
|
Yu Q, Wu J. Involvement of cyclins in mammalian spermatogenesis. Mol Cell Biochem 2008; 315:17-24. [PMID: 18470654 DOI: 10.1007/s11010-008-9783-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 04/30/2008] [Indexed: 11/29/2022]
Abstract
Mammalian spermatogenesis is a complicated developmental process by which undifferentiated germ cells continuously produce mature sperm throughout a lifetime. Stringent control of the cell cycle during spermatogenesis is required to ensure self-renewal of male germ line cells and differentiation of appropriate numbers of cells for the various lineages. Cyclins are key factors of cell cycle regulation and play crucial roles in governing both the mitotic and meiotic divisions that characterize spermatogenesis. Abnormal expression of some types of cyclins in the testes can induce apoptosis, infertility, testicular tumors, and other problems related to spermatogenesis in mammals. In this review, available data regarding cellular and molecular regulation of several different types of cyclins during mammalian spermatogenesis are collected and further discussed.
Collapse
Affiliation(s)
- Qingsheng Yu
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai, China
| | | |
Collapse
|
49
|
Sartini BL, Wang H, Wang W, Millette CF, Kilpatrick DL. Pre-Messenger RNA Cleavage Factor I (CFIm): Potential Role in Alternative Polyadenylation During Spermatogenesis1. Biol Reprod 2008; 78:472-82. [DOI: 10.1095/biolreprod.107.064774] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
50
|
|