1
|
Buglak DB, Holmes KHM, Galletta BJ, Rusan NM. The proximal centriole-like structure maintains nucleus-centriole architecture in sperm. J Cell Sci 2024; 137:jcs262311. [PMID: 39166297 PMCID: PMC11423811 DOI: 10.1242/jcs.262311] [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: 05/30/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Proper connection between the sperm head and tail is critical for sperm motility and fertilization. Head-tail linkage is mediated by the head-tail coupling apparatus (HTCA), which secures the axoneme (tail) to the nucleus (head). However, the molecular architecture of the HTCA is poorly understood. Here, we use Drosophila to investigate formation and remodeling of the HTCA throughout spermiogenesis by visualizing key components of this complex. Using structured illumination microscopy, we demonstrate that key HTCA proteins Spag4 and Yuri form a 'centriole cap' that surrounds the centriole (or basal body) as it invaginates into the surface of the nucleus. As development progresses, the centriole is laterally displaced to the side of the nucleus while the HTCA expands under the nucleus, forming what we term the 'nuclear shelf'. We next show that the proximal centriole-like (PCL) structure is positioned under the nuclear shelf, functioning as a crucial stabilizer of centriole-nucleus attachment. Together, our data indicate that the HTCA is a complex, multi-point attachment site that simultaneously engages the PCL, the centriole and the nucleus to ensure proper head-tail connection during late-stage spermiogenesis.
Collapse
Affiliation(s)
- Danielle B. Buglak
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kathleen H. M. Holmes
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian J. Galletta
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nasser M. Rusan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Kage A, Takahashi K, Nozaki H, Higashiyama T, Baba SA, Nishizaka T. Swimming ability and flagellar motility of sperm packets of the volvocine green alga Pleodorina starrii. PLoS One 2024; 19:e0287561. [PMID: 39024288 PMCID: PMC11257277 DOI: 10.1371/journal.pone.0287561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Eukaryotic flagella collectively form metachronal waves that facilitate the ability to cause flow or swim. Among such flagellated and planktonic swimmers, large volvocine genera such as Eudorina, Pleodorina and Volvox form bundles of small male gametes (sperm) called "sperm packets" for sexual reproduction. Although these sperm packets reportedly have flagella and the ability to swim, previous studies on volvocine motility have focused on asexual forms and the swimming characteristics of sperm packets remain unknown. However, it is important to quantify the motility of sperm packets and sperm in order to gain insights into the significance of motility in the sexual reproduction of planktonic algae. In this study, we quantitatively described the behavior of three flagellated forms of a male strain of Pleodorina starrii-asexual colonies, sperm packets, and single dissociated sperm-with emphasis on comparison of the two multicellular forms. Despite being smaller, sperm packets swam approximately 1.4 times faster than the asexual colonies of the same male strain. Body length was approximately 0.5 times smaller in the sperm packets than in asexual colonies. The flagella from sperm packets and asexual colonies showed asymmetric waveforms, whereas those from dissociated single sperm showed symmetric waveforms, suggesting the presence of a switching mechanism between sperm packets and dissociated sperm. Flagella from sperm packets were approximately 0.5 times shorter and had a beat period approximately twice as long as those from asexual colonies. The flagella of sperm packets were densely distributed over the anterior part of the body, whereas the flagella of asexual colonies were sparse and evenly distributed. The distribution of flagella, but not the number of flagella, appear to illustrate a significant difference in the speeds of sperm packets and asexual colonies. Our findings reveal novel aspects of the regulation of eukaryotic flagella and shed light on the role of flagellar motility in sexual reproduction of planktonic algae.
Collapse
Affiliation(s)
- Azusa Kage
- Department of Physics, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Kohei Takahashi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hisayoshi Nozaki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tetsuya Higashiyama
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shoji A. Baba
- Department of Biology, Ochanomizu University, Bunkyo-ku, Tokyo, Japan
| | | |
Collapse
|
3
|
Guseva EA, Buev VS, Mirzaeva SE, Pletnev PI, Dontsova OA, Sergiev PV. Structure and Composition of Spermatozoa Fibrous Sheath in Diverse Groups of Metazoa. Int J Mol Sci 2024; 25:7663. [PMID: 39062905 PMCID: PMC11276731 DOI: 10.3390/ijms25147663] [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: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
The proper functioning and assembly of the sperm flagella structures contribute significantly to spermatozoa motility and overall male fertility. However, the fine mechanisms of assembly steps are poorly studied due to the high diversity of cell types, low solubility of the corresponding protein structures, and high tissue and cell specificity. One of the open questions for investigation is the attachment of longitudinal columns to the doublets 3 and 8 of axonemal microtubules through the outer dense fibers. A number of mutations affecting the assembly of flagella in model organisms are known. Additionally, evolutionary genomics data and comparative analysis of flagella morphology are available for a set of non-model species. This review is devoted to the analysis of diverse ultrastructures of sperm flagellum of Metazoa combined with an overview of the evolutionary distribution and function of the mammalian fibrous sheath proteins.
Collapse
Affiliation(s)
- Ekaterina A. Guseva
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 143025 Skolkovo, Russia; (E.A.G.); (O.A.D.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.S.B.); (S.E.M.); (P.I.P.)
| | - Vitaly S. Buev
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.S.B.); (S.E.M.); (P.I.P.)
- Faculty of Bioengeneering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Sabina E. Mirzaeva
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.S.B.); (S.E.M.); (P.I.P.)
| | - Philipp I. Pletnev
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.S.B.); (S.E.M.); (P.I.P.)
| | - Olga A. Dontsova
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 143025 Skolkovo, Russia; (E.A.G.); (O.A.D.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.S.B.); (S.E.M.); (P.I.P.)
- Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Petr V. Sergiev
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 143025 Skolkovo, Russia; (E.A.G.); (O.A.D.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Faculty of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (V.S.B.); (S.E.M.); (P.I.P.)
| |
Collapse
|
4
|
Yazdan Parast F, Veeraragavan S, Gaikwad AS, Powar S, Prabhakar R, O'Bryan MK, Nosrati R. Viscous Loading Regulates the Flagellar Energetics of Human and Bull Sperm. SMALL METHODS 2024; 8:e2300928. [PMID: 38135876 DOI: 10.1002/smtd.202300928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/04/2023] [Indexed: 12/24/2023]
Abstract
The viscoelastic properties of the female reproductive tract influence sperm swimming behavior, but the exact role of these rheological changes in regulating sperm energetics remains unknown. Using high-speed dark-field microscopy, the flagellar dynamics of free-swimming sperm across a physiologically relevant range of viscosities is resolved. A transition from 3D to 2D slither swimming under an increased viscous loading is revealed, in the absence of any geometrical or chemical stimuli. This transition is species-specific, aligning with viscosity variations within each species' reproductive tract. Despite substantial drag increase, 2D slithering sperm maintain a steady swimming speed across a wide viscosity range (20-250 and 75-1000 mPa s for bull and human sperm) by dissipating over sixfold more energy into the fluid without elevating metabolic activity, potentially by altering the mechanisms of dynein motor activity. This energy-efficient motility mode is ideally suited for the viscous environment of the female reproductive tract.
Collapse
Affiliation(s)
- Farin Yazdan Parast
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Shibani Veeraragavan
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Avinash S Gaikwad
- Institute of Reproductive Genetics, University of Münster, 48149, Münster, Germany
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Sushant Powar
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Ranganathan Prabhakar
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Institute, Faculty of Science, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Reza Nosrati
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
5
|
Houston BJ, Merriner DJ, Stathatos GG, Nguyen JH, O'Connor AE, Lopes AM, Conrad DF, Baker M, Dunleavy JE, O'Bryan MK. Genetic mutation of Cep76 results in male infertility due to abnormal sperm tail composition. Life Sci Alliance 2024; 7:e202302452. [PMID: 38570187 PMCID: PMC10992998 DOI: 10.26508/lsa.202302452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
The transition zone is a specialised gate at the base of cilia/flagella, which separates the ciliary compartment from the cytoplasm and strictly regulates protein entry. We identified a potential new regulator of the male germ cell transition zone, CEP76. We demonstrated that CEP76 was involved in the selective entry and incorporation of key proteins required for sperm function and fertility into the ciliary compartment and ultimately the sperm tail. In the mutant, sperm tails were shorter and immotile as a consequence of deficits in essential sperm motility proteins including DNAH2 and AKAP4, which accumulated at the sperm neck in the mutant. Severe annulus, fibrous sheath, and outer dense fibre abnormalities were also detected in sperm lacking CEP76. Finally, we identified that CEP76 dictates annulus positioning and structure. This study suggests CEP76 as a male germ cell transition zone protein and adds further evidence to the hypothesis that the spermatid transition zone and annulus are part of the same functional structure.
Collapse
Affiliation(s)
- Brendan J Houston
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - D Jo Merriner
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - G Gemma Stathatos
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Joseph H Nguyen
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Anne E O'Connor
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Alexandra M Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology & Immunology, University of Porto, Porto, Portugal
| | - Donald F Conrad
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Mark Baker
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Jessica Em Dunleavy
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Moira K O'Bryan
- https://ror.org/01ej9dk98 School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| |
Collapse
|
6
|
Liu Y, Li T, Shi M, Wan Y, Li H, Zhang M, Wang Z, Wang S, Lv Y, Lu G, Liu H, Zhang H, Huang T. MORN2 regulates the morphology and energy metabolism of mitochondria and is required for male fertility in mice. J Transl Med 2024; 22:240. [PMID: 38443933 PMCID: PMC10916217 DOI: 10.1186/s12967-024-05010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Mitochondria produce adenosine triphosphate through respiratory activities to power sperm differentiation and motility, and decreased mitochondrial respiratory activity can result in poor sperm motility and asthenospermia. The mitochondrial sheath is a component of the mid-piece of the sperm flagellum, and dysfunction of the sheath can reduce sperm motility and cause male infertility. The membrane occupation and recognition nexus-motif protein 2 (MORN2) is testis enriched in mice, and the MORN motif was reported to play a role in the regulation of bioelectrical signal homeostasis in cardiomyocytes. METHODS We generated Morn2-/- mice using CRISPR/Cas9 and evaluated the potential functions of MORN2 in spermiogenesis through histological analysis, fertility examination, RT-PCR, CASA, immunofluorescence, TUNEL, electron microscopy analysis, mitochondrial energy metabolism analysis, etc. RESULTS: The Morn2-/- mice were infertile, and their sperm showed severe motility defects. Morn2-/- sperm also had abnormal morphology characterized by bent heads, aberrant mitochondrial sheath formation, lower mitochondrial membrane potential, higher levels of reactive oxygen species, and decreased mitochondrial respiratory activity. CONCLUSIONS Our study demonstrates that MORN2 is essential for male fertility and indicates that MORN2 functions in mitochondrial sheath formation and regulates mitochondrial respiratory activity.
Collapse
Affiliation(s)
- Yining Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Tongtong Li
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Mingze Shi
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Yanling Wan
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Hanzhen Li
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Mingyu Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Ziqi Wang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Shiyu Wang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
| | - Yue Lv
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, 250012, Shandong, China
- CUHK-SDU Joint Laboratory On Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Lu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- CUHK-SDU Joint Laboratory On Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- CUHK-SDU Joint Laboratory On Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Haobo Zhang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Center for Reproductive Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| | - Tao Huang
- Center for Reproductive Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
| |
Collapse
|
7
|
Antonouli S, Di Nisio V, Messini C, Samara M, Salumets A, Daponte A, Anifandis G. Sperm plasma membrane ion transporters and male fertility potential: A perspective under the prism of cryopreservation. Cryobiology 2024; 114:104845. [PMID: 38184269 DOI: 10.1016/j.cryobiol.2023.104845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Intracellular calcium homeostasis plays a crucial role in spermatozoa by regulating physiological functions associated with sperm quality and male fertility potential. Intracellular calcium fine balance in the sperm cytoplasm is strictly dependent on sperm surface channels including the CatSper channel. CatSpers' role is to ensure the influx of extracellular calcium, while intracellular pH alkalinization serves as a stimulus for the activation of several channels, including CatSper. Overall, the generation of intracellular calcium spikes through CatSper is essential for fertilization-related processes, such as sperm hyperactivation, acrosome reaction, egg chemotaxis, and zona pellucida penetration. Multiple lines of evidence suggest that disruption in the close interaction among ions, pH, and CatSper could impair male fertility potential. In contemporary times, the growing reliance on Medically Assisted Reproduction procedures underscores the impact of cryopreservation on gametes. In fact, a large body of literature raises concerns about the cryo-damages provoked by the freeze-thawing processes, that can affect the plasma membrane integrity, thus the structure of pivotal ion channels, and the fine regulation of both intracellular calcium and pH. This review aims to provide an overview of the importance of the CatSper channel in sperm quality and further fertilization potential. Additionally, it addresses the emerging issue of cryopreservation's impact on the functionality of this sperm channel.
Collapse
Affiliation(s)
- Sevastiani Antonouli
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece.
| | - Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden.
| | - Christina Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece.
| | - Maria Samara
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece.
| | - Andres Salumets
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Stockholm, Sweden; Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia; Competence Centre on Health Technologies, Tartu, Estonia.
| | - Alexandros Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece.
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece.
| |
Collapse
|
8
|
Shimada K, Lu Y, Ikawa M. Disruption of testis-enriched cytochrome c oxidase subunit COX6B2 but not COX8C leads to subfertility. Exp Anim 2024; 73:1-10. [PMID: 37423748 PMCID: PMC10877148 DOI: 10.1538/expanim.23-0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023] Open
Abstract
Mammalian sperm flagellum contains the midpiece characterized by a mitochondrial sheath that packs tightly around the axoneme and outer dense fibers. Mitochondria are known as the "powerhouse" of the cell, and produce ATP through the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS). However, the contribution of the TCA cycle and OXPHOS to sperm motility and male fertility is less clear. Cytochrome c oxidase (COX) is an oligomeric complex localized within the mitochondrial inner membrane, and the terminal enzyme of the mitochondrial electron transport chain in eukaryotes. Both COX6B2 and COX8C are testis-enriched COX subunits whose functions in vivo are poorly studied. Here, we generated Cox6b2 and Cox8c knockout (KO) mice using the CRISPR/Cas9 system. We examined their fertility and sperm mitochondrial function to determine the significance of testis-enriched COX subunits in male fertility. The mating test revealed that disrupting COX6B2 induces male subfertility, while disrupting COX8C does not affect male fertility. Cox6b2 KO spermatozoa showed low sperm motility, but mitochondrial function was normal according to oxygen consumption rates. Therefore, low sperm motility seems to cause subfertility in Cox6b2 KO male mice. These results also indicate that testis-enriched COX, COX6B2 and COX8C, are not essential for OXPHOS in mouse spermatozoa.
Collapse
Affiliation(s)
- Keisuke Shimada
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Yonggang Lu
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
9
|
Bashiri Z, Movahedin M, Pirhajati V, Asgari H, Koruji M. Ultrastructural study: in vitro and in vivo differentiation of mice spermatogonial stem cells. ZYGOTE 2024; 32:87-95. [PMID: 38149356 DOI: 10.1017/s096719942300062x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Mouse testicular tissue is composed of seminiferous tubules and interstitial tissue. Mammalian spermatogenesis is divided into three stages: spermatocytogenesis (mitotic divisions) in which spermatogonial stem cells (SSCs) turn into spermatocytes, followed by two consecutive meiotic divisions in which spermatocytes form spermatids. Spermatids differentiate into spermatozoa during spermiogenesis. Various factors affect the process of spermatogenesis and the organization of cells in the testis. Any disorder in different stages of spermatogenesis will have negative effects on male fertility. The aim of the current study was to compare the in vitro and in vivo spermatogenesis processes before and after transplantation to azoospermic mice using ultrastructural techniques. In this study, mice were irradiated with single doses of 14 Gy 60Co radiation. SSCs isolated from neonatal mice were cultured in vitro for 1 week and were injected into the seminiferous tubule recipient's mice. Testicular cells of neonatal mice were cultured in the four groups on extracellular matrix-based 3D printing scaffolds. The transplanted testes (8 weeks after transplantation) and cultured testicular cells in vitro (after 3 weeks) were then processed for transmission electron microscopy studies. Our study's findings revealed that the morphology and ultrastructure of testicular cells after transplantation and in vitro culture are similar to those of in vivo spermatogenesis, indicating that spermatogenic cell nature is unaltered in vitro.
Collapse
Affiliation(s)
- Zahra Bashiri
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Mansoureh Movahedin
- Department of Anatomical Sciences, Medical Sciences Faculty, Tarbiat Modares University, Tehran, Iran
| | - Vahid Pirhajati
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Asgari
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Subbiah A, Caswell DL, Turner K, Jaiswal A, Avidor-Reiss T. CP110 and CEP135 Localize Near the Proximal Centriolar Remnants of Mice Spermatozoa. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001083. [PMID: 38351906 PMCID: PMC10862134 DOI: 10.17912/micropub.biology.001083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Centrioles form centrosomes that organize microtubules, assist in cell structure, and nucleate cilia that provide motility and sensation. Within the sperm, the centrosome consists of two centrioles (proximal and distal centriole) and a pericentriolar material known as the striated column and capitulum. The distal centriole nucleates the flagellum. Mice spermatozoa, unlike other mammal spermatozoa (e.g., human and bovine), have no ultra-structurally recognizable centrioles, but their neck has the centriolar proteins POC1B and FAM161A, suggesting mice spermatozoa have remnant centrioles. Here, we examine whether other centriolar proteins, CP110 and CEP135, found in the human and bovine spermatozoa centrioles are also found in the mouse spermatozoa neck. CP110 is a tip protein controlling ciliogenesis, and CEP135 is a centriole-specific structural protein in the centriole base of canonical centrioles found in most cell types. Here, we report that CP110 and CEP135 were both located in the mice spermatozoa neck around the proximal centriolar remnants labeled by POC1B, increasing the number of centriolar proteins found in the mice spermatozoa neck, further supporting the hypothesis that a remnant proximal centriole is present in mice.
Collapse
|
11
|
Khalphallah A, Al-Daek T, Abdelhamid M, Elmeligy E, El-Hawari SF, Khesruf KA, Nasr HA, Mohamed RH. Camel filariasis (Dipetalonema evansi) and its association with clinical balanoposthitis with reference to prominent changes in clinical findings, serum testosterone, semen analysis, and testicular histopathology. BMC Vet Res 2024; 20:1. [PMID: 38172872 PMCID: PMC10763322 DOI: 10.1186/s12917-023-03844-5] [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: 04/25/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Camel filariasis induced variable clinical syndromes characterized by fever, lethargy, localized dermal lesions, loss of condition, and testicular and scrotal swelling. The objective of the present work focused on clarifying the diagnostic importance of clinical findings, serum testosterone, and semen analysis as well as blood smear and testicular histopathology as a differential tool between only balanoposthitis without filariasis male camels group (OnlyBpgr) and balanoposthitis-filariasis infected male camels group (BpFlgr). The study also monitored the associations between the severity of ticks' infestations in investigated male camels and the occurrence of balanoposthitis only or balanoposthitis with filariasis. RESULTS AND CONCLUSIONS The study reported significant correlation between serum testosterone, serum cortisol, and sperm vitality and abnormalities percentages. The study included male camels (n = 250) classified into three groups: healthy control group (Contgr; n = 30), OnlyBpgr (n = 210), and BpFlgr (n = 10). These male camels were clinically and laboratory examined, and skin scraping tests and testicular histopathology were conducted. The study confirmed the association of the changes in clinical findings, whole blood picture, serum testosterone, serum cortisol, and semen analysis, with OnlyBpgr and BpFlgr. These changes were more prominent in BpFlgr than in OnlyBpgr. Skin scraping test results revealed a higher severity of live ticks' infestation in BpFlgr than in OnlyBpgr because, unlike OnlyBpgr, all camels in BpFlgr (n = 10) were suffering from live ticks' infestation. It also concluded the higher efficacy of histopathology of testicular tissues in male camels as a diagnostic tool for adult filaria in balanoposthitis-affected male camels than blood smear because all cases of camel filariasis in the current work were negative for microfilaria on microscopic examination of diurnal blood smear as well as testicular histopathology revealed detection of adult filaria in all camel filariasis associated with balanoposthitis. Strong correlation relationships were demonstrated between serum testosterone, serum cortisol, and semen analysis results. Positive correlations were reported between serum testosterone levels and sperm vitality percentages. However, negative correlations were stated between serum testosterone and each of serum cortisol and sperm abnormalities either in Contgr, OnlyBpgr, or BpFlgr.
Collapse
Affiliation(s)
- Arafat Khalphallah
- Division of Internal Medicine, Department of Animal Medicine, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Taher Al-Daek
- Faculty of Veterinary Medicine, Omar Al-Mukhtar University, Al-bayda, 919, Libya
| | - Mahmoud Abdelhamid
- Department of Parasitology, Faculty of Veterinary Medicine, Aswan University, Aswan, 81528, Egypt
| | - Enas Elmeligy
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Sayed Fathi El-Hawari
- Department of clinical studies, Collage of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Khaled A Khesruf
- Department of Animal Diseases, Faculty of Veterinary Medicine, Aleppo University, Aleppo, Syria
| | - Heba A Nasr
- Division of Clinical Laboratory Diagnosis, Department of Animal Medicine, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Ragab H Mohamed
- Department of Theriogenology, Obstetrics, and Artificial Insemination, Faculty of Veterinary Medicine, Aswan University, Aswan, 81528, Egypt
| |
Collapse
|
12
|
Khanal S, Jaiswal A, Chowdanayaka R, Puente N, Turner K, Assefa KY, Nawras M, Back ED, Royfman A, Burkett JP, Cheong SH, Fisher HS, Sindhwani P, Gray J, Ramachandra NB, Avidor-Reiss T. The evolution of centriole degradation in mouse sperm. Nat Commun 2024; 15:117. [PMID: 38168044 PMCID: PMC10761967 DOI: 10.1038/s41467-023-44411-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 12/12/2023] [Indexed: 01/05/2024] Open
Abstract
Centrioles are subcellular organelles found at the cilia base with an evolutionarily conserved structure and a shock absorber-like function. In sperm, centrioles are found at the flagellum base and are essential for embryo development in basal animals. Yet, sperm centrioles have evolved diverse forms, sometimes acting like a transmission system, as in cattle, and sometimes becoming dispensable, as in house mice. How the essential sperm centriole evolved to become dispensable in some organisms is unclear. Here, we test the hypothesis that this transition occurred through a cascade of evolutionary changes to the proteins, structure, and function of sperm centrioles and was possibly driven by sperm competition. We found that the final steps in this cascade are associated with a change in the primary structure of the centriolar inner scaffold protein FAM161A in rodents. This information provides the first insight into the molecular mechanisms and adaptive evolution underlying a major evolutionary transition within the internal structure of the mammalian sperm neck.
Collapse
Affiliation(s)
- Sushil Khanal
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Ankit Jaiswal
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Rajanikanth Chowdanayaka
- Department of Studies in Genetics and Genomics, University of Mysore, Manasagangotri, Mysuru, India
| | - Nahshon Puente
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Katerina Turner
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | | | - Mohamad Nawras
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Ezekiel David Back
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Abigail Royfman
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - James P Burkett
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Soon Hon Cheong
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Heidi S Fisher
- Department of Biology, University of Maryland College Park, College Park, MD, USA
| | - Puneet Sindhwani
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - John Gray
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | | | - Tomer Avidor-Reiss
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA.
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
13
|
Ma Y, Wu B, Chen Y, Ma S, Wang L, Han T, Lin X, Yang F, Liu C, Zhao J, Li W. CCDC146 is required for sperm flagellum biogenesis and male fertility in mice. Cell Mol Life Sci 2023; 81:1. [PMID: 38038747 PMCID: PMC11072088 DOI: 10.1007/s00018-023-05025-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/19/2023] [Accepted: 10/28/2023] [Indexed: 12/02/2023]
Abstract
Multiple morphological abnormalities of the flagella (MMAF) is a severe disease of male infertility, while the pathogenetic mechanisms of MMAF are still incompletely understood. Previously, we found that the deficiency of Ccdc38 might be associated with MMAF. To understand the underlying mechanism of this disease, we identified the potential partner of this protein and found that the coiled-coil domain containing 146 (CCDC146) can interact with CCDC38. It is predominantly expressed in the testes, and the knockout of this gene resulted in complete infertility in male mice but not in females. The knockout of Ccdc146 impaired spermiogenesis, mainly due to flagellum and manchette organization defects, finally led to MMAF-like phenotype. Furthermore, we demonstrated that CCDC146 could interact with both CCDC38 and CCDC42. It also interacts with intraflagellar transport (IFT) complexes IFT88 and IFT20. The knockout of this gene led to the decrease of ODF2, IFT88, and IFT20 protein levels, but did not affect CCDC38, CCDC42, or ODF1 expression. Additionally, we predicted and validated the detailed interactions between CCDC146 and CCDC38 or CCDC42, and built the interaction models at the atomic level. Our results suggest that the testis predominantly expressed gene Ccdc146 is essential for sperm flagellum biogenesis and male fertility, and its mutations might be associated with MMAF in some patients.
Collapse
Affiliation(s)
- Yanjie Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bingbing Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yinghong Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liying Wang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Tingting Han
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Xiaolei Lin
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Fulin Yang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing, 100101, China.
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, No. 9 Jinsui Road, Tianhe District, Guangzhou, 510623, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
14
|
Tai L, Yin G, Huang X, Sun F, Zhu Y. In-cell structural insight into the stability of sperm microtubule doublet. Cell Discov 2023; 9:116. [PMID: 37989994 PMCID: PMC10663601 DOI: 10.1038/s41421-023-00606-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/21/2023] [Indexed: 11/23/2023] Open
Abstract
The propulsion for mammalian sperm swimming is generated by flagella beating. Microtubule doublets (DMTs) along with microtubule inner proteins (MIPs) are essential structural blocks of flagella. However, the intricate molecular architecture of intact sperm DMT remains elusive. Here, by in situ cryo-electron tomography, we solved the in-cell structure of mouse sperm DMT at 4.5-7.5 Å resolutions, and built its model with 36 kinds of MIPs in 48 nm periodicity. We identified multiple copies of Tektin5 that reinforce Tektin bundle, and multiple MIPs with different periodicities that anchor the Tektin bundle to tubulin wall. This architecture contributes to a superior stability of A-tubule than B-tubule of DMT, which was revealed by structural comparison of DMTs from the intact and deformed axonemes. Our work provides an overall molecular picture of intact sperm DMT in 48 nm periodicity that is essential to understand the molecular mechanism of sperm motility as well as the related ciliopathies.
Collapse
Affiliation(s)
- Linhua Tai
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Guoliang Yin
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaojun Huang
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Fei Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China.
| | - Yun Zhu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
15
|
Corkidi G, Montoya F, González-Cota AL, Hernández-Herrera P, Bruce NC, Bloomfield-Gadêlha H, Darszon A. Human sperm rotate with a conserved direction during free swimming in four dimensions. J Cell Sci 2023; 136:jcs261306. [PMID: 37902031 PMCID: PMC10729817 DOI: 10.1242/jcs.261306] [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: 05/11/2023] [Accepted: 10/23/2023] [Indexed: 10/31/2023] Open
Abstract
Head rotation in human spermatozoa is essential for different swimming modes and fertilisation, as it links the molecular workings of the flagellar beat with sperm motion in three-dimensional (3D) space over time. Determining the direction of head rotation has been hindered by the symmetry and translucent nature of the sperm head, and by the fast 3D motion driven by the helical flagellar beat. Analysis has been mostly restricted to two-dimensional (2D) single focal plane image analysis, which enables tracking of head centre position but not tracking of head rotation. Despite the conserved helical beating of the human sperm flagellum, human sperm head rotation has been reported to be uni- or bi-directional, and even to intermittently change direction in a given cell. Here, we directly measure the head rotation of freely swimming human sperm using multi-plane 4D (3D+t) microscopy and show that: (1) 2D microscopy is unable to distinguish head rotation direction in human spermatozoa; (2) head rotation direction in non-capacitating and capacitating solutions, for both aqueous and viscous media, is counterclockwise (CCW), as seen from head to tail, in all rotating spermatozoa, regardless of the experimental conditions; and (3) head rotation is suppressed in 36% of spermatozoa swimming in non-capacitating viscous medium, although CCW rotation is recovered after incubation in capacitating conditions within the same viscous medium, possibly unveiling an unexplored aspect of the essential need of capacitation for fertilisation. Our observations show that the CCW head rotation in human sperm is conserved. It constitutes a robust and persistent helical driving mechanism that influences sperm navigation in 3D space over time, and thus is of critical importance in cell motility, propulsion of flagellated microorganisms, sperm motility assessments, human reproduction research, and self-organisation of flagellar beating patterns and swimming in 3D space.
Collapse
Affiliation(s)
- Gabriel Corkidi
- Laboratorio de Imágenes y Visión por Computadora, Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, México
| | - Fernando Montoya
- Laboratorio de Imágenes y Visión por Computadora, Departamento de Ingeniería Celular y Biocatálisis, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, México
| | - Ana L. González-Cota
- Departamento de Genética del Desarrollo y Fisiología Molecular and Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, México
| | - Paul Hernández-Herrera
- Laboratorio Nacional de Microscopía Avanzada, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, México
| | - Neil C. Bruce
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México, Circuito Exterior S/N, Ciudad Universitaria, 04510 Ciudad de México, México
| | - Hermes Bloomfield-Gadêlha
- School of Engineering Mathematics and Technology & Bristol Robotics Laboratory, University of Bristol, Bristol BS8 1TW, UK
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular and Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca 62210, México
| |
Collapse
|
16
|
Chen Z, Shiozaki M, Haas KM, Skinner WM, Zhao S, Guo C, Polacco BJ, Yu Z, Krogan NJ, Lishko PV, Kaake RM, Vale RD, Agard DA. De novo protein identification in mammalian sperm using in situ cryoelectron tomography and AlphaFold2 docking. Cell 2023; 186:5041-5053.e19. [PMID: 37865089 PMCID: PMC10842264 DOI: 10.1016/j.cell.2023.09.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 08/02/2023] [Accepted: 09/16/2023] [Indexed: 10/23/2023]
Abstract
To understand the molecular mechanisms of cellular pathways, contemporary workflows typically require multiple techniques to identify proteins, track their localization, and determine their structures in vitro. Here, we combined cellular cryoelectron tomography (cryo-ET) and AlphaFold2 modeling to address these questions and understand how mammalian sperm are built in situ. Our cellular cryo-ET and subtomogram averaging provided 6.0-Å reconstructions of axonemal microtubule structures. The well-resolved tertiary structures allowed us to unbiasedly match sperm-specific densities with 21,615 AlphaFold2-predicted protein models of the mouse proteome. We identified Tektin 5, CCDC105, and SPACA9 as novel microtubule-associated proteins. These proteins form an extensive interaction network crosslinking the lumen of axonemal doublet microtubules, suggesting their roles in modulating the mechanical properties of the filaments. Indeed, Tekt5 -/- sperm possess more deformed flagella with 180° bends. Together, our studies presented a cellular visual proteomics workflow and shed light on the in vivo functions of Tektin 5.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| | - Momoko Shiozaki
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Kelsey M Haas
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA; J. David Gladstone Institutes, San Francisco, CA, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Will M Skinner
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Shumei Zhao
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Caiying Guo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Benjamin J Polacco
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Zhiheng Yu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA; J. David Gladstone Institutes, San Francisco, CA, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Polina V Lishko
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Robyn M Kaake
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA; J. David Gladstone Institutes, San Francisco, CA, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA
| | - Ronald D Vale
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - David A Agard
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Luo ZY, Jiang TX, Zhang T, Xu P, Qiu XB. Ubiquitin Ligase Nrdp1 Controls Autophagy-Associated Acrosome Biogenesis and Mitochondrial Arrangement during Spermiogenesis. Cells 2023; 12:2211. [PMID: 37759433 PMCID: PMC10527437 DOI: 10.3390/cells12182211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/23/2023] [Accepted: 07/27/2023] [Indexed: 09/29/2023] Open
Abstract
Autophagy is critical to acrosome biogenesis and mitochondrial quality control, but the underlying mechanisms remain unclear. The ubiquitin ligase Nrdp1/RNF41 promotes ubiquitination of the mitophagy-associated Parkin and interacts with the pro-autophagic protein SIP/CacyBP. Here, we report that global deletion of Nrdp1 leads to formation of the round-headed sperm and male infertility by disrupting autophagy. Quantitative proteome analyses demonstrated that the expression of many proteins associated with mitochondria, lysosomes, and acrosomes was dysregulated in either spermatids or sperm of the Nrdp1-deficient mice. Deletion of Nrdp1 increased the levels of Parkin but decreased the levels of SIP, the mitochondrial fission protein Drp1 and the mitochondrial protein Tim23 in sperm, accompanied by the inhibition of autophagy, the impairment of acrosome biogenesis and the disruption of mitochondrial arrangement in sperm. Thus, our results uncover an essential role of Nrdp1 in spermiogenesis and male fertility by promoting autophagy, providing important clues to cope with the related male reproductive diseases.
Collapse
Affiliation(s)
- Zi-Yu Luo
- Ministry of Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China; (Z.-Y.L.); (T.-X.J.)
| | - Tian-Xia Jiang
- Ministry of Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China; (Z.-Y.L.); (T.-X.J.)
| | - Tao Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Institute of Lifeomics, 38 Science Park Road, Beijing 102206, China;
| | - Ping Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Institute of Lifeomics, 38 Science Park Road, Beijing 102206, China;
| | - Xiao-Bo Qiu
- Ministry of Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China; (Z.-Y.L.); (T.-X.J.)
| |
Collapse
|
18
|
Li P, Messina G, Lehner CF. Nuclear elongation during spermiogenesis depends on physical linkage of nuclear pore complexes to bundled microtubules by Drosophila Mst27D. PLoS Genet 2023; 19:e1010837. [PMID: 37428798 PMCID: PMC10359004 DOI: 10.1371/journal.pgen.1010837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023] Open
Abstract
Spermatozoa in animal species are usually highly elongated cells with a long motile tail attached to a head that contains the haploid genome in a compact and often elongated nucleus. In Drosophila melanogaster, the nucleus is compacted two hundred-fold in volume during spermiogenesis and re-modeled into a needle that is thirty-fold longer than its diameter. Nuclear elongation is preceded by a striking relocalization of nuclear pore complexes (NPCs). While NPCs are initially located throughout the nuclear envelope (NE) around the spherical nucleus of early round spermatids, they are later confined to one hemisphere. In the cytoplasm adjacent to this NPC-containing NE, the so-called dense complex with a strong bundle of microtubules is assembled. While this conspicuous proximity argued for functional significance of NPC-NE and microtubule bundle, experimental confirmation of their contributions to nuclear elongation has not yet been reported. Our functional characterization of the spermatid specific Mst27D protein now resolves this deficit. We demonstrate that Mst27D establishes physical linkage between NPC-NE and dense complex. The C-terminal region of Mst27D binds to the nuclear pore protein Nup358. The N-terminal CH domain of Mst27D, which is similar to that of EB1 family proteins, binds to microtubules. At high expression levels, Mst27D promotes bundling of microtubules in cultured cells. Microscopic analyses indicated co-localization of Mst27D with Nup358 and with the microtubule bundles of the dense complex. Time-lapse imaging revealed that nuclear elongation is accompanied by a progressive bundling of microtubules into a single elongated bundle. In Mst27D null mutants, this bundling process does not occur and nuclear elongation is abnormal. Thus, we propose that Mst27D permits normal nuclear elongation by promoting the attachment of the NPC-NE to the microtubules of the dense complex, as well as the progressive bundling of these microtubules.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Molecular Life Science (DMLS), University of Zurich, Zurich, Switzerland
| | - Giovanni Messina
- Department of Molecular Life Science (DMLS), University of Zurich, Zurich, Switzerland
| | - Christian F Lehner
- Department of Molecular Life Science (DMLS), University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Leung MR, Zeng J, Wang X, Roelofs MC, Huang W, Zenezini Chiozzi R, Hevler JF, Heck AJR, Dutcher SK, Brown A, Zhang R, Zeev-Ben-Mordehai T. Structural specializations of the sperm tail. Cell 2023; 186:2880-2896.e17. [PMID: 37327785 PMCID: PMC10948200 DOI: 10.1016/j.cell.2023.05.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/16/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Sperm motility is crucial to reproductive success in sexually reproducing organisms. Impaired sperm movement causes male infertility, which is increasing globally. Sperm are powered by a microtubule-based molecular machine-the axoneme-but it is unclear how axonemal microtubules are ornamented to support motility in diverse fertilization environments. Here, we present high-resolution structures of native axonemal doublet microtubules (DMTs) from sea urchin and bovine sperm, representing external and internal fertilizers. We identify >60 proteins decorating sperm DMTs; at least 15 are sperm associated and 16 are linked to infertility. By comparing DMTs across species and cell types, we define core microtubule inner proteins (MIPs) and analyze evolution of the tektin bundle. We identify conserved axonemal microtubule-associated proteins (MAPs) with unique tubulin-binding modes. Additionally, we identify a testis-specific serine/threonine kinase that links DMTs to outer dense fibers in mammalian sperm. Our study provides structural foundations for understanding sperm evolution, motility, and dysfunction at a molecular level.
Collapse
Affiliation(s)
- Miguel Ricardo Leung
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Jianwei Zeng
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Xiangli Wang
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Marc C Roelofs
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry & Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Johannes F Hevler
- Biomolecular Mass Spectrometry & Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry & Proteomics, Bijvoet Centre for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Susan K Dutcher
- Department of Genetics, Washington University in St. Louis, St Louis, MO, USA
| | - Alan Brown
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.
| | - Tzviya Zeev-Ben-Mordehai
- Structural Biochemistry, Bijvoet Centre for Biomolecular Research, Utrecht University, 3584 CG Utrecht, the Netherlands.
| |
Collapse
|
20
|
Gönczy P, Balestra FR. Sperm-contributed centrioles segregate stochastically into blastomeres of 4-cell stage Caenorhabditis elegans embryos. Genetics 2023; 224:iyad048. [PMID: 36988082 PMCID: PMC10158834 DOI: 10.1093/genetics/iyad048] [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: 03/06/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/30/2023] Open
Abstract
Whereas both sperm and egg contribute nuclear genetic material to the zygote in metazoan organisms, the inheritance of other cellular constituents is unequal between the 2 gametes. Thus, 2 copies of the centriole are contributed solely by the sperm to the zygote in most species. Centrioles can have a stereotyped distribution in some asymmetric divisions, but whether sperm-contributed centrioles are distributed in a stereotyped manner in the resulting embryo is not known. Here, we address this question in Caenorhabditis elegans using marked mating experiments, whereby the presence of the 2 sperm-contributed centrioles is monitored in the embryo using the stable centriolar component SAS-4::GFP, as well as GFP::SAS-7. Our analysis demonstrates that the distribution of sperm-contributed centrioles is stochastic in 4-cell stage embryos. Moreover, using sperm from zyg-1 mutant males that harbor a single centriole, we show that the older sperm-contributed centriole is likewise distributed stochastically in the resulting embryo. Overall, we conclude that, in contrast to the situation during some asymmetric cell divisions, centrioles contributed by the male germ line are distributed stochastically in embryos of C. elegans.
Collapse
Affiliation(s)
- Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | - Fernando R Balestra
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland
- Departamento de Genética, Universidad de Sevilla, Sevilla 41080, Spain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla 41092, Spain
| |
Collapse
|
21
|
Yap YT, Li W, Huang Q, Zhou Q, Zhang D, Sheng Y, Mladenovic-Lucas L, Yee SP, Orwig KE, Granneman JG, Williams DC, Hess RA, Toure A, Zhang Z. DNALI1 interacts with the MEIG1/PACRG complex within the manchette and is required for proper sperm flagellum assembly in mice. eLife 2023; 12:e79620. [PMID: 37083624 PMCID: PMC10185345 DOI: 10.7554/elife.79620] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 03/12/2023] [Indexed: 04/22/2023] Open
Abstract
The manchette is a transient and unique structure present in elongating spermatids and required for proper differentiation of the germ cells during spermatogenesis. Previous work indicated that the MEIG1/PACRG complex locates in the manchette and is involved in the transport of cargos, such as SPAG16L, to build the sperm flagellum. Here, using co-immunoprecipitation and pull-down approaches in various cell systems, we established that DNALI1, an axonemal component originally cloned from Chlamydomonas reinhardtii, recruits and stabilizes PACRG and we confirm in vivo, the co-localization of DNALI1 and PACRG in the manchette by immunofluorescence of elongating murine spermatids. We next generated mice with a specific deficiency of DNALI1 in male germ cells, and observed a dramatic reduction of the sperm cells, which results in male infertility. In addition, we observed that the majority of the sperm cells exhibited abnormal morphology including misshapen heads, bent tails, enlarged midpiece, discontinuous accessory structure, emphasizing the importance of DNALI1 in sperm differentiation. Examination of testis histology confirmed impaired spermiogenesis in the mutant mice. Importantly, while testicular levels of MEIG1, PACRG, and SPAG16L proteins were unchanged in the Dnali1 mutant mice, their localization within the manchette was greatly affected, indicating that DNALI1 is required for the formation of the MEIG1/PACRG complex within the manchette. Interestingly, in contrast to MEIG1 and PACRG-deficient mice, the DNALI1-deficient mice also showed impaired sperm spermiation/individualization, suggesting additional functions beyond its involvement in the manchette structure. Overall, our work identifies DNALI1 as a protein required for sperm development.
Collapse
Affiliation(s)
- Yi Tian Yap
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - Wei Li
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - Qian Huang
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
- Department of Occupational and Environmental Medicine, School of Public Health, Wuhan University of Science and TechnologyWuhanChina
| | - Qi Zhou
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
- Department of Occupational and Environmental Medicine, School of Public Health, Wuhan University of Science and TechnologyWuhanChina
| | - David Zhang
- College of William and MaryWilliamsburgUnited States
| | - Yi Sheng
- Molecular Genetics and Developmental Biology Graduate Program, Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Ljljiana Mladenovic-Lucas
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health CenterFarmingtonUnited States
| | - Kyle E Orwig
- Molecular Genetics and Developmental Biology Graduate Program, Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
| | - David C Williams
- Department of Pathology and Laboratory Medicine, University of North CarolinaChapel HillUnited States
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of IllinoisUrbanaUnited States
| | - Aminata Toure
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team Physiology and Pathophysiology of Sperm cells, Institute for Advanced BiosciencesGrenobleFrance
| | - Zhibing Zhang
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics & Gynecology, Wayne State UniversityDetroitUnited States
| |
Collapse
|
22
|
Chen Z, Greenan GA, Shiozaki M, Liu Y, Skinner WM, Zhao X, Zhao S, Yan R, Yu Z, Lishko PV, Agard DA, Vale RD. In situ cryo-electron tomography reveals the asymmetric architecture of mammalian sperm axonemes. Nat Struct Mol Biol 2023; 30:360-369. [PMID: 36593309 PMCID: PMC10023559 DOI: 10.1038/s41594-022-00861-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/11/2022] [Indexed: 01/04/2023]
Abstract
The flagella of mammalian sperm display non-planar, asymmetric beating, in contrast to the planar, symmetric beating of flagella from sea urchin sperm and unicellular organisms. The molecular basis of this difference is unclear. Here, we perform in situ cryo-electron tomography of mouse and human sperm, providing the highest-resolution structural information to date. Our subtomogram averages reveal mammalian sperm-specific protein complexes within the microtubules, the radial spokes and nexin-dynein regulatory complexes. The locations and structures of these complexes suggest potential roles in enhancing the mechanical strength of mammalian sperm axonemes and regulating dynein-based axonemal bending. Intriguingly, we find that each of the nine outer microtubule doublets is decorated with a distinct combination of sperm-specific complexes. We propose that this asymmetric distribution of proteins differentially regulates the sliding of each microtubule doublet and may underlie the asymmetric beating of mammalian sperm.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Garrett A Greenan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Momoko Shiozaki
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Yanxin Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Will M Skinner
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Xiaowei Zhao
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Shumei Zhao
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Rui Yan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Zhiheng Yu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Polina V Lishko
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA
| | - David A Agard
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| | - Ronald D Vale
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| |
Collapse
|
23
|
Liu W, Wei X, Liu X, Chen G, Zhang X, Liang X, Isachenko V, Sha Y, Wang Y. Biallelic mutations in ARMC12 cause asthenozoospermia and multiple midpiece defects in humans and mice. J Med Genet 2023; 60:154-162. [PMID: 35534203 DOI: 10.1136/jmedgenet-2021-108137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 04/14/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Asthenozoospermia is a major factor contributing to male infertility. The mitochondrial sheath (MS), an important organelle in the midpiece of spermatozoa, is crucial to sperm motility. ARMC12 is a mitochondrial peripheral membrane protein. Deletion of Armc12 impairs the arrangement of MS and causes infertility in mice. However, the role of ARMC12 in human asthenozoospermia remains unknown. OBJECTIVE To study the genetic defects in patients with asthenozoospermia. METHODS A total of 125 patients with asthenozoospermia and 120 men with proven fertility were recruited. Whole-exome sequencing and Sanger sequencing were performed for genetic analysis. Papanicolaou staining, HE staining, immunofluorescent staining, transmission electron microscopy and field emission scanning electron microscopy were employed to observe the morphological and structural defects of the spermatozoa and testes. Armc12-knockout mice were generated using the CRISPR-Cas9 system. Intracytoplasmic sperm injection was used to treat the patients. RESULTS Biallelic ARMC12 mutations were identified in three patients, including homozygous mutations in two siblings from a consanguineous family and compound heterozygous mutations in one sporadic patient. ARMC12 is mainly expressed in the midpiece of elongated and late spermatids in the human testis. The patients' spermatozoa displayed multiple midpiece defects, including absent MS and central pair, scattered or forked axoneme and incomplete plasma membrane. Spermatozoa from Armc12-/- mice showed parallel defects in the midpiece. Moreover, two patients were treated with intracytoplasmic sperm injection and achieved good outcomes. CONCLUSION Our findings prove for the first time that defects in ARMC12 cause asthenozoospermia and multiple midpiece defects in humans.
Collapse
Affiliation(s)
- Wensheng Liu
- Obstetrics and Gynecology Center, Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoli Wei
- School of Medicine, Yunnan University, Kunming, Yunnan, China
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyan Liu
- Reproductive Medicine Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Gaowen Chen
- Obstetrics and Gynecology Center, Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoya Zhang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xiaomei Liang
- Obstetrics and Gynecology Center, Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Vladimir Isachenko
- Research Group for Reproductive Medicine, Department of Obstetrics and Gynecology, Medical Faculty, University of Cologne, Cologne, North Rhine-Westphalia, Germany
| | - Yanwei Sha
- Department of Andrology, United Diagnostic and Research Center for Clinical Genetics, Fujian Provincial Key Laboratory of Reproductive Health Research, Women and Children's Hospital & School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yifeng Wang
- Obstetrics and Gynecology Center, Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
24
|
Liu C, Shen Y, Tang S, Wang J, Zhou Y, Tian S, Wu H, Cong J, He X, Jin L, Cao Y, Yang Y, Zhang F. Homozygous variants in AKAP3 induce asthenoteratozoospermia and male infertility. J Med Genet 2023; 60:137-143. [PMID: 35228300 DOI: 10.1136/jmedgenet-2021-108271] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 02/08/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND As a common type of asthenoteratozoospermia, multiple morphological abnormalities of the sperm flagella (MMAF) can cause male infertility. Previous studies have revealed genetic factors as a major cause of MMAF. The known MMAF-associated genes are involved in the mitochondrial sheath, outer dense fibre or axoneme of the sperm flagella. These findings indicate the genetic heterogeneity of MMAF. METHODS AND RESULTS Here, we conducted genetic analyses using whole-exome sequencing in a cohort of 150 Han Chinese men with asthenoteratozoospermia. Homozygous deleterious variants of AKAP3 (A-kinase anchoring protein 3) were identified in two MMAF-affected men from unrelated families. One AKAP3 variant was a frameshift (c.2286_2287del, p.His762Glnfs*22) and the other variant was a missense mutation (c.44G>A, p.Cys15Tyr), which was predicted to be damaging by multiple bioinformatics tools. Further western blotting and immunofluorescence assays revealed the absence of AKAP3 in the spermatozoa from the man harbouring the homozygous frameshift variant, whereas the expression of AKAP3 was markedly reduced in the spermatozoa of the man with the AKAP3 missense variant p.Cys15Tyr. Notably, the clinical outcomes after intracytoplasmic sperm injection (ICSI) were divergent between these two cases, suggesting a possibility of AKAP3 dosage-dependent prognosis of ICSI treatment. CONCLUSIONS Our study revealed AKAP3 as a novel gene involved in human asthenoteratozoospermia.
Collapse
Affiliation(s)
- Chunyu Liu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Ying Shen
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuyan Tang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Jiaxiong Wang
- Center for Reproduction and Genetics, State Key Laboratory of Reproductive Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Yiling Zhou
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Shixiong Tian
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China, Anhui Medical University, Hefei, Anhui, China
| | - Jiangshan Cong
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China, Anhui Medical University, Hefei, Anhui, China
| | - Li Jin
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Ministry of Education of the People's Republic of China, Anhui Medical University, Hefei, Anhui, China
| | - Yihong Yang
- Center of Reproductive Medicine, West China Second University Hospital, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai, China .,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Institute of Reproduction and Development, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Comparative oxidative metabolism in mammalian sperm. Anim Reprod Sci 2022; 247:107095. [DOI: 10.1016/j.anireprosci.2022.107095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022]
|
26
|
Hoyer-Fender S. Development of the Connecting Piece in ODF1-Deficient Mouse Spermatids. Int J Mol Sci 2022; 23:ijms231810280. [PMID: 36142191 PMCID: PMC9499666 DOI: 10.3390/ijms231810280] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/30/2022] [Accepted: 09/04/2022] [Indexed: 11/30/2022] Open
Abstract
ODF1 is a major protein of the accessory fibres of the mammalian sperm tail. In addition, ODF1 is found in the connecting piece, a complex structure located at the posterior end of the nucleus that connects the sperm head and tail. The tight coupling of the sperm head and tail is critical for the progressive motility of the sperm to reach the oocyte for fertilisation. The depletion of ODF1 by homologous recombination in mice led to male infertility. Although sperm tails were present in the epididymis, no intact spermatozoa were found. Instead, the depletion of ODF1 resulted in sperm decapitation, suggesting that ODF1 is essential for the formation of the coupling apparatus and the tight linkage of the sperm head and tail. However, the development of the linkage complex in the absence of ODF1 has never been investigated. Here, I analysed the fine structure of the developing connecting piece by transmission electron microscopy. I show that the connecting piece develops as in wild-type spermatids. Structural abnormalities were not observed when ODF1 was absent. Thus, ODF1 is dispensable for the development of the connecting piece. However, the decapitation of ODF1-deficient spermatozoa indicates that the heads and tails of the spermatozoa are not linked, so that they separate when force is applied.
Collapse
Affiliation(s)
- Sigrid Hoyer-Fender
- Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology-Developmental Biology, GZMB, Ernst-Caspari-Haus, Justus-von-Liebig-Weg 11, Georg-August-Universität Göttingen, 37077 Göttingen, Germany
| |
Collapse
|
27
|
Chen Y, Chen X, Zhang H, Sha Y, Meng R, Shao T, Yang X, Jin P, Zhuang Y, Min W, Xu D, Jiang Z, Li Y, Li L, Yue W, Yin C. TBC1D21 is an essential factor for sperm mitochondrial sheath assembly and male fertility‡. Biol Reprod 2022; 107:619-634. [PMID: 35403672 DOI: 10.1093/biolre/ioac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/03/2022] [Accepted: 03/29/2022] [Indexed: 11/12/2022] Open
Abstract
During spermiogenesis, the formation of the mitochondrial sheath is critical for male fertility. The molecular processes that govern the development of the mitochondrial sheath remain unknown. Whether TBC1D21 serves as a GTPase-activating protein (GAP) for GTP hydrolysis in the testis is unclear, despite recent findings indicating that it collaborates with numerous proteins to regulate the formation of the mitochondrial sheath. To thoroughly examine the property of TBC1D21 in spermiogenesis, we applied the CRISPR/Cas9 technology to generate the Tbc1d21-/- mice, Tbc1d21D125A R128K mice with mutation in the GAP catalytic residues (IxxDxxR), and Tbc1d21-3xFlag mice. Male Tbc1d21-/- mice were infertile due to the curved spermatozoa flagella. In vitro fertilization is ineffective for Tbc1d21-/- sperm, although healthy offspring were obtained by intracytoplasmic sperm injection. Electron microscopy revealed aberrant ultrastructural changes in the mitochondrial sheath. Thirty-four Rab vectors were constructed followed by co-immunoprecipitation, which identified RAB13 as a novel TBC1D21 binding protein. Interestingly, infertility was not observed in Tbc1d21D125A R128K mice harboring the catalytic residue, suggesting that TBC1D21 is not a typical GAP for Rab-GTP hydrolysis. Moreover, TBC1D21 was expressed in the sperm mitochondrial sheath in Tbc1d21-3xFlag mice. Immunoprecipitation-mass spectrometry demonstrated the interactions of TBC1D21 with ACTB, TPM3, SPATA19, and VDAC3 to regulate the architecture of the sperm midpiece. The collective findings suggest that TBC1D21 is a scaffold protein required for the organization and stabilization of the mitochondrial sheath morphology.
Collapse
Affiliation(s)
- Yongjie Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Xiu Chen
- Department of Pharmacy, Heze University, Heze, Shandong, China
| | - Haihang Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Yanwei Sha
- Department of Andrology, United Diagnostic and Research Center for Clinical Genetics, School of Public Health & Women and Children's Hospital, Xiamen University, Xiamen, China
| | - Ranran Meng
- National Institute of Biological Sciences, Beijing, China
| | - Tianyu Shao
- National Institute of Biological Sciences, Beijing, China
| | - Xiaoyan Yang
- National Institute of Biological Sciences, Beijing, China
| | - Pengpeng Jin
- National Institute of Biological Sciences, Beijing, China
| | - Yinghua Zhuang
- National Institute of Biological Sciences, Beijing, China
| | - Wanping Min
- National Institute of Biological Sciences, Beijing, China
| | - Dan Xu
- National Institute of Biological Sciences, Beijing, China
| | - Zhaodi Jiang
- National Institute of Biological Sciences, Beijing, China
| | - Yuhua Li
- National Institute of Biological Sciences, Beijing, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Wentao Yue
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chenghong Yin
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| |
Collapse
|
28
|
Palacios Martínez S, Greaney J, Zenker J. Beyond the centrosome: The mystery of microtubule organising centres across mammalian preimplantation embryos. Curr Opin Cell Biol 2022; 77:102114. [PMID: 35841745 DOI: 10.1016/j.ceb.2022.102114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/25/2022] [Accepted: 06/11/2022] [Indexed: 11/18/2022]
Abstract
Mammalian preimplantation embryogenesis depends on the spatio-temporal dynamics of the microtubule cytoskeleton to enable exceptionally fast changes in cell number, function, architecture, and fate. Microtubule organising centres (MTOCs), which coordinate the remodelling of microtubules, are therefore of fundamental significance during the first days of a new life. Despite its indispensable role during early mammalian embryogenesis, the origin of microtubule growth remains poorly understood. In this review, we summarise the most recent discoveries on microtubule organisation and function during early human embryogenesis and compare these to innovative studies conducted in alternative mammalian models. We emphasise the differences and analogies of centriole inheritance and their role during the first cleavage. Furthermore, we highlight the significance of non-centrosomal MTOCs for embryo viability and discuss the potential of novel in vitro models and light-inducible approaches towards unravelling microtubule formation in research and assisted reproductive technologies.
Collapse
Affiliation(s)
| | - Jessica Greaney
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Jennifer Zenker
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
29
|
Zhao Y, Wang H, Wiesehoefer C, Shah NB, Reetz E, Hwang JY, Huang X, Wang TE, Lishko PV, Davies KM, Wennemuth G, Nicastro D, Chung JJ. 3D structure and in situ arrangements of CatSper channel in the sperm flagellum. Nat Commun 2022; 13:3439. [PMID: 35715406 PMCID: PMC9205950 DOI: 10.1038/s41467-022-31050-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 05/31/2022] [Indexed: 12/14/2022] Open
Abstract
The sperm calcium channel CatSper plays a central role in successful fertilization as a primary Ca2+ gateway. Here, we applied cryo-electron tomography to visualize the higher-order organization of the native CatSper complex in intact mammalian sperm. The repeating CatSper units form long zigzag-rows along mouse and human sperm flagella. Above each tetrameric channel pore, most of the extracellular domains form a canopy that interconnects to a zigzag-shaped roof. Murine CatSper contains an additional wing-structure connected to the tetrameric channel. The intracellular domains link two neighboring channels to a diagonal array, suggesting a dimer formation. Fitting of an atomic model of isolated monomeric CatSper to the in situ map reveals supramolecular interactions and assembly of the CatSper complex. Loss of EFCAB9-CATSPERζ alters the architecture and interactions of the channels, resulting in fragmentation and misalignment of the zigzag-rows and disruption of flagellar movement in Efcab9-/- sperm. This work offers unique insights into the structural basis for understanding CatSper regulation of sperm motility.
Collapse
Affiliation(s)
- Yanhe Zhao
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Huafeng Wang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Caroline Wiesehoefer
- Department of Anatomy, University of Duisburg-Essen, Medical Faculty, 45147, Essen, Germany
| | - Naman B Shah
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Molecular Biophysics and Integrative Bioimaging division, Bioscience Area, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Evan Reetz
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jae Yeon Hwang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Xiaofang Huang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Tse-En Wang
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Polina V Lishko
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- The Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Karen M Davies
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Molecular Biophysics and Integrative Bioimaging division, Bioscience Area, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Gunther Wennemuth
- Department of Anatomy, University of Duisburg-Essen, Medical Faculty, 45147, Essen, Germany
| | - Daniela Nicastro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Jean-Ju Chung
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, 06510, USA.
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
30
|
Zhang R, Wu B, Liu C, Zhang Z, Wang X, Wang L, Xiao S, Chen Y, Wei H, Jiang H, Gao F, Yuan L, Li W. CCDC38 is required for sperm flagellum biogenesis and male fertility in mice. Development 2022; 149:275684. [DOI: 10.1242/dev.200516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/14/2022] [Indexed: 12/18/2022]
Abstract
ABSTRACT
The sperm flagellum is essential for male fertility, and defects in flagellum biogenesis are associated with male infertility. Deficiency of coiled-coil domain-containing (CCDC) 42 (CCDC42) is specifically associated with malformation of mouse sperm flagella. Here, we find that the testis-specific protein CCDC38 interacts with CCDC42, localizing on the manchette and sperm tail during spermiogenesis. Inactivation of CCDC38 in male mice results in a distorted manchette, multiple morphological abnormalities of the flagella of spermatozoa and eventually male sterility. Furthermore, we find that CCDC38 interacts with intraflagellar transport protein 88 (IFT88), as well as outer dense fibrous 2 (ODF2), and the knockout of Ccdc38 reduces transport of ODF2 to the flagellum. Altogether, our results uncover the essential role of CCDC38 in sperm flagellum biogenesis, and suggest that some mutations of these genes might be associated with male infertility in humans.
Collapse
Affiliation(s)
- Ruidan Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University 2 , Guangzhou 510623 , China
- University of the Chinese Academy of Sciences 3 , Beijing 100049 , China
| | - Bingbing Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University 2 , Guangzhou 510623 , China
- University of the Chinese Academy of Sciences 3 , Beijing 100049 , China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University 2 , Guangzhou 510623 , China
| | - Zhe Zhang
- Peking University Third Hospital 4 Department of Urology , , Beijing 100191 , China
- Peking University Third Hospital 5 Department of Andrology , , Beijing 100191 , China
- Peking University Third Hospital 6 Department of Reproductive Medicine Center , , Beijing 100191 , China
- Peking University Third Hospital 7 Department of Human Sperm Bank , , Beijing 100191 , China
| | - Xiuge Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- University of the Chinese Academy of Sciences 3 , Beijing 100049 , China
| | - Liying Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University 2 , Guangzhou 510623 , China
| | - Sai Xiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- University of the Chinese Academy of Sciences 3 , Beijing 100049 , China
| | - Yinghong Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- University of the Chinese Academy of Sciences 3 , Beijing 100049 , China
| | - Huafang Wei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University 2 , Guangzhou 510623 , China
| | - Hui Jiang
- Peking University Third Hospital 4 Department of Urology , , Beijing 100191 , China
- Peking University Third Hospital 5 Department of Andrology , , Beijing 100191 , China
- Peking University Third Hospital 6 Department of Reproductive Medicine Center , , Beijing 100191 , China
- Peking University Third Hospital 7 Department of Human Sperm Bank , , Beijing 100191 , China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- University of the Chinese Academy of Sciences 3 , Beijing 100049 , China
| | - Li Yuan
- Savaid Medical School, University of Chinese Academy of Sciences 8 , Beijing 100049 , China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Stem Cell and Regenerative Medicine Innovation Institute, Chinese Academy of Sciences 1 , Beijing 100101 , China
- Institute of Reproductive Health and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University 2 , Guangzhou 510623 , China
| |
Collapse
|
31
|
Li S, Wang Q, Huang L, Fan S, Li T, Shu Y, Zhang C, Zhou Y, Liu Q, Luo K, Tao M, Liu S. miR-199-5p regulates spermiogenesis at the posttranscriptional level via targeting Tekt1 in allotriploid crucian carp. J Anim Sci Biotechnol 2022; 13:44. [PMID: 35418106 PMCID: PMC9009052 DOI: 10.1186/s40104-022-00693-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sperm abnormalities are one of the primary factors leading to male sterility, but their pathogenesis is still unclear. Although miRNAs are suggested to exert important roles in the regulation of spermatogenesis at both transcriptional and posttranscriptional levels, little is currently known regarding the regulation of sperm flagella assembly by microRNAs (miRNAs). The role of miRNAs in the development of sperm abnormalities in sterile triploid fish has not been studied. RESULTS In this study, we found that miR-199-5p was widely expressed in all detected tissues of different-ploidy crucian carp. As one of the testis-specific candidate markers, Tekt1 was predominantly expressed in the testis. Quantitative real-time PCR (qRT-PCR) analyses showed that the expression trend of miR-199-5p was exactly opposite to that of Tekt1. Through bioinformatics analysis, we identified a putative miR-199-5p binding site in the Tekt1 mRNA. We further identified Tekt1 as a target of miR-199-5p using luciferase reporter assay. Finally, we confirmed that miR-199-5p was necessary for sperm flagellar assembly and spermatogenesis in vivo via intraperitoneal injection of miR-199-5p antagomir or agomir in diploid red crucian carp. Moreover, miR-199-5p gain-of-function could lead to spermatids apoptosis and abnormal spermatozoa structure, which is similar to that of allotriploid crucian carp. CONCLUSIONS Our studies suggested that abnormally elevated miR-199-5p inhibited the sperm flagella formation in spermiogenesis by negatively regulating the expression of Tekt1, thereby causing sperm abnormalities of male allotriploid crucian carp.
Collapse
Affiliation(s)
- Shengnan Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Qiubei Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Lu Huang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Siyu Fan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Ting Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Yuqing Shu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Chun Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, PR China
| | - Yi Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China.,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, PR China
| | - Qingfeng Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Kaikun Luo
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Min Tao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China. .,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, PR China.
| | - Shaojun Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China. .,Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, PR China.
| |
Collapse
|
32
|
Wang X, Yin L, Wen Y, Yuan S. Mitochondrial regulation during male germ cell development. Cell Mol Life Sci 2022; 79:91. [PMID: 35072818 PMCID: PMC11072027 DOI: 10.1007/s00018-022-04134-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/21/2021] [Accepted: 01/05/2022] [Indexed: 12/16/2022]
Abstract
Mitochondria tailor their morphology to execute their specialized functions in different cell types and/or different environments. During spermatogenesis, mitochondria undergo continuous morphological and distributional changes with germ cell development. Deficiencies in these processes lead to mitochondrial dysfunction and abnormal spermatogenesis, thereby causing male infertility. In recent years, mitochondria have attracted considerable attention because of their unique role in the regulation of piRNA biogenesis in male germ cells. In this review, we describe the varied characters of mitochondria and focus on key mitochondrial factors that play pivotal roles in the regulation of spermatogenesis, from primordial germ cells to spermatozoa, especially concerning metabolic shift, stemness and reprogramming, mitochondrial transformation and rearrangement, and mitochondrial defects in human sperm. Further, we discuss the molecular mechanisms underlying these processes.
Collapse
Affiliation(s)
- Xiaoli Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lisha Yin
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yujiao Wen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Laboratory Animal Center, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
33
|
Teves ME, Roldan ERS. Sperm bauplan and function and underlying processes of sperm formation and selection. Physiol Rev 2022; 102:7-60. [PMID: 33880962 PMCID: PMC8812575 DOI: 10.1152/physrev.00009.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
The spermatozoon is a highly differentiated and polarized cell, with two main structures: the head, containing a haploid nucleus and the acrosomal exocytotic granule, and the flagellum, which generates energy and propels the cell; both structures are connected by the neck. The sperm's main aim is to participate in fertilization, thus activating development. Despite this common bauplan and function, there is an enormous diversity in structure and performance of sperm cells. For example, mammalian spermatozoa may exhibit several head patterns and overall sperm lengths ranging from ∼30 to 350 µm. Mechanisms of transport in the female tract, preparation for fertilization, and recognition of and interaction with the oocyte also show considerable variation. There has been much interest in understanding the origin of this diversity, both in evolutionary terms and in relation to mechanisms underlying sperm differentiation in the testis. Here, relationships between sperm bauplan and function are examined at two levels: first, by analyzing the selective forces that drive changes in sperm structure and physiology to understand the adaptive values of this variation and impact on male reproductive success and second, by examining cellular and molecular mechanisms of sperm formation in the testis that may explain how differentiation can give rise to such a wide array of sperm forms and functions.
Collapse
Affiliation(s)
- Maria Eugenia Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia
| | - Eduardo R S Roldan
- Department of Biodiversity and Evolutionary Biology, Museo Nacional de Ciencias Naturales (CSIC), Madrid, Spain
| |
Collapse
|
34
|
Ogata H, Tsukamoto M, Yamashita K, Iwamori T, Takahashi H, Kaneko T, Iwamori N, Inai T, Iida H. Effects of Calyculin a on the Motility and Protein Phosphorylation in Frozen-Thawed Bull Spermatozoa. Zoolog Sci 2021; 38:531-543. [PMID: 34854285 DOI: 10.2108/zs210046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/06/2021] [Indexed: 11/17/2022]
Abstract
In this study, we examined the effects of calyculin A, a phosphatase inhibitor, on motility, protein phosphorylation, and the distribution of phospho-(Ser/Thr) PKA substrates in frozen-thawed bull spermatozoa that are actually used by most farmers for breeding. The data showed that calyculin A, which has been reported to have a positive effect on the motility of ejaculated fresh spermatozoa, distinctly decreased the motility of frozen-thawed bull spermatozoa even if a cell activator, such as caffeine, was present in the incubation medium and that the suppressive effect of calyculin A was dose-dependent and continued for at least 200 min. Immunoblot analyses revealed that de novo protein phosphorylation was not detected in spermatozoa exposed to caffeine or dbcAMP (a cell-permeable cAMP analog), while the addition of calyculin A to the medium brought about the appearance of several phosphorylated proteins at 50 kDa and 75 kDa, suggesting that 50 kDa and 75 kDa proteins, which were phosphorylated by activation of cAMP-dependent PKA, were not dephosphorylated and were accumulated in spermatozoa due to the suppression of calyculin A-sensitive protein phosphatases. Immunofluorescence microscopy revealed that calyculin A caused, alone or in conjunction with caffeine or dbcAMP, the accumulation of phospho-PKA substrates at the annulus, although caffeine or dbcAMP alone did not. This study suggested that calyculin A decreases the motility of frozen-thawed bull spermatozoa concomitant with the accumulation of phospho-(Ser/Thr) PKA substrates at the annulus of flagella.
Collapse
Affiliation(s)
- Honami Ogata
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Mariko Tsukamoto
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Kenichi Yamashita
- Sensing System Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Tosu, Saga 841-0052, Japan
| | - Tokuko Iwamori
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Hideyuki Takahashi
- Kuju Agriculture Research Center, Kyushu University, Oita 878-0201, Japan
| | - Takane Kaneko
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Naoki Iwamori
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
| | - Tetsuichiro Inai
- Department of Morphological Biology, Fukuoka Dental College, Sawara-ku, Fukuoka 814-0193, Japan
| | - Hiroshi Iida
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka 819-0395, Japan,
| |
Collapse
|
35
|
Giaccagli MM, Gómez-Elías MD, Herzfeld JD, Marín-Briggiler CI, Cuasnicú PS, Cohen DJ, Da Ros VG. Capacitation-Induced Mitochondrial Activity Is Required for Sperm Fertilizing Ability in Mice by Modulating Hyperactivation. Front Cell Dev Biol 2021; 9:767161. [PMID: 34765607 PMCID: PMC8576324 DOI: 10.3389/fcell.2021.767161] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 01/22/2023] Open
Abstract
To become fully competent to fertilize an egg, mammalian sperm undergo a series of functional changes within the female tract, known as capacitation, that require an adequate supply and management of energy. However, the contribution of each ATP generating pathway to sustain the capacitation-associated changes remains unclear. Based on this, we investigated the role of mitochondrial activity in the acquisition of sperm fertilizing ability during capacitation in mice. For this purpose, the dynamics of the mitochondrial membrane potential (MMP) was studied by flow cytometry with the probe tetramethylrhodamine ethyl ester (TMRE). We observed a time-dependent increase in MMP only in capacitated sperm as well as a specific staining with the probe in the flagellar region where mitochondria are confined. The MMP rise was prevented when sperm were exposed to the mitochondrial uncoupler carbonyl cyanide m-chlorophenyl hydrazine (CCCP) or the protein kinase A (PKA) inhibitor H89 during capacitation, indicating that MMP increase is dependent on capacitation and H89-sensitive events. Results showed that whereas nearly all motile sperm were TMRE positive, immotile cells were mostly TMRE negative, supporting an association between high MMP and sperm motility. Furthermore, CCCP treatment during capacitation did not affect PKA substrate and tyrosine phosphorylations but produced a decrease in hyperactivation measured by computer assisted sperm analysis (CASA), similar to that observed after H89 exposure. In addition, CCCP inhibited the in vitro sperm fertilizing ability without affecting cumulus penetration and gamete fusion, indicating that the hyperactivation supported by mitochondrial function is needed mainly for zona pellucida penetration. Finally, complementary in vivo fertilization experiments further demonstrated the fundamental role of mitochondrial activity for sperm function. Altogether, our results show the physiological relevance of mitochondrial functionality for sperm fertilization competence.
Collapse
Affiliation(s)
- María Milagros Giaccagli
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Matías Daniel Gómez-Elías
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Jael Dafne Herzfeld
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Clara Isabel Marín-Briggiler
- Laboratorio de Biología Celular y Molecular de la Reproducción, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Patricia Sara Cuasnicú
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Débora Juana Cohen
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| | - Vanina Gabriela Da Ros
- Laboratorio de Mecanismos Moleculares de la Fertilización, Instituto de Biología y Medicina Experimental (IByME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
36
|
In-cell structures of conserved supramolecular protein arrays at the mitochondria-cytoskeleton interface in mammalian sperm. Proc Natl Acad Sci U S A 2021; 118:2110996118. [PMID: 34737233 PMCID: PMC8609336 DOI: 10.1073/pnas.2110996118] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2021] [Indexed: 11/24/2022] Open
Abstract
Spatial organization of mitochondria is vital for cellular function. In many specialized cell types, mitochondria are immobilized at specific subcellular loci through interactions with the cytoskeleton. One of the most striking mitochondrial configurations occurs in mammalian sperm, where mitochondria wrap around the flagellum. Malformation of the mitochondrial sheath causes infertility, but the molecular structures underlying this intricate arrangement are unknown. Here, we analyzed the mitochondrial sheath in sperm from three mammalian species. We find that although mitochondrial dimensions and cristae architecture vary across species, molecular assemblies mediating intermitochondria and mitochondria–cytoskeleton interactions are conserved. These findings yield important insight into sperm physiology and evolution and are relevant for other polarized cell types, such as muscles, neurons, photoreceptors, and hair cells. Mitochondria–cytoskeleton interactions modulate cellular physiology by regulating mitochondrial transport, positioning, and immobilization. However, there is very little structural information defining mitochondria–cytoskeleton interfaces in any cell type. Here, we use cryofocused ion beam milling-enabled cryoelectron tomography to image mammalian sperm, where mitochondria wrap around the flagellar cytoskeleton. We find that mitochondria are tethered to their neighbors through intermitochondrial linkers and are anchored to the cytoskeleton through ordered arrays on the outer mitochondrial membrane. We use subtomogram averaging to resolve in-cell structures of these arrays from three mammalian species, revealing they are conserved across species despite variations in mitochondrial dimensions and cristae organization. We find that the arrays consist of boat-shaped particles anchored on a network of membrane pores whose arrangement and dimensions are consistent with voltage-dependent anion channels. Proteomics and in-cell cross-linking mass spectrometry suggest that the conserved arrays are composed of glycerol kinase-like proteins. Ordered supramolecular assemblies may serve to stabilize similar contact sites in other cell types in which mitochondria need to be immobilized in specific subcellular environments, such as in muscles and neurons.
Collapse
|
37
|
Lester WC, Johnson T, Hale B, Serra N, Elgart B, Wang R, Geyer CB, Sperry AO. Aurora a kinase (AURKA) is required for male germline maintenance and regulates sperm motility in the mouse. Biol Reprod 2021; 105:1603-1616. [PMID: 34518881 DOI: 10.1093/biolre/ioab168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/12/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Aurora A kinase (AURKA) is an important regulator of cell division and is required for assembly of the mitotic spindle. We recently reported the unusual finding that this mitotic kinase is also found on the sperm flagellum. To determine its requirement in spermatogenesis, we generated conditional knockout animals with deletion of the Aurka gene in either spermatogonia or spermatocytes to assess its role in mitotic and postmitotic cells, respectively. Deletion of Aurka in spermatogonia resulted in disappearance of all developing germ cells in the testis, as expected given its vital role in mitotic cell division. Deletion of Aurka in spermatocytes reduced testis size, sperm count, and fertility, indicating disruption of meiosis or an effect on spermiogenesis in developing mice. Interestingly, deletion of Aurka in spermatocytes increased apoptosis in spermatocytes along with an increase in the percentage of sperm with abnormal morphology. Despite the increase in abnormal sperm, sperm from spermatocyte Aurka knockout mice displayed increased progressive motility. In addition, sperm lysate prepared from Aurka knockout animals had decreased protein phosphatase 1 (PP1) activity. Together, our results show that AURKA plays multiple roles in spermatogenesis, from mitotic divisions of spermatogonia to sperm morphology and motility.
Collapse
Affiliation(s)
- William C Lester
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| | - Taylor Johnson
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Ben Hale
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Nicholas Serra
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Brian Elgart
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| | - Rong Wang
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology at the Brody School of Medicine.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville NC, USA 27834
| | - Ann O Sperry
- Department of Anatomy and Cell Biology at the Brody School of Medicine
| |
Collapse
|
38
|
Tapia Contreras C, Hoyer-Fender S. The Transformation of the Centrosome into the Basal Body: Similarities and Dissimilarities between Somatic and Male Germ Cells and Their Relevance for Male Fertility. Cells 2021; 10:2266. [PMID: 34571916 PMCID: PMC8471410 DOI: 10.3390/cells10092266] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
The sperm flagellum is essential for the transport of the genetic material toward the oocyte and thus the transmission of the genetic information to the next generation. During the haploid phase of spermatogenesis, i.e., spermiogenesis, a morphological and molecular restructuring of the male germ cell, the round spermatid, takes place that includes the silencing and compaction of the nucleus, the formation of the acrosomal vesicle from the Golgi apparatus, the formation of the sperm tail, and, finally, the shedding of excessive cytoplasm. Sperm tail formation starts in the round spermatid stage when the pair of centrioles moves toward the posterior pole of the nucleus. The sperm tail, eventually, becomes located opposed to the acrosomal vesicle, which develops at the anterior pole of the nucleus. The centriole pair tightly attaches to the nucleus, forming a nuclear membrane indentation. An articular structure is formed around the centriole pair known as the connecting piece, situated in the neck region and linking the sperm head to the tail, also named the head-to-tail coupling apparatus or, in short, HTCA. Finally, the sperm tail grows out from the distal centriole that is now transformed into the basal body of the flagellum. However, a centriole pair is found in nearly all cells of the body. In somatic cells, it accumulates a large mass of proteins, the pericentriolar material (PCM), that together constitute the centrosome, which is the main microtubule-organizing center of the cell, essential not only for the structuring of the cytoskeleton and the overall cellular organization but also for mitotic spindle formation and chromosome segregation. However, in post-mitotic (G1 or G0) cells, the centrosome is transformed into the basal body. In this case, one of the centrioles, which is always the oldest or mother centriole, grows the axoneme of a cilium. Most cells of the body carry a single cilium known as the primary cilium that serves as an antenna sensing the cell's environment. Besides, specialized cells develop multiple motile cilia differing in substructure from the immotile primary cilia that are essential in moving fluids or cargos over the cellular surface. Impairment of cilia formation causes numerous severe syndromes that are collectively subsumed as ciliopathies. This comparative overview serves to illustrate the molecular mechanisms of basal body formation, their similarities, and dissimilarities, in somatic versus male germ cells, by discussing the involved proteins/genes and their expression, localization, and function. The review, thus, aimed to provide a deeper knowledge of the molecular players that is essential for the expansion of clinical diagnostics and treatment of male fertility disorders.
Collapse
Affiliation(s)
| | - Sigrid Hoyer-Fender
- Göttingen Center of Molecular Biosciences, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology-Developmental Biology, Faculty of Biology and Psychology, Georg-August University of Göttingen, 37077 Göttingen, Germany;
| |
Collapse
|
39
|
Zhang G, Li D, Tu C, Meng L, Tan Y, Ji Z, Cheng J, Lu G, Lin G, Zhang H, Sun J, Wang M, Du J, Xu W. Loss-of-function missense variant of AKAP4 induced male infertility through reduced interaction with QRICH2 during sperm flagella development. Hum Mol Genet 2021; 31:219-231. [PMID: 34415320 DOI: 10.1093/hmg/ddab234] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/25/2022] Open
Abstract
Sperm fibrous sheath is closely related to sperm maturation, capacitation and motility, and A-kinase anchor protein 4 (AKAP4) is the most abundant protein in sperm fibrous sheath. Previous studies found incomplete sperm fibrous sheaths and abnormal flagella in Akap4 knockout (KO) mice. Meanwhile, it was reported that the partial deletion in AKAP4 is highly relevant to the dysplasia of the fibrous sheath in an infertile man, and so far, there is no report about male infertility caused by hemizygous AKAP4 variant. Furthermore, the specific mechanisms of how the variant is relevant to the phenotype remain elusive. In this study, we investigated three multiple morphological abnormalities of the sperm flagella (MMAF)-affected men from three independent families (including one consanguine family) carried hemizygous c.C1285T variant in AKAP4. The patients carried thisvariant showed dysplastic sperm fibrous sheath and the protein expression of AKAP4 was decreased in flagella which was further confirmed in HEK-293 T cells in vitro. In addition, the co-localization and interaction between AKAP4 and glutamine-rich protein 2 (QRICH2) on the molecular level were identified by immunofluorescence and Co-immunoprecipitation (CO-IP). The hemizygous c.1285C > T variant in AKAP4 induced decreased protein expression of QRICH2 in spermatozoa. These results suggested that the normal expression of AKAP4 is required for maintaining the expression of QRICH2 and the decreased protein expression of AKAP4 and QRICH2,as well as the interaction between them induced by the hemizygous variant of AKAP4 caused dysplastic fibrous sheath, which eventually led to reduced sperm motility and male infertility.
Collapse
Affiliation(s)
- Guohui Zhang
- Department of Obstetrics and Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Dongyan Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, China
| | - Chaofeng Tu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Lanlan Meng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Yueqiu Tan
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Zhiliang Ji
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jiao Cheng
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Guangxiu Lu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Huan Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Jinpeng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Mingwei Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, 250012, China
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410078, China.,Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Wenming Xu
- Department of Obstetrics and Gynecology, Joint Laboratory of Reproductive Medicine (SCU-CUHK), Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
40
|
Wu B, Gao H, Liu C, Li W. The coupling apparatus of the sperm head and tail†. Biol Reprod 2021; 102:988-998. [PMID: 31995163 DOI: 10.1093/biolre/ioaa016] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/05/2019] [Accepted: 01/26/2020] [Indexed: 12/23/2022] Open
Abstract
A strong sperm head-tail coupling apparatus (HTCA) is needed to ensure the integrity of spermatozoa during their fierce competition to fertilize the egg. A lot of HTCA-specific components have evolved to strengthen the attachment of the tail to the implantation fossa at the sperm head. Defects in HTCA formation lead to acephalic spermatozoa syndrome and pathologies of some male infertility. Recent studies have provided insights into the pathogenic molecular mechanisms of acephalic spermatozoa syndrome. Here, we summarize the proteins involved in sperm neck development and focus on their roles in the formation of HTCA. In addition, we discuss the fine structures of the sperm neck in different species from an evolutionary view, highlighting the potential conservative mechanism of HTCA formation.
Collapse
Affiliation(s)
- Bingbing Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chao Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
41
|
Kumar P, Wang M, Isachenko E, Rahimi G, Mallmann P, Wang W, von Brandenstein M, Isachenko V. Unraveling Subcellular and Ultrastructural Changes During Vitrification of Human Spermatozoa: Effect of a Mitochondria-Targeted Antioxidant and a Permeable Cryoprotectant. Front Cell Dev Biol 2021; 9:672862. [PMID: 34277615 PMCID: PMC8284099 DOI: 10.3389/fcell.2021.672862] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Mitochondria-targeted antioxidants have great potential to counterbalance the generated reactive oxygen species (ROS) because they cross the inner membrane of the mitochondria. Still, their use was not reported in vitrified human spermatozoa. Our laboratory has successfully vitrified spermatozoa without the use of permeable cryoprotectants, but subcellular-level evidence was missing. Therefore, this study aimed to improve spermatozoa vitrification using a mitochondria-targeted antioxidant (mitoquinone, MitoQ), reveal ultrastructural changes in the spermatozoa due to the use of a permeable cryoprotectant, and report alterations of functional proteins during the spermatozoa vitrification process. For this, each of 20 swim-up-prepared ejaculates was divided into seven aliquots and diluted with a vitrification medium supplemented with varying concentrations of MitoQ (0.02 and 0.2 μM), glycerol (1, 4, and 6%), and a combination of MitoQ and glycerol. All aliquots were vitrified by the aseptic capillary method developed in our laboratory. The spermatozoa function assays revealed that the addition of either MitoQ (0.02 μM), glycerol (1%), or a combination of MitoQ (0.02 μM) and glycerol (1%) in the vitrification medium results in better or equivalent spermatozoa quality relative to the control. Transmission electron microscopy revealed that MitoQ protects the spermatozoa from undergoing ultrastructural alterations, but glycerol induced ultrastructural alterations during the vitrification process. Next, we performed label-free quantitative proteomics and identified 1,759 proteins, of which 69, 60, 90, and 81 were altered in the basal medium, 0.02 μM MitoQ, 1% glycerol, and Mito-glycerol groups, respectively. Actin, tubulins, and outer dense fiber proteins were not affected during the vitrification process. Some of the identified ubiquitinating enzymes were affected during spermatozoa vitrification. Only a few proteins responsible for phosphorylation were altered during vitrification. Similarly, several proteins involved in spermatozoa–egg fusion and fertilization (IZUMO1 and Tektin) were not affected during the vitrification process. In conclusion, MitoQ attenuates the vitrification-induced ultrastructural changes and alterations in the key proteins involved in spermatozoa functions and fertilization.
Collapse
Affiliation(s)
- Pradeep Kumar
- Department of Animal Physiology and Reproduction, ICAR-Central Institute for Research on Buffaloes, Hisar, India.,Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, Cologne, Germany
| | - Mengying Wang
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, Cologne, Germany
| | - Evgenia Isachenko
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, Cologne, Germany
| | - Gohar Rahimi
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, Cologne, Germany
| | - Peter Mallmann
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, Cologne, Germany
| | - Wanxue Wang
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, Cologne, Germany
| | | | - Vladimir Isachenko
- Department of Obstetrics and Gynaecology, Medical Faculty, Cologne University, Cologne, Germany
| |
Collapse
|
42
|
Zhang Y, Yang L, Huang L, Liu G, Nie X, Zhang X, Xing X. SUN5 Interacting With Nesprin3 Plays an Essential Role in Sperm Head-to-Tail Linkage: Research on Sun5 Gene Knockout Mice. Front Cell Dev Biol 2021; 9:684826. [PMID: 34268309 PMCID: PMC8276135 DOI: 10.3389/fcell.2021.684826] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Acephalic spermatozoa syndrome is a rare genetic and reproductive disease. Recent studies have shown that approximately 33–47% of patients with acephalic spermatozoa syndrome have SUN5 mutations, but the molecular mechanism underlying this phenomenon has not been elucidated. In this study, we generated Sun5 knockout mice and found that the head-to-tail linkage was broken in Sun5–/– mice, which was similar to human acephalic spermatozoa syndrome. Furthermore, ultrastructural imaging revealed that the head-tail coupling apparatus (HTCA) and the centrosome were distant from the nucleus at steps 9–10 during spermatid elongation. With the manchette disappearing at steps 13–14, the head and the tail segregated. To explore the molecular mechanism underlying this process, bioinformatic analysis was performed and showed that Sun5 may interact with Nesprin3. Further coimmunoprecipitation (Co-IP) and immunofluorescence assays confirmed that Sun5 and Nesprin3 were indeed bona fide interaction partners that formed the linker of the nucleoskeleton and cytoskeleton (LINC) complex participating in the connection of the head and tail of spermatozoa. Nesprin3 was located posterior and anterior to the nucleus during spermiogenesis in wild-type mice, whereas it lost its localization at the implantation fossa of the posterior region in Sun5–/– mice. Without correct localization of Nesprin3 at the nuclear membrane, the centrosome, which is the originator of the flagellum, was distant from the nucleus, which led to the separation of the head and tail. In addition, isobaric tag for relative and absolute quantitation results showed that 47 proteins were upregulated, and 56 proteins were downregulated, in the testis in Sun5–/– mice, and the downregulation of spermatogenesis-related proteins (Odf1 and Odf2) may also contribute to the damage to the spermatozoa head-to-tail linkage. Our findings suggested that Sun5 is essential for the localization of Nesprin3 at the posterior nuclear membrane, which plays an essential role in the sperm head-tail connection.
Collapse
Affiliation(s)
- Yunfei Zhang
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Linfei Yang
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Huang
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Gang Liu
- The Institute of Reproduction and Stem Cell Engineering, Central South University, Changsha, China
| | - Xinmin Nie
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xinxing Zhang
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaowei Xing
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
43
|
ARMC12 regulates spatiotemporal mitochondrial dynamics during spermiogenesis and is required for male fertility. Proc Natl Acad Sci U S A 2021; 118:2018355118. [PMID: 33536340 PMCID: PMC8017931 DOI: 10.1073/pnas.2018355118] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Although formation of the mitochondrial sheath is a critical process in the formation of mature spermatozoa, the molecular mechanisms involved in mitochondrial sheath genesis remain unclear. Using gene-manipulated mice, we discovered that ARMC12 regulates spatiotemporal “sperm mitochondrial dynamics” during mitochondrial sheath formation through interactions with mitochondrial proteins MIC60, VDAC2, and VDAC3 as well as testis-specific proteins TBC1D21 and GK2. In addition, we demonstrated that ARMC12-interacting proteins TBC1D21 and GK2 are also essential for mitochondrial sheath formation. Our paper sheds light on the molecular mechanisms of mitochondrial sheath formation and the regulation of sperm mitochondrial dynamics, allowing us to further understand the biology of spermatogenesis and the etiology of infertility in men. The mammalian sperm midpiece has a unique double-helical structure called the mitochondrial sheath that wraps tightly around the axoneme. Despite the remarkable organization of the mitochondrial sheath, the molecular mechanisms involved in mitochondrial sheath formation are unclear. In the process of screening testis-enriched genes for functions in mice, we identified armadillo repeat-containing 12 (ARMC12) as an essential protein for mitochondrial sheath formation. Here, we engineered Armc12-null mice, FLAG-tagged Armc12 knock-in mice, and TBC1 domain family member 21 (Tbc1d21)-null mice to define the functions of ARMC12 in mitochondrial sheath formation in vivo. We discovered that absence of ARMC12 causes abnormal mitochondrial coiling along the flagellum, resulting in reduced sperm motility and male sterility. During spermiogenesis, sperm mitochondria in Armc12-null mice cannot elongate properly at the mitochondrial interlocking step which disrupts abnormal mitochondrial coiling. ARMC12 is a mitochondrial peripheral membrane protein and functions as an adherence factor between mitochondria in cultured cells. ARMC12 in testicular germ cells interacts with mitochondrial proteins MIC60, VDAC2, and VDAC3 as well as TBC1D21 and GK2, which are required for mitochondrial sheath formation. We also observed that TBC1D21 is essential for the interaction between ARMC12 and VDAC proteins in vivo. These results indicate that ARMC12 uses integral mitochondrial membrane proteins VDAC2 and VDAC3 as scaffolds to link mitochondria and works cooperatively with TBC1D21. Thus, our studies have revealed that ARMC12 regulates spatiotemporal mitochondrial dynamics to form the mitochondrial sheath through cooperative interactions with several proteins on the sperm mitochondrial surface.
Collapse
|
44
|
Khanal S, Leung MR, Royfman A, Fishman EL, Saltzman B, Bloomfield-Gadêlha H, Zeev-Ben-Mordehai T, Avidor-Reiss T. A dynamic basal complex modulates mammalian sperm movement. Nat Commun 2021; 12:3808. [PMID: 34155206 PMCID: PMC8217517 DOI: 10.1038/s41467-021-24011-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/28/2021] [Indexed: 01/04/2023] Open
Abstract
Reproductive success depends on efficient sperm movement driven by axonemal dynein-mediated microtubule sliding. Models predict sliding at the base of the tail - the centriole - but such sliding has never been observed. Centrioles are ancient organelles with a conserved architecture; their rigidity is thought to restrict microtubule sliding. Here, we show that, in mammalian sperm, the atypical distal centriole (DC) and its surrounding atypical pericentriolar matrix form a dynamic basal complex (DBC) that facilitates a cascade of internal sliding deformations, coupling tail beating with asymmetric head kinking. During asymmetric tail beating, the DC's right side and its surroundings slide ~300 nm rostrally relative to the left side. The deformation throughout the DBC is transmitted to the head-tail junction; thus, the head tilts to the left, generating a kinking motion. These findings suggest that the DBC evolved as a dynamic linker coupling sperm head and tail into a single self-coordinated system.
Collapse
Affiliation(s)
- Sushil Khanal
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Miguel Ricardo Leung
- The Division of Structural Biology, Wellcome Centre for Human Genetics, The University of Oxford, Oxford, UK
- Cryo-Electron Microscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Abigail Royfman
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Emily L Fishman
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Barbara Saltzman
- School of Population Health, College of Health and Human Services, University of Toledo, Toledo, OH, USA
| | - Hermes Bloomfield-Gadêlha
- Department of Engineering Mathematics and Bristol Robotics Laboratory, University of Bristol, Bristol, UK
| | - Tzviya Zeev-Ben-Mordehai
- The Division of Structural Biology, Wellcome Centre for Human Genetics, The University of Oxford, Oxford, UK.
- Cryo-Electron Microscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.
| | - Tomer Avidor-Reiss
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA.
- Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
45
|
Fang X, Gamallat Y, Chen Z, Mai H, Zhou P, Sun C, Li X, Li H, Zheng S, Liao C, Yang M, Li Y, Yang Z, Ma C, Han D, Zuo L, Xu W, Hu H, Sun L, Li N. Hypomorphic and hypermorphic mouse models of Fsip2 indicate its dosage-dependent roles in sperm tail and acrosome formation. Development 2021; 148:269073. [PMID: 34125190 DOI: 10.1242/dev.199216] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/04/2021] [Indexed: 02/03/2023]
Abstract
Loss-of-function mutations in multiple morphological abnormalities of the sperm flagella (MMAF)-associated genes lead to decreased sperm motility and impaired male fertility. As an MMAF gene, the function of fibrous sheath-interacting protein 2 (FSIP2) remains largely unknown. In this work, we identified a homozygous truncating mutation of FSIP2 in an infertile patient. Accordingly, we constructed a knock-in (KI) mouse model with this mutation. In parallel, we established an Fsip2 overexpression (OE) mouse model. Remarkably, KI mice presented with the typical MMAF phenotype, whereas OE mice showed no gross anomaly except for sperm tails with increased length. Single-cell RNA sequencing of the testes uncovered altered expression of genes related to sperm flagellum, acrosomal vesicle and spermatid development. We confirmed the expression of Fsip2 at the acrosome and the physical interaction of this gene with Acrv1, an acrosomal marker. Proteomic analysis of the testes revealed changes in proteins sited at the fibrous sheath, mitochondrial sheath and acrosomal vesicle. We also pinpointed the crucial motifs of Fsip2 that are evolutionarily conserved in species with internal fertilization. Thus, this work reveals the dosage-dependent roles of Fsip2 in sperm tail and acrosome formation.
Collapse
Affiliation(s)
- Xiang Fang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Yaser Gamallat
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Zhiheng Chen
- Center of Reproductive Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Hanran Mai
- Department of Andrology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Pei Zhou
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Chuanbo Sun
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Xiaoliang Li
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 610041 Chengdu, China
| | - Hong Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Shuxin Zheng
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Caihua Liao
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Miaomiao Yang
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Yan Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Zeyu Yang
- Guangdong Technion-Israel Institute of Technology, Shantou, 515063 Guangdong, China
| | - Caiqi Ma
- Center of Reproductive Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Dingding Han
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Liandong Zuo
- Department of Andrology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Wenming Xu
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 610041 Chengdu, China
| | - Hao Hu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China.,Third Affiliated Hospital of Zhengzhou University, 450052 Zhengzhou, China.,Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Ling Sun
- Center of Reproductive Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| | - Na Li
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623 Guangzhou, China
| |
Collapse
|
46
|
Tung CK, Suarez SS. Co-Adaptation of Physical Attributes of the Mammalian Female Reproductive Tract and Sperm to Facilitate Fertilization. Cells 2021; 10:cells10061297. [PMID: 34073739 PMCID: PMC8225031 DOI: 10.3390/cells10061297] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 12/20/2022] Open
Abstract
The functions of the female reproductive tract not only encompass sperm migration, storage, and fertilization, but also support the transport and development of the fertilized egg through to the birth of offspring. Further, because the tract is open to the external environment, it must also provide protection against invasive pathogens. In biophysics, sperm are considered “pusher microswimmers”, because they are propelled by pushing fluid behind them. This type of swimming by motile microorganisms promotes the tendency to swim along walls and upstream in gentle fluid flows. Thus, the architecture of the walls of the female tract, and the gentle flows created by cilia, can guide sperm migration. The viscoelasticity of the fluids in the tract, such as mucus secretions, also promotes the cooperative swimming of sperm that can improve fertilization success; at the same time, the mucus can also impede the invasion of pathogens. This review is focused on how the mammalian female reproductive tract and sperm interact physically to facilitate the movement of sperm to the site of fertilization. Knowledge of female/sperm interactions can not only explain how the female tract can physically guide sperm to the fertilization site, but can also be applied for the improvement of in vitro fertilization devices.
Collapse
Affiliation(s)
- Chih-Kuan Tung
- Department of Physics, North Carolina A&T State University, Greensboro, NC 27411, USA
- Correspondence:
| | - Susan S. Suarez
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA;
| |
Collapse
|
47
|
Kobayashi M, Tsuzuki C, Kobayashi M, Tsuchiya H, Yamashita Y, Ueno K, Onozawa M, Kobayashi M, Kawakami E, Hori T. Effect of supplementation with the reduced form of coenzyme Q10 on semen quality and antioxidant status in dogs with poor semen quality: Three case studies. J Vet Med Sci 2021; 83:1044-1049. [PMID: 34011783 PMCID: PMC8349810 DOI: 10.1292/jvms.21-0174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Oxidative stress owing to an imbalance between reactive oxygen species and antioxidants, such as coenzyme Q10 (CoQ10), is a major contributor to male
infertility. We investigated the effects of the reduced form of CoQ10 (ubiquinol) supplementation on semen quality in dogs with poor semen quality. Three dogs
received 100 mg of ubiquinol orally once daily for 12 weeks. Semen quality, serum testosterone, and seminal plasma superoxide dismutase (SOD) activity were
examined at 2-week intervals from 2 weeks before ubiquinol supplementation to 4 weeks after the treatment. Ubiquinol improved sperm motility, reduced
morphologically abnormal sperm, and increased seminal plasma SOD activity; however, it had no effect on testosterone level, semen volume, and sperm number.
Ubiquinol supplementation could be used as a non-endocrine therapy for infertile dogs.
Collapse
Affiliation(s)
- Masanori Kobayashi
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Chie Tsuzuki
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Marika Kobayashi
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Hinano Tsuchiya
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Yume Yamashita
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Kanako Ueno
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Moe Onozawa
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Masato Kobayashi
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | - Eiichi Kawakami
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan.,Japan Institute of Small Animal Reproduction (Bio Art), 3-16-9 Uchikanda, Chiyoda-ku, Tokyo 101-0047, Japan
| | - Tatsuya Hori
- Laboratory of Reproduction, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| |
Collapse
|
48
|
Characterization of Bottlenose Dolphin (Tursiops truncatus) Sperm Based on Morphometric Traits. BIOLOGY 2021; 10:biology10050355. [PMID: 33922110 PMCID: PMC8143526 DOI: 10.3390/biology10050355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 11/19/2022]
Abstract
Simple Summary Dolphins are one of the best adapted aquatic mammalians in captivity. While these animals can reproduce naturally in aquaria, many aspects related to their reproduction are still unknown. Their behavior, sociability with humans and ability to learn make dolphins ideal subjects for investigating their reproduction features for conservation programs aimed at this aquatic species and others. It is known that dolphins use a multi-male mating strategy, in which sperm competition could play a fundamental role. This study aims to characterize the shape and dimensions of dolphin sperm from two mature males and putatively classifies them into subpopulations. Moreover, the influence of sex hormone levels (testosterone) and refrigeration (temperature and storage period) with sperm dimension was evaluated. The results indicated that sperm dimensions and shape differed between the two males studied and that the sperm of both males could be classified into two subpopulations depending on their dimensions. Moreover, both testosterone levels and refrigeration were seen to influence sperm dimensions. This investigation provides new insights into sperm competition in dolphin species, and the results could be extrapolated to other endangered aquatic mammalian species. Abstract Bottlenose dolphin (Tursiops truncatus) males follow many reproductive strategies to ensure their paternity. However, little is known about the sperm traits, including morphometric features, that contribute to their reproductive success. Our aim was to study dolphin sperm morphometry (a total of 13 parameters) in two adult males to evaluate (i) presumptive sperm subpopulations, (ii) the correlation of sperm morphometry with testosterone levels and (iii) the effect of refrigerated storage on the sperm morphometry. Sperm populations were classified into four principal components (PCs) based on morphometry (>94% of cumulative variance). The PCs clustered into two different sperm subpopulations, which differed between males. Furthermore, the levels of serum testosterone were positively correlated with the length of the midpiece but negatively correlated with head width and the principal piece, flagellum and total sperm lengths. Most of the sperm morphometric parameters changed during the storage period (day 1 vs. day 7), but only the principal piece length was affected by the storage temperature (5 °C vs. 15 °C). This is the first study to identify dolphin sperm subpopulations based on morphometry and the influence of serum testosterone and refrigeration on sperm morphometry.
Collapse
|
49
|
Leung MR, Roelofs MC, Ravi RT, Maitan P, Henning H, Zhang M, Bromfield EG, Howes SC, Gadella BM, Bloomfield‐Gadêlha H, Zeev‐Ben‐Mordehai T. The multi-scale architecture of mammalian sperm flagella and implications for ciliary motility. EMBO J 2021; 40:e107410. [PMID: 33694216 PMCID: PMC8013824 DOI: 10.15252/embj.2020107410] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/27/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022] Open
Abstract
Motile cilia are molecular machines used by a myriad of eukaryotic cells to swim through fluid environments. However, available molecular structures represent only a handful of cell types, limiting our understanding of how cilia are modified to support motility in diverse media. Here, we use cryo-focused ion beam milling-enabled cryo-electron tomography to image sperm flagella from three mammalian species. We resolve in-cell structures of centrioles, axonemal doublets, central pair apparatus, and endpiece singlets, revealing novel protofilament-bridging microtubule inner proteins throughout the flagellum. We present native structures of the flagellar base, which is crucial for shaping the flagellar beat. We show that outer dense fibers are directly coupled to microtubule doublets in the principal piece but not in the midpiece. Thus, mammalian sperm flagella are ornamented across scales, from protofilament-bracing structures reinforcing microtubules at the nano-scale to accessory structures that impose micron-scale asymmetries on the entire assembly. Our structures provide vital foundations for linking molecular structure to ciliary motility and evolution.
Collapse
Affiliation(s)
- Miguel Ricardo Leung
- Cryo‐Electron MicroscopyBijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
- The Division of Structural BiologyWellcome Centre for Human GeneticsThe University of OxfordOxfordUK
| | - Marc C Roelofs
- Cryo‐Electron MicroscopyBijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
| | - Ravi Teja Ravi
- Cryo‐Electron MicroscopyBijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
| | - Paula Maitan
- Department of Equine SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
- Veterinary DepartmentUniversidade Federal de ViçosaViçosaBrazil
| | - Heiko Henning
- Department of Equine SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Min Zhang
- Department of Farm & Animal Health and Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Elizabeth G Bromfield
- Department of Farm & Animal Health and Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
- Priority Research Centre for Reproductive ScienceFaculty of ScienceThe University of NewcastleCallaghanNSWAustralia
| | - Stuart C Howes
- Cryo‐Electron MicroscopyBijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
| | - Bart M Gadella
- Department of Farm & Animal Health and Biomolecular Health SciencesFaculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | - Tzviya Zeev‐Ben‐Mordehai
- Cryo‐Electron MicroscopyBijvoet Center for Biomolecular ResearchUtrecht UniversityUtrechtThe Netherlands
- The Division of Structural BiologyWellcome Centre for Human GeneticsThe University of OxfordOxfordUK
| |
Collapse
|
50
|
Lançoni R, Celeghini ECC, Giuli VD, de Carvalho CPT, Zoca GB, Garcia-Oliveros LN, Batissaco L, Oliveira LZ, de Arruda RP. Coenzyme Q-10 improves preservation of mitochondrial functionality and actin structure of cryopreserved stallion sperm. Anim Reprod 2021; 18:e20200218. [PMID: 33936294 PMCID: PMC8078863 DOI: 10.1590/1984-3143-ar2020-0218] [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] [Indexed: 11/21/2022] Open
Abstract
Coenzyme Q-10 (CoQ-10) is a cofactor for mitochondrial electron transport chain and may be an alternative to improve sperm quality of cryopreserved equine semen. This work aimed to improve stallion semen quality after freezing by adding CoQ-10 to the cryopreservation protocol. Seven saddle stallions were utilized. Each animal was submitted to five semen collections and freezing procedures. For cryopreservation, each ejaculate was divided in three treatments: 1) Botucrio® diluent (control); 2) 50 μmol CoQ-10 added to Botucrio® diluent; 3) 1 mmol CoQ-10 added to Botucrio® diluent. Semen batches were analyzed for sperm motility characteristics (CASA), plasma and acrosomal membranes integrity and mitochondrial membrane potential (by fluorescence probes propidium iodide, Hoechst 33342, FITC-PSA and JC-1, respectively), alterations in cytoskeletal actin (phalloidin-FITC) and mitochondrial function (diaminobenzidine; DAB). The 1 mmol CoQ-10 treatment presented higher (P<0.05) amount (66.8%) of sperm cells with fully stained midpiece (indicating high mitochondrial activity) and higher (P<0.05) amount (81.6%) of cells without actin reorganization to the post-acrosomal region compared to control group (60.8% and 76.0%, respectively). It was concluded that the addition of 1 mmol CoQ-10 to the freezing diluent was more effective in preserving mitochondria functionality and cytoskeleton of sperm cells submitted to cryopreservation process.
Collapse
Affiliation(s)
- Renata Lançoni
- Departamento de Reprodução Animal, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
| | | | - Valdemar De Giuli
- Central Internacional de Reprodução Equina Rancho das Américas, Porto Feliz, SP, Brasil
| | | | | | | | - Leonardo Batissaco
- Departamento de Reprodução Animal, Universidade de São Paulo, Pirassununga, SP, Brasil
| | - Letícia Zoccolaro Oliveira
- Departamento de Clínica Veterinária e Cirurgia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Rubens Paes de Arruda
- Departamento de Reprodução Animal, Universidade de São Paulo, Pirassununga, SP, Brasil
| |
Collapse
|