151
|
Gunes S, Sengupta P, Henkel R, Alguraigari A, Sinigaglia MM, Kayal M, Joumah A, Agarwal A. Microtubular Dysfunction and Male Infertility. World J Mens Health 2018; 38:9-23. [PMID: 30350487 PMCID: PMC6920067 DOI: 10.5534/wjmh.180066] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 01/27/2023] Open
Abstract
Microtubules are the prime component of the cytoskeleton along with microfilaments. Being vital for organelle transport and cellular divisions during spermatogenesis and sperm motility process, microtubules ascertain functional capacity of sperm. Also, microtubule based structures such as axoneme and manchette are crucial for sperm head and tail formation. This review (a) presents a concise, yet detailed structural overview of the microtubules, (b) analyses the role of microtubule structures in various male reproductive functions, and (c) presents the association of microtubular dysfunctions with male infertility. Considering the immense importance of microtubule structures in the formation and maintenance of physiological functions of sperm cells, this review serves as a scientific trigger in stimulating further male infertility research in this direction.
Collapse
Affiliation(s)
- Sezgin Gunes
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey.,American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, MAHSA University, Selangor, Malaysia.,American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ralf Henkel
- Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa.,American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Aabed Alguraigari
- Batterjee Medical College, Jeddah, Saudi Arabia.,American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Mariana Marques Sinigaglia
- University of Sao Paulo, Sao Paulo, Brazil.,American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Malik Kayal
- Alfaisal University Medical School, Riyadh, Saudi Arabia.,American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmad Joumah
- Alfaisal University Medical School, Riyadh, Saudi Arabia.,American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
152
|
Biallelic mutations of CFAP251 cause sperm flagellar defects and human male infertility. J Hum Genet 2018; 64:49-54. [PMID: 30310178 DOI: 10.1038/s10038-018-0520-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 11/08/2022]
Abstract
Multiple morphological abnormalities of flagella (MMAF) are human reproduction disorders due to the dysplastic development of sperm flagella. The spermatozoa of men with MMAF manifest absent, short, coiled, bent, and/or irregular-caliber flagella. Previous studies revealed genetic contributions to human MMAF, but known MMAF-associated genes only explained approximately 50% MMAF cases. In this study, we employed human whole-exome sequencing for genetic analysis and identified biallelic mutations of CFAP251 (cilia- and flagella-associated protein 251, also known as WDR66) in three (5%) of 65 Han Chinese men with MMAF. All these CFAP251 mutations are loss-of-function. The population genome data suggested that these CFAP251 mutations are extremely rare (only heterozygous) or absent from human populations. Our functional assays of gene expression and immunofluorescence staining in a CFAP251-deficient man, together with previous experimental evidence from model organisms, suggested that CFAP251 is involved in flagellar functions. Our observations suggested that CFAP251 is associated with sperm flagellar development and human male infertility.
Collapse
|
153
|
Abbasi F, Miyata H, Shimada K, Morohoshi A, Nozawa K, Matsumura T, Xu Z, Pratiwi P, Ikawa M. RSPH6A is required for sperm flagellum formation and male fertility in mice. J Cell Sci 2018; 131:jcs.221648. [PMID: 30185526 DOI: 10.1242/jcs.221648] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/23/2018] [Indexed: 01/09/2023] Open
Abstract
The flagellum is an evolutionarily conserved appendage used for sensing and locomotion. Its backbone is the axoneme and a component of the axoneme is the radial spoke (RS), a protein complex implicated in flagellar motility regulation. Numerous diseases occur if the axoneme is improperly formed, such as primary ciliary dyskinesia (PCD) and infertility. Radial spoke head 6 homolog A (RSPH6A) is an ortholog of Chlamydomonas RSP6 in the RS head and is evolutionarily conserved. While some RS head proteins have been linked to PCD, little is known about RSPH6A. Here, we show that mouse RSPH6A is testis-enriched and localized in the flagellum. Rsph6a knockout (KO) male mice are infertile as a result of their short immotile spermatozoa. Observation of the KO testis indicates that the axoneme can elongate but is disrupted before accessory structures are formed. Manchette removal is also impaired in the KO testis. Further, RSPH9, another radial spoke protein, disappeared in the Rsph6a KO flagella. These data indicate that RSPH6A is essential for sperm flagellar assembly and male fertility in mice.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Ferheen Abbasi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Haruhiko Miyata
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Akane Morohoshi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kaori Nozawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takafumi Matsumura
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Zoulan Xu
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Putri Pratiwi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan .,Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan.,The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
154
|
RNA processing in the male germline: Mechanisms and implications for fertility. Semin Cell Dev Biol 2018; 79:80-91. [DOI: 10.1016/j.semcdb.2017.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 12/22/2022]
|
155
|
Rapid Evolution of Sperm Produces Diverse Centriole Structures that Reveal the Most Rudimentary Structure Needed for Function. Cells 2018; 7:cells7070067. [PMID: 29949922 PMCID: PMC6071034 DOI: 10.3390/cells7070067] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 11/17/2022] Open
Abstract
Centrioles are ancient subcellular protein-based organelles that maintain a conserved number and structure across many groups of eukaryotes. Centriole number (two per cells) is tightly regulated; each pre-existing centriole nucleates only one centriole as the cell prepares for division. The structure of centrioles is barrel-shaped, with a nine-fold symmetry of microtubules. This organization of microtubules is essential for the ancestral function of centriole–cilium nucleation. In animal cells, centrioles have gained an additional role: recruiting pericentriolar material (PCM) to form a centrosome. Therefore, it is striking that in animal spermatozoa, the centrioles have a remarkable diversity of structures, where some are so anomalous that they are referred to as atypical centrioles and are barely recognizable. The atypical centriole maintains the ability to form a centrosome and nucleate a new centriole, and therefore reveals the most rudimentary structure that is needed for centriole function. However, the atypical centriole appears to be incapable of forming a cilium. Here, we propose that the diversity in sperm centriole structure is due to rapid evolution in the shape of the spermatozoa head and neck. The enhanced diversity may be driven by a combination of direct selection for novel centriole functions and pleiotropy, which eliminates centriole properties that are dispensable in the spermatozoa function.
Collapse
|
156
|
Ali A, Mistry BV, Ahmed HA, Abdulla R, Amer HA, Prince A, Alazami AM, Alkuraya FS, Assiri A. Deletion of DDB1- and CUL4- associated factor-17 (Dcaf17) gene causes spermatogenesis defects and male infertility in mice. Sci Rep 2018; 8:9202. [PMID: 29907856 PMCID: PMC6003934 DOI: 10.1038/s41598-018-27379-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/31/2018] [Indexed: 01/26/2023] Open
Abstract
DDB1– and CUL4–associated factor 17 (Dcaf17) is a member of DCAF family genes that encode substrate receptor proteins for Cullin-RING E3 ubiquitin ligases, which play critical roles in many cellular processes. To unravel the function of DCAF17, we performed expression profiling of Dcaf17 in different tissues of wild type mouse by qRT-PCR and generated Dcaf17 knockout mice by gene targeting. Expression profiling of Dcaf17 showed highest expression in testis. Analyses of Dcaf17 transcripts during post-natal development of testis at different ages displayed gradual increase in Dcaf17 mRNA levels with the age. Although Dcaf17 disruption did not have any effect on female fertility, Dcaf17 deletion led to male infertility due to abnormal sperm development. The Dcaf17−/− mice produced low number of sperm with abnormal shape and significantly low motility. Histological examination of the Dcaf17−/− testis revealed impaired spermatogenesis with presence of vacuoles and sloughed cells in the seminiferous tubules. Disruption of Dcaf17 caused asymmetric acrosome capping, impaired nuclear compaction and abnormal round spermatid to elongated spermatid transition. For the first time, these data indicate that DCAF17 is essential for spermiogenesis.
Collapse
Affiliation(s)
- Asmaa Ali
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Bhavesh V Mistry
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Hala A Ahmed
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Razan Abdulla
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Hassan A Amer
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12613, Egypt
| | - Abdelbary Prince
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12613, Egypt
| | - Anas M Alazami
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Abdullah Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia. .,College of Medicine, AlFaisal University, Riyadh, Saudi Arabia. .,Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| |
Collapse
|
157
|
Liu H, Li W, Zhang Y, Zhang Z, Shang X, Zhang L, Zhang S, Li Y, Somoza AV, Delpi B, Gerton GL, Foster JA, Hess RA, Pazour GJ, Zhang Z. IFT25, an intraflagellar transporter protein dispensable for ciliogenesis in somatic cells, is essential for sperm flagella formation. Biol Reprod 2018; 96:993-1006. [PMID: 28430876 DOI: 10.1093/biolre/iox029] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 04/13/2017] [Indexed: 12/25/2022] Open
Abstract
Intraflagellar transport (IFT) is a conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella. However, IFT25, a component of the IFT complex, is not required for the formation of cilia in somatic tissues. In mice, the gene is highly expressed in the testis, and its expression is upregulated during the final phase when sperm flagella are formed. To investigate the role of IFT25 in sperm flagella formation, the gene was specifically disrupted in male germ cells. All homozygous knockout mice survived to adulthood and did not show any gross abnormalities. However, all homozygous knockout males were completely infertile. Sperm numbers were reduced and these sperm were completely immotile. Multiple morphological abnormalities were observed in sperm, including round heads, short and bent tails, with some tails showing branched flagella and others with frequent abnormal thicknesses, as well as swollen tips of the tail. Transmission electron microscopy revealed that flagellar accessory structures, including the fibrous sheath and outer dense fibers, were disorganized, and most sperm had also lost the "9+2" microtubule structure. In the testis, IFT25 forms a complex with other IFT proteins. In Ift25 knockout testes, IFT27, an IFT25 binding partner, was missing, and IFT20 and IFT81 levels were also reduced. Our findings suggest that IFT25, although not necessary for the formation of cilia in somatic cells, is indispensable for sperm flagellum formation and male fertility in mice.
Collapse
Affiliation(s)
- Hong Liu
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Wei Li
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yong Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Dermatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengang Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuejun Shang
- Department of Andrology, Jinling Hospital, Nanjing University, School of Medicine, Nanjing, China
| | - Ling Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Shiyang Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yanwei Li
- Department of Computer Science, Wellesley College, Wellesley, Massachusetts, USA
| | - Andres V Somoza
- Department of Humanities and Sciences, Honor College, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Brandon Delpi
- Department of Biology, Randolph-Macon College, Ashland, Virginia, USA
| | - George L Gerton
- Center for Research on Reproduction and Women's Health Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James A Foster
- Department of Biology, Randolph-Macon College, Ashland, Virginia, USA
| | - Rex A Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois, USA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zhibing Zhang
- School of Public Health and Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, China.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
158
|
Zhao YQ, Mu DL, Wang D, Han YL, Hou CC, Zhu JQ. Analysis of the function of KIF3A and KIF3B in the spermatogenesis in Boleophthalmus pectinirostris. FISH PHYSIOLOGY AND BIOCHEMISTRY 2018; 44:769-788. [PMID: 29511984 DOI: 10.1007/s10695-017-0461-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 12/18/2017] [Indexed: 06/08/2023]
Abstract
Spermatogenesis represents one of the most complicated morphological transformation procedures. During this process, the assembly and maintenance of the flagella and intracellular transport of membrane-bound organelles required KIF3A and KIF3B. Our main goal was to test KIF3A and KIF3B location during spermatogenesis of Boleophthalmus pectinirostris. We cloned complete cDNA of KIF3A/3B from the testis of B. pectinirostris by PCR and rapid amplification of cDNA ends (RACE). The predicted secondary and tertiary structures of B. pectinirostris KIF3A/3B contained three domains: (a) the head region, (b) the stalk region, and (c) the tail region. Real-time quantitative PCR (qPCR) results revealed that KIF3A and KIF3B mRNA were presented in all the tissues examined, with the highest expression seen in the testis. In situ hybridization (ISH) showed that KIF3A and KIF3B were distributed in the periphery of the nuclear in the spermatocyte and the early spermatid. In the late spermatid and mature sperm, the KIF3A and KIF3B mRNA were gradually gathered to one side where the flagella formed. Immunofluorescence (IF) showed that KIF3A, tubulin, and mitochondria were co-localized in different stages during spermiogenesis in B. pectinirostris. The temporal and spatial expression dynamics of KIF3A/3B indicate that KIF3A and KIF3B might be involved in flagellar assembly and maintenance at the mRNA and protein levels. Moreover, these proteins may transport the mitochondria resulting in flagellum formation in B. pectinirostris.
Collapse
Affiliation(s)
- Yong-Qiang Zhao
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Dan-Li Mu
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Di Wang
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Ying-Li Han
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, People's Republic of China
| | - Cong-Cong Hou
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, People's Republic of China.
| | - Jun-Quan Zhu
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, People's Republic of China.
| |
Collapse
|
159
|
SPAG17 Is Required for Male Germ Cell Differentiation and Fertility. Int J Mol Sci 2018; 19:ijms19041252. [PMID: 29690537 PMCID: PMC5979577 DOI: 10.3390/ijms19041252] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/05/2018] [Accepted: 04/12/2018] [Indexed: 01/08/2023] Open
Abstract
Spag17 encodes a protein present in the axoneme central pair complex of motile cilia and flagella. A mutation in this gene has been reported to be associated with infertility caused by defects in sperm motility. Here, we report that Spag17 knockout mice are infertile because of a severe defect in spermatogenesis. The histological evaluation of testis sections from mutant mice revealed seminiferous tubules with spermatogenesis arrested at the spermatid stage and cell debris in the cauda epididymis. The few sperm collected from the cauda epididymis were immotile and displayed abnormal tail and head morphology. Immunofluorescence analysis of Spag17 knockout germ cells showed spermatids with abnormally long manchette structures and morphological defects in the head. Electron microscopy showed altered manchette microtubules, reduced chromatin condensation, irregular nuclear shape, and detached acrosomes. Additionally, the transport of proteins (Pcdp1 and IFT20) along the manchette microtubules was disrupted in the knockout elongating spermatids. Our results show for the first time that Spag17 is essential for normal manchette structure, protein transport, and formation of the sperm head and flagellum, in addition to its role in sperm motility.
Collapse
|
160
|
Yang K, Adham IM, Meinhardt A, Hoyer-Fender S. Ultra-structure of the sperm head-to-tail linkage complex in the absence of the spermatid-specific LINC component SPAG4. Histochem Cell Biol 2018; 150:49-59. [PMID: 29663073 DOI: 10.1007/s00418-018-1668-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2018] [Indexed: 12/31/2022]
Abstract
Tight connection between sperm head and tail is crucial for the transport of the male genome and fertilization. The linkage complex, the sperm head-to-tail coupling apparatus (HTCA), originates from the centrosome and anchors to the nuclear membrane. In contrast to its ultra-structural organization, which is already well known for decades, its protein composition largely still awaits future deciphering. SUN-domain proteins are essential components of a complex that links the cytoskeleton to the peripheral nucleoskeleton, which is the nuclear lamina. Here, we studied the impact of the SUN protein SPAG4/SUN4 on the formation of the HTCA. SPAG4/SUN4 is specifically expressed in haploid male germ cells showing a polarized distribution towards the posterior pole in late spermatids that corresponds to the tail attachment site. SPAG4-deficient male mice are infertile with compromised manchette formation and malformed sperm heads. Nonetheless, sperm tails are present demonstrating dispensability of a proper manchette for their formation. Ultra-structural analyses revealed that the development of the sperm head-to-tail linkage complex in the absence of SPAG4 resembles that in the wild type. However, in SPAG4-deficient sperm, the attachment site is diminished with obvious lateral detachment of the HTCA from the nucleus. Our results thus indicate that SPAG4, albeit not essential for the formation of the HTCA per se, is, nevertheless, required for tightening the sperm head-to-tail anchorage by provoking the correct attachment of the lateral parts of the basal plate to the implantation fossa.
Collapse
Affiliation(s)
- Kefei Yang
- Department of Developmental Biology, Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology, GZMB, Ernst-Caspari-Haus, Georg-August-Universität Göttingen, Justus-von-Liebig-Weg 11, Göttingen, Germany
| | - Ibrahim M Adham
- Department of Human Genetics, University Medicine, Georg-August-Universität Göttingen, Heinrich-Düker-Weg 12, Göttingen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus-Liebig-University Giessen, Aulweg 123, Giessen, Germany
| | - Sigrid Hoyer-Fender
- Department of Developmental Biology, Johann-Friedrich-Blumenbach-Institute of Zoology and Anthropology, GZMB, Ernst-Caspari-Haus, Georg-August-Universität Göttingen, Justus-von-Liebig-Weg 11, Göttingen, Germany.
| |
Collapse
|
161
|
Dong FN, Amiri-Yekta A, Martinez G, Saut A, Tek J, Stouvenel L, Lorès P, Karaouzène T, Thierry-Mieg N, Satre V, Brouillet S, Daneshipour A, Hosseini SH, Bonhivers M, Gourabi H, Dulioust E, Arnoult C, Touré A, Ray PF, Zhao H, Coutton C. Absence of CFAP69 Causes Male Infertility due to Multiple Morphological Abnormalities of the Flagella in Human and Mouse. Am J Hum Genet 2018; 102:636-648. [PMID: 29606301 DOI: 10.1016/j.ajhg.2018.03.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/05/2018] [Indexed: 10/17/2022] Open
Abstract
The multiple morphological abnormalities of the flagella (MMAF) phenotype is among the most severe forms of sperm defects responsible for male infertility. The phenotype is characterized by the presence in the ejaculate of immotile spermatozoa with severe flagellar abnormalities including flagella being short, coiled, absent, and of irregular caliber. Recent studies have demonstrated that MMAF is genetically heterogeneous, and genes thus far associated with MMAF account for only one-third of cases. Here we report the identification of homozygous truncating mutations (one stop-gain and one splicing variant) in CFAP69 of two unrelated individuals by whole-exome sequencing of a cohort of 78 infertile men with MMAF. CFAP69 encodes an evolutionarily conserved protein found at high levels in the testis. Immunostaining experiments in sperm from fertile control individuals showed that CFAP69 localized to the midpiece of the flagellum, and the absence of CFAP69 was confirmed in both individuals carrying CFPA69 mutations. Additionally, we found that sperm from a Cfap69 knockout mouse model recapitulated the MMAF phenotype. Ultrastructural analysis of testicular sperm from the knockout mice showed severe disruption of flagellum structure, but histological analysis of testes from these mice revealed the presence of all stages of the seminiferous epithelium, indicating that the overall progression of spermatogenesis is preserved and that the sperm defects likely arise during spermiogenesis. Together, our data indicate that CFAP69 is necessary for flagellum assembly/stability and that in both humans and mice, biallelic truncating mutations in CFAP69 cause autosomal-recessive MMAF and primary male infertility.
Collapse
|
162
|
Chen H, Zhu Y, Zhu Z, Zhi E, Lu K, Wang X, Liu F, Li Z, Xia W. Detection of heterozygous mutation in hook microtubule-tethering protein 1 in three patients with decapitated and decaudated spermatozoa syndrome. J Med Genet 2018; 55:150-157. [PMID: 29330334 DOI: 10.1136/jmedgenet-2016-104404] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 07/30/2017] [Accepted: 07/31/2017] [Indexed: 11/04/2022]
Abstract
BACKGROUND The mechanism of intramanchette transport is crucial to the transformation of sperm tail and the nuclear condensation during spermiogenesis. Although few dysfunctional proteins could result in abnormal junction between the head and tail of spermatozoon, little is known about the genetic cues in this process. OBJECTIVE Based on patients with severe decapitated and decaudated spermatozoa (DDS) syndrome, the study aimed to validate whether new mutation exists on their Hook microtubule-tethering protein 1 (HOOK1) genes and follow their results of assisted reproduction treatment (ART). METHODS 7 severe teratozoospermia patients with DDS (proportion >95%) and three relative members in one pedigree were collected to sequence the whole genomic DNA. The fertilisation rates (FRs) of these patients were followed. Morphological observation and interspecies intracytoplasmic sperm injection (ICSI) assays were applied. RESULTS A novel missense mutation of A to G (p.Q286R) in patients with DDS (n=3/7) was found in the HOOK1 gene, which was inherited from the mother in one patient. This variant was absent in 160 fertile population-matched control individuals. Morphological observation showed that almost all the DDS broke into decaudated heads and headless tails at the implantation fossa or the basal plate. The clinical studies indicated that the mutation might cause reduced FRs on both ART (FR=18.07%) and interspecies ICSI (FR=16.98%). CONCLUSIONS An unreported mutation in HOOK1 gene was identified, which might be responsible to some patients with DDS. Further studies need to uncover the molecular mechanism of spermiogenesis for genomic therapy.
Collapse
Affiliation(s)
- Huixing Chen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Med-X Research Institute, Shanghai, China.,Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Zhu
- Shanghai Human Sperm Bank, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zijue Zhu
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Erlei Zhi
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keming Lu
- Shanghai Human Sperm Bank, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaobo Wang
- Shanghai Human Sperm Bank, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Med-X Research Institute, Shanghai, China.,Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Human Sperm Bank, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Med-X Research Institute, Shanghai, China.,Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Human Sperm Bank, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- School of Biomedical Engineering, Shanghai Jiao Tong University, Med-X Research Institute, Shanghai, China
| |
Collapse
|
163
|
Zhang Y, Liu H, Li W, Zhang Z, Zhang S, Teves ME, Stevens C, Foster JA, Campbell GE, Windle JJ, Hess RA, Pazour GJ, Zhang Z. Intraflagellar transporter protein 140 (IFT140), a component of IFT-A complex, is essential for male fertility and spermiogenesis in mice. Cytoskeleton (Hoboken) 2018; 75:70-84. [PMID: 29236364 DOI: 10.1002/cm.21427] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 11/11/2022]
Abstract
Intraflagellar transport (IFT) is a conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella. However, little is known about its role in sperm flagella formation and male fertility. IFT140 is a component of IFT-A complex. In mouse, it is highly expressed in the testis. Ift140 gene was inactivated specifically in mouse spermatocytes/spermatids. The mutant mice did not show any gross abnormalities, but all were infertile and associated with significantly reduced sperm number and motility. Multiple sperm morphological abnormalities were discovered, including amorphous heads, short/bent flagella and swollen tail tips, as well as vesicles along the flagella due to spermiogenesis defects. The epididymides contained round bodies of cytoplasm derived from the sloughing of the cytoplasmic lobes and residual bodies. Knockout of Ift140 did not significantly affect testicular expression levels of selective IFT components but localization of IFT27 and IFT88, two components of IFT-B complex, was changed. Our findings demonstrate that IFT140 is a key regulator for male fertility and normal spermiogenesis in mice. It not only plays a role in sperm flagella assembling, but is also involved in critical assembly of proteins that interface between the germ cell plasma and the Sertoli cell.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Hong Liu
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,School of Public Health, Wuhan University of Science and Technology, Wuhan 430060, Hubei
| | - Wei Li
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Zhengang Zhang
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,Department of Gastroenterology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei
| | - Shiyang Zhang
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,School of Public Health, Wuhan University of Science and Technology, Wuhan 430060, Hubei
| | - Maria E Teves
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Courtney Stevens
- Department of Biology, Randolph-Macon College, Ashland, Virginia 23005
| | - James A Foster
- Department of Biology, Randolph-Macon College, Ashland, Virginia 23005
| | - Gregory E Campbell
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Jolene J Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia 23298
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, 2001 S. Lincoln, Urbana, Illinois 61802-6199
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Zhibing Zhang
- Department of Obstetrics & Gynecology, Virginia Commonwealth University, Richmond, Virginia 23298.,School of Public Health, Wuhan University of Science and Technology, Wuhan 430060, Hubei
| |
Collapse
|
164
|
Martins LR, Bung RK, Koch S, Richter K, Schwarzmüller L, Terhardt D, Kurtulmus B, Niehrs C, Rouhi A, Lohmann I, Pereira G, Fröhling S, Glimm H, Scholl C. Stk33 is required for spermatid differentiation and male fertility in mice. Dev Biol 2018; 433:84-93. [DOI: 10.1016/j.ydbio.2017.11.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 11/29/2022]
|
165
|
Liu M, Ru Y, Gu Y, Tang J, Zhang T, Wu J, Yu F, Yuan Y, Xu C, Wang J, Shi H. Disruption of Ssp411 causes impaired sperm head formation and male sterility in mice. Biochim Biophys Acta Gen Subj 2017; 1862:660-668. [PMID: 29247744 DOI: 10.1016/j.bbagen.2017.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/10/2017] [Accepted: 12/12/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND We previously cloned the Ssp411 gene. We found that the Ssp411 protein is predominantly expressed in elongated spermatids in the rat testis in a stage-dependent manner. Although our findings strongly suggested that Ssp411 might play an important role in mammalian spermatogenesis, this hypothesis has not been studied. METHODS We first used real-time PCR, Western blotting and immunohistochemistry to confirm that the expression pattern of Ssp411 in several murine tissues is similar to its expression pattern in corresponding rat tissues. To better understand the roles of Ssp411 in male reproduction in vivo, we identified and characterized an Ssp411 expression-disrupted murine strain (Ssp411PB/PB) that was generated by piggyBac (PB) transposon insertion. We studied Ssp411-interacting proteins using proteome microarray, co-IP and GST pull-down assay. RESULTS Both Ssp411 mRNA and protein were detected exclusively in spermatids after step 9 during spermiogenesis in testis. Phenotypic analysis suggested that only Ssp411PB/PB males are sterile. These males have smaller testes, reduced sperm counts, decreased sperm motility and deformed spermatozoa. Microscopy analysis indicated that the manchette, a structurally reshaped sperm head, is aberrant in Ssp411PB/PB spermatids. The results of proteome microarray analysis and GST pull-down assays suggested that Ssp411 participates the ubiquitin-proteasome system by interacting with PSMC3. This has been reported to be manchette-associated and important for the head shaping of spermatids. CONCLUSIONS Our study suggested that Ssp411 is required for spermiogenesis. It seems to play a role in sperm head shaping. The lack of Ssp411 causes sperm deformation and results in male infertility. GENERAL SIGNIFICANCE Ssp411PB/PB mouse strain is an animal model of idiopathic oligoasthenoteratozoospermia (iOAT), and the gene may represent a therapeutic target for iOAT patients.
Collapse
Affiliation(s)
- Miao Liu
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Yanfei Ru
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Yihua Gu
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Jianan Tang
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Tiancheng Zhang
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Jun Wu
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Fudong Yu
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Yao Yuan
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China
| | - Chen Xu
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, China
| | - Jian Wang
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China.
| | - Huijuan Shi
- NHFPC Key Lab of Reproduction Regulation, SIPPR, Pharmacy School, Fudan University, China.
| |
Collapse
|
166
|
Dunleavy JEM, Okuda H, O’Connor AE, Merriner DJ, O’Donnell L, Jamsai D, Bergmann M, O’Bryan MK. Katanin-like 2 (KATNAL2) functions in multiple aspects of haploid male germ cell development in the mouse. PLoS Genet 2017; 13:e1007078. [PMID: 29136647 PMCID: PMC5705150 DOI: 10.1371/journal.pgen.1007078] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 11/28/2017] [Accepted: 10/16/2017] [Indexed: 11/19/2022] Open
Abstract
The katanin microtubule-severing proteins are essential regulators of microtubule dynamics in a diverse range of species. Here we have defined critical roles for the poorly characterised katanin protein KATNAL2 in multiple aspects of spermatogenesis: the initiation of sperm tail growth from the basal body, sperm head shaping via the manchette, acrosome attachment, and ultimately sperm release. We present data suggesting that depending on context, KATNAL2 can partner with the regulatory protein KATNB1 or act autonomously. Moreover, our data indicate KATNAL2 may regulate δ- and ε-tubulin rather than classical α-β-tubulin microtubule polymers, suggesting the katanin family has a greater diversity of function than previously realised. Together with our previous research, showing the essential requirement of katanin proteins KATNAL1 and KATNB1 during spermatogenesis, our data supports the concept that in higher order species the presence of multiple katanins has allowed for subspecialisation of function within complex cellular settings such as the seminiferous epithelium. Male infertility affects one in twenty men of reproductive age in western countries. Despite this, the biochemical basis of common defects, including reduced sperm count and abnormal sperm structure and function, remains poorly defined. Microtubules are cellular “scaffolds” that serve critical roles in all cells, including developing male germ cells wherein they facilitate mitosis and meiosis (cell division), sperm head remodelling and sperm tail formation. The precise regulation of microtubule number, length and movement is thus, essential for male fertility. Within this manuscript, we have used spermatogenesis to define the function of the putative microtubule-severing protein katanin-like 2 (KATNAL2). We show that mice with compromised KATNAL2 function are male sterile as a consequence of defects in the structural remodelling of germ cells. Notably, we show the function of microtubule-based structures involved in sperm head shaping and tail formation are disrupted. Further, we show for the first time, that KATNAL2 can function both independently or in concert with the katanin regulatory protein KATNB1 and that it can target the poorly characterized tubulin subunits delta and epsilon. Our research has immediate relevance to the origins of human male infertility and provides novel insights into aspects of microtubule regulation relevant to numerous tissues and species.
Collapse
Affiliation(s)
- Jessica E. M. Dunleavy
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and The Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria; Australia
| | - Hidenobu Okuda
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
| | - Anne E. O’Connor
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
| | - D. Jo Merriner
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
| | - Liza O’Donnell
- Hudson Institute of Medical Research and Department of Molecular and Translational Science, Monash University, Melbourne, Victoria; Australia
| | - Duangporn Jamsai
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and The Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria; Australia
| | - Martin Bergmann
- Institute of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Hesse; Germany
| | - Moira K. O’Bryan
- School of Biological Sciences, Monash University, Melbourne, Victoria; Australia
- * E-mail:
| |
Collapse
|
167
|
Intraflagellar transporter protein (IFT27), an IFT25 binding partner, is essential for male fertility and spermiogenesis in mice. Dev Biol 2017; 432:125-139. [PMID: 28964737 DOI: 10.1016/j.ydbio.2017.09.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/04/2017] [Accepted: 09/18/2017] [Indexed: 01/08/2023]
Abstract
Intraflagellar transport (IFT) is an evolutionarily conserved mechanism essential for the assembly and maintenance of most eukaryotic cilia and flagella. In mice, mutations in IFT proteins have been shown to cause several ciliopathies including retinal degeneration, polycystic kidney disease, and hearing loss. However, little is known about its role in the formation of the sperm tail, which has the longest flagella of mammalian cells. IFT27 is a component of IFT-B complex and binds to IFT25 directly. In mice, IFT27 is highly expressed in the testis. To investigate the role of IFT27 in male germ cells, the floxed Ift27 mice were bred with Stra8-iCre mice so that the Ift27 gene was disrupted in spermatocytes/spermatids. The Ift27: Stra8-iCre mutant mice did not show any gross abnormalities, and all of the mutant mice survived to adulthood. There was no difference between testis weight/body weight between controls and mutant mice. All adult homozygous mutant males examined were completely infertile. Histological examination of the testes revealed abnormally developed germ cells during the spermiogenesis phase. The epididymides contained round bodies of cytoplasm. Sperm number was significantly reduced compared to the controls and only about 2% of them remained significantly reduced motility. Examination of epididymal sperm by light microscopy and SEM revealed multiple morphological abnormalities including round heads, short and bent tails, abnormal thickness of sperm tails in some areas, and swollen tail tips in some sperm. TEM examination of epididymal sperm showed that most sperm lost the "9+2″ axoneme structure, and the mitochondria sheath, fibrous sheath, and outer dense fibers were also disorganized. Some sperm flagella also lost cell membrane. Levels of IFT25 and IFT81 were significantly reduced in the testis of the conditional Ift27 knockout mice, and levels of IFT20, IFT74, and IFT140 were not changed. Sperm lipid rafts, which were disrupted in the conditional Ift25 knockout mice, appeared to be normal in the conditional Ift27 knockout mice. Our findings suggest that like IFT25, IFT27, even though not required for ciliogenesis in somatic cells, is essential for sperm flagella formation, sperm function, and male fertility in mice. IFT25 and IFT27 control sperm formation/function through many common mechanisms, but IFT25 has additional roles beyond IFT27.
Collapse
|
168
|
Lehti MS, Sironen A. Formation and function of sperm tail structures in association with sperm motility defects†. Biol Reprod 2017; 97:522-536. [DOI: 10.1093/biolre/iox096] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022] Open
|
169
|
Zhao YQ, Yang HY, Zhang DD, Han YL, Hou CC, Zhu JQ. Dynamic transcription and expression patterns of KIF3A and KIF3B genes during spermiogenesis in the shrimp, Palaemon carincauda. Anim Reprod Sci 2017; 184:59-77. [PMID: 28689636 DOI: 10.1016/j.anireprosci.2017.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/29/2017] [Accepted: 06/22/2017] [Indexed: 01/20/2023]
Abstract
Spermiogenesis is a highly ordered and complex process in the male germ cell differentiation. The microtubule-based motor proteins KIF3A and KIF3B are required for the progression of the stages of spermiogenesis. In this study, the main goal was to determine whether KIF3A and KIF3B have a key role in spermiogenesis in Palaemon carincauda. The complete cDNA of KIF3A/3B from the testis of P. carincauda was cloned by using PCR and rapid amplification of cDNA ends (RACE). The predicted secondary and tertiary structures of KIF3A/3B contained three domains which were the: a) head region, b) stalk region, and c) tail region. Real-time quantitative PCR (qPCR) results revealed that KIF3A and KIF3B mRNAs were obtained for all the tissues examined, with the greatest gene expression in the testis. In situ hybridization indicated the KIF3A and KIF3B mRNAs were distributed in the periphery of the nuclear in the early spermatid of spermiogenesis. In the middle and late spermatid stages, KIF3A and KIF3B mRNAs were gradually upregulated and assembled to one side where acrosome biogenesis begins. In the mature sperm, KIF3A and KIF3B mRNAs were distributed in the acrosome cap and spike. Immunofluorescence studies indicated that KIF3A, tubulin, mitochondria, and Golgi were co-localized in different stages during spermiogenesis in P. carincauda. The temporal and spatial gene expression dynamics of KIF3A/3B indicate that KIF3A and KIF3B proteins may be involved in acrosome formation and nucleus shaping. Moreover, these proteins can transport the mitochondria and Golgi that facilitate acrosome formation in P. carincauda.
Collapse
Affiliation(s)
- Yong-Qiang Zhao
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Hai-Yan Yang
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Dan-Dan Zhang
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Ying-Li Han
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Cong-Cong Hou
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China
| | - Jun-Quan Zhu
- Key Laboratory of Applied Marine Biotechnology by the Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315211, People's Republic of China.
| |
Collapse
|
170
|
Pleuger C, Fietz D, Hartmann K, Schuppe HC, Weidner W, Kliesch S, Baker M, O'Bryan MK, Bergmann M. Expression of ciliated bronchial epithelium 1 during human spermatogenesis. Fertil Steril 2017; 108:47-54. [DOI: 10.1016/j.fertnstert.2017.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/05/2017] [Accepted: 05/14/2017] [Indexed: 12/19/2022]
|
171
|
Ma DD, Pan MY, Hou CC, Tan FQ, Yang WX. KIFC1 and myosin Va: two motors for acrosomal biogenesis and nuclear shaping during spermiogenesis of Portunus trituberculatus. Cell Tissue Res 2017. [DOI: 10.1007/s00441-017-2638-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
172
|
Abstract
Myosin VI (MVI) is a versatile actin-based motor protein that has been implicated in a variety of different cellular processes, including endo- and exocytic vesicle trafficking, Golgi morphology, and actin structure stabilization. A role for MVI in crucial actin-based processes involved in sperm maturation was demonstrated in Drosophila. Because of the prominence and importance of actin structures in mammalian spermiogenesis, we investigated whether MVI was associated with actin-mediated maturation events in mammals. Both immunofluorescence and ultrastructural analyses using immunogold labeling showed that MVI was strongly linked with key structures involved in sperm development and maturation. During the early stage of spermiogenesis, MVI is associated with the Golgi and with coated and uncoated vesicles, which fuse to form the acrosome. Later, as the acrosome spreads to form a cap covering the sperm nucleus, MVI is localized to the acroplaxome, an actin-rich structure that anchors the acrosome to the nucleus. Finally, during the elongation/maturation phase, MVI is associated with the actin-rich structures involved in nuclear shaping: the acroplaxome, manchette, and Sertoli cell actin hoops. Since this is the first report of MVI expression and localization during mouse spermiogenesis and MVI partners in developing sperm have not yet been identified, we discuss some probable roles for MVI in this process. During early stages, MVI is hypothesized to play a role in Golgi morphology and function as well as in actin dynamics regulation important for attachment of developing acrosome to the nuclear envelope. Next, the protein might also play anchoring roles to help generate forces needed for spermatid head elongation. Moreover, association of MVI with actin that accumulates in the Sertoli cell ectoplasmic specialization and other actin structures in surrounding cells suggests additional MVI functions in spermatid movement across the seminiferous epithelium and in sperm release.
Collapse
|
173
|
Simón L, Funes AK, Yapur MA, Cabrillana ME, Monclus MA, Boarelli PV, Vincenti AE, Saez Lancellotti TE, Fornés MW. Manchette-acrosome disorders during spermiogenesis and low efficiency of seminiferous tubules in hypercholesterolemic rabbit model. PLoS One 2017; 12:e0172994. [PMID: 28241054 PMCID: PMC5328279 DOI: 10.1371/journal.pone.0172994] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/13/2017] [Indexed: 12/31/2022] Open
Abstract
Hypercholesterolemia is a marker for several adult chronic diseases. Recently we demonstrated that sub/infertility is also associated to Hypercholesterolemia in rabbits. Seminal alterations included: abnormal sperm morphology, decreased sperm number and declined percentage of motile sperm, among others. In this work, our objective was to evaluate the effects of hypercholesterolemia on testicular efficiency and spermiogenesis, as the latter are directly related to sperm number and morphology respectively. Tubular efficiency was determined by comparing total number of spermatogenic cells with each cell type within the proliferation/differentiation compartments. We found lower testicular efficiency related to both a decrease in spermatogonial cells and an increase in germ cell apoptosis in hypercholesterolemic rabbits. On the other hand, spermiogenesis-the last step of spermatogenesis involved in sperm shaping-was detaily analyzed, particularly the acrosome-nucleus-manchette complex. The manchette is a microtubular-based temporary structure responsible in sperm cell elongation. We analyzed the contribution of actin filaments and raft microdomains in the arrangement of the manchette. Under fluorescence microscopy, spermatocyte to sperm cell development was followed in cells isolated from V to VIII tubular stages. In cells from hypercholesterolemic rabbits, abnormal development of acrosome, nucleus and inaccurate tail implantation were associated with actin-alpha-tubulin-GM1 sphingolipid altered distribution. Morphological alterations were also observed at electron microscopy. We demonstrated for the first time that GM1-enriched microdomains together with actin filaments and microtubules are involved in allowing the correct anchoring of the manchette complex. In conclusion, cholesterol enriched diets promote male fertility alterations by affecting critical steps in sperm development: spermatogenesis and spermiogenesis. It was also demonstrated that hypercholesterolemic rabbit model is a useful tool to study serum cholesterol increment linked to sub/infertility.
Collapse
Affiliation(s)
- Layla Simón
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
- Instituto de investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - Abi K. Funes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Martín A. Yapur
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María E. Cabrillana
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
- Instituto de investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - María A. Monclus
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
- Instituto de investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - Paola V. Boarelli
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
- Instituto de investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - Amanda E. Vincenti
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Tania E. Saez Lancellotti
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
- Instituto de investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - Miguel W. Fornés
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM). Instituto y Área de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo y Centro Científico Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
- Instituto de investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
- * E-mail:
| |
Collapse
|
174
|
Okuda H, DeBoer K, O'Connor AE, Merriner DJ, Jamsai D, O'Bryan MK. LRGUK1 is part of a multiprotein complex required for manchette function and male fertility. FASEB J 2016; 31:1141-1152. [PMID: 28003339 DOI: 10.1096/fj.201600909r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/28/2016] [Indexed: 11/11/2022]
Abstract
Infertility occurs in 1 in 20 young men and is idiopathic in origin in most. We have reported that the leucine-rich repeat (LRR) and guanylate kinase-like domain containing, isoform (LRGUK)-1 is essential for sperm head shaping, via the manchette, and the initiation of sperm tail growth from the centriole/basal body, and thus, male fertility. Within this study we have used a yeast 2-hybrid screen of an adult testis library to identify LRGUK1-binding partners, which were then validated with a range of techniques. The data indicate that LRGUK1 likely achieves its function in partnership with members of the HOOK family of proteins (HOOK-1-3), Rab3-interacting molecule binding protein (RIMBP)-3 and kinesin light chain (KLC)-3, all of which are associated with intracellular protein transport as cargo adaptor proteins and are localized to the manchette. LRGUK1 consists of 3 domains; an LRR, a guanylate kinase (GUK)-like and an unnamed domain. In the present study, we showed that the GUK-like domain is essential for binding to HOOK2 and RIMBP3, and the LRR domain is essential for binding to KLC3. These findings establish LRGUK1 as a key component of a multiprotein complex with an essential role in microtubule dynamics within haploid male germ cells.-Okuda, H., DeBoer, K., O'Connor, A. E., Merriner, D. J., Jamsai, D., O'Bryan, M. K. LRGUK1 is part of a multiprotein complex required for manchette function and male fertility.
Collapse
Affiliation(s)
- Hidenobu Okuda
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; and.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Kathleen DeBoer
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; and.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Anne E O'Connor
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; and.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - D Jo Merriner
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; and.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Duangporn Jamsai
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; and.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Moira K O'Bryan
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; and .,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
175
|
The control of male fertility by spermatid-specific factors: searching for contraceptive targets from spermatozoon's head to tail. Cell Death Dis 2016; 7:e2472. [PMID: 27831554 PMCID: PMC5260884 DOI: 10.1038/cddis.2016.344] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 09/18/2016] [Accepted: 09/26/2016] [Indexed: 02/06/2023]
Abstract
Male infertility due to abnormal spermatozoa has been reported in both animals and humans, but its pathogenic causes, including genetic abnormalities, remain largely unknown. On the other hand, contraceptive options for men are limited, and a specific, reversible and safe method of male contraception has been a long-standing quest in medicine. Some progress has recently been made in exploring the effects of spermatid-specifical genetic factors in controlling male fertility. A comprehensive search of PubMed for articles and reviews published in English before July 2016 was carried out using the search terms 'spermiogenesis failure', 'globozoospermia', 'spermatid-specific', 'acrosome', 'infertile', 'manchette', 'sperm connecting piece', 'sperm annulus', 'sperm ADAMs', 'flagellar abnormalities', 'sperm motility loss', 'sperm ion exchanger' and 'contraceptive targets'. Importantly, we have opted to focus on articles regarding spermatid-specific factors. Genetic studies to define the structure and physiology of sperm have shown that spermatozoa appear to be one of the most promising contraceptive targets. Here we summarize how these spermatid-specific factors regulate spermiogenesis and categorize them according to their localization and function from spermatid head to tail (e.g., acrosome, manchette, head-tail conjunction, annulus, principal piece of tail). In addition, we emphatically introduce small-molecule contraceptives, such as BRDT and PPP3CC/PPP3R2, which are currently being developed to target spermatogenic-specific proteins. We suggest that blocking the differentiation of haploid germ cells, which rarely affects early spermatogenic cell types and the testicular microenvironment, is a better choice than spermatogenic-specific proteins. The studies described here provide valuable information regarding the genetic and molecular defects causing male mouse infertility to improve our understanding of the importance of spermatid-specific factors in controlling fertility. Although a male contraceptive 'pill' is still many years away, research into the production of new small-molecule contraceptives targeting spermatid-specific proteins is the right avenue.
Collapse
|
176
|
Zhang Z, Li W, Zhang Y, Zhang L, Teves ME, Liu H, Strauss JF, Pazour GJ, Foster JA, Hess RA, Zhang Z. Intraflagellar transport protein IFT20 is essential for male fertility and spermiogenesis in mice. Mol Biol Cell 2016; 27:mbc.E16-05-0318. [PMID: 27682589 PMCID: PMC5170554 DOI: 10.1091/mbc.e16-05-0318] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/06/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022] Open
Abstract
Intraflagellar transport (IFT) is a conserved mechanism thought to be essential for the assembly and maintenance of cilia and flagella. However, little is known about its role in mammalian sperm flagella formation. To fill this gap, we disrupted the Ift20 gene in male germ cells. Homozygous mutant mice were infertile with significantly reduced sperm counts and motility. In addition, abnormally shaped elongating spermatid heads and bulbous round spermatids were found in the lumen of the seminiferous tubules. Electron microscopy revealed increased cytoplasmic vesicles, fiber-like structures, abnormal accumulation of mitochondria and a decrease in mature lysosomes. The few developed sperm had disrupted axonemes and some retained cytoplasmic lobe components on the flagella. ODF2 and SPAG16L, two sperm flagella proteins failed to be incorporated into sperm tails of the mutant mice, and in the germ cells, both were assembled into complexes with lighter density in the absence of IFT20. Disrupting IFT20 did not significantly change expression levels of IFT88, a component of IFT-B complex, and IFT140, a component of IFT-A complex. Even though the expression level of an autophagy core protein that associates with IFT20, ATG16, was reduced in the testis of the Ift20 mutant mice, expression levels of other major autophagy markers, including LC3 and ubiquitin were not changed. Our studies suggest that IFT20 is essential for male fertility and spermiogenesis in mice, and its major function is to transport cargo proteins for sperm flagella formation. It also appears to be involved in removing excess cytoplasmic components.
Collapse
Affiliation(s)
- Zhengang Zhang
- Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China, 430030 Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Wei Li
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Yong Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298 Department of Dermatology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China, 430030
| | - Ling Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298 School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Maria E Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Hong Liu
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298 School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - James A Foster
- Department of Biology, Randolph-Macon College, Ashland, VA 23005
| | - Rex A Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois, 2001 S. Lincoln, Urbana, IL 61802-6199
| | - Zhibing Zhang
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| |
Collapse
|
177
|
Fraser R, Lin CJ. Epigenetic reprogramming of the zygote in mice and men: on your marks, get set, go! Reproduction 2016; 152:R211-R222. [PMID: 27601712 PMCID: PMC5097126 DOI: 10.1530/rep-16-0376] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/02/2016] [Indexed: 12/19/2022]
Abstract
Gametogenesis (spermatogenesis and oogenesis) is accompanied by the acquisition of gender-specific epigenetic marks, such as DNA methylation, histone modifications and regulation by small RNAs, to form highly differentiated, but transcriptionally silent cell-types in preparation for fertilisation. Upon fertilisation, extensive global epigenetic reprogramming takes place to remove the previously acquired epigenetic marks and produce totipotent zygotic states. It is the aim of this review to delineate the cellular and molecular events involved in maternal, paternal and zygotic epigenetic reprogramming from the time of gametogenesis, through fertilisation, to the initiation of zygotic genome activation for preimplantation embryonic development. Recent studies have begun to uncover the indispensable functions of epigenetic players during gametogenesis, fertilisation and preimplantation embryo development, and a more comprehensive understanding of these early events will be informative for increasing pregnancy success rates, adding particular value to assisted fertility programmes.
Collapse
Affiliation(s)
- Rupsha Fraser
- The University of EdinburghMRC Centre for Reproductive Health, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Chih-Jen Lin
- The University of EdinburghMRC Centre for Reproductive Health, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| |
Collapse
|