1
|
Noguchi Y, Onodera Y, Miyamoto T, Maruoka M, Kosako H, Suzuki J. In vivo CRISPR screening directly targeting testicular cells. CELL GENOMICS 2024; 4:100510. [PMID: 38447574 PMCID: PMC10943590 DOI: 10.1016/j.xgen.2024.100510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/10/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024]
Abstract
CRISPR-Cas9 short guide RNA (sgRNA) library screening is a powerful approach to understand the molecular mechanisms of biological phenomena. However, its in vivo application is currently limited. Here, we developed our previously established in vitro revival screening method into an in vivo one to identify factors involved in spermatogenesis integrity by utilizing sperm capacitation as an indicator. By introducing an sgRNA library into testicular cells, we successfully pinpointed the retinal degeneration 3 (Rd3) gene as a significant factor in spermatogenesis. Single-cell RNA sequencing (scRNA-seq) analysis highlighted the high expression of Rd3 in round spermatids, and proteomics analysis indicated that Rd3 interacts with mitochondria. To search for cell-type-specific signaling pathways based on scRNA-seq and proteomics analyses, we developed a computational tool, Hub-Explorer. Through this, we discovered that Rd3 modulates oxidative stress by regulating mitochondrial distribution upon ciliogenesis induction. Collectively, our screening system provides a valuable in vivo approach to decipher molecular mechanisms in biological processes.
Collapse
Affiliation(s)
- Yuki Noguchi
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto 606-8501, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto 606-8501, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, N15W7 Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Tatsuo Miyamoto
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Masahiro Maruoka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto 606-8501, Japan; Center for Integrated Biosystems, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Jun Suzuki
- Graduate School of Biostudies, Kyoto University, Konoe-cho, Yoshida, Sakyoku, Kyoto 606-8501, Japan; Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida-Honmachi, Sakyoku, Kyoto 606-8501, Japan; Center for Integrated Biosystems, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
2
|
Mebendazole-Induced Blood-Testis Barrier Injury in Mice Testes by Disrupting Microtubules in Addition to Triggering Programmed Cell Death. Int J Mol Sci 2022; 23:ijms23084220. [PMID: 35457043 PMCID: PMC9029725 DOI: 10.3390/ijms23084220] [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] [Received: 03/18/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 01/27/2023] Open
Abstract
Mebendazole (MBZ) is a synthetic benzimidazole known for its antiparasitic properties. In recent years, growing evidence showed that MBZ was also used as an anti-tumor agent. However, whether (and to what extent) this drug treatment affected the male reproductive system was not well-understood. In this study, male C57BL/6 mice were injected with 40 mg/kg/day of MBZ. The treatment was for 3 and 7 days. Our results showed that the injected mice exhibited an abnormal spermatogenic phase with a significant decrease in sperm. We further detected microtubule disruption and transient functional destruction of the blood–testes barrier (BTB) in the MBZ-injected mice testes (BTB). Our data confirmed that MBZ suppressed the expression of the BTB junction-associated proteins and disrupted the Sertoli cells’ function in vivo. Moreover, MBZ-treated mice demonstrated an aberrant caspase-3 signalling pathway, which resulted in the apoptosis of the germ cells. Here, we present our data, indicating that MBZ impairs BTB by reducing the expression of the microtubules’ and BTB junction-associated proteins. The last leads to activating the caspase-3 pathway, which triggers extensive germ cell apoptosis.
Collapse
|
3
|
Darbey A, Rebourcet D, Curley M, Kilcoyne K, Jeffery N, Reed N, Milne L, Roesl C, Brown P, Smith LB. A comparison of in vivo viral targeting systems identifies adeno-associated virus serotype 9 (AAV9) as an effective vector for genetic manipulation of Leydig cells in adult mice. Andrology 2020; 9:460-473. [PMID: 32996275 DOI: 10.1111/andr.12915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/01/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite the increasing popularity of deliverable transgenics, a robust and fully validated method for targeting Leydig cells, capable of delivering long-term transgene expression, is yet to be defined. OBJECTIVES We compared three viral vector systems in terms of their cell targeting specificity, longevity of gene expression and impact on targeted cell types when delivered to the interstitial compartment of the mouse testis. MATERIALS & METHODS We delivered lentiviral, adenoviral and adeno-associated (AAV) viral particles to the interstitial compartment of adult mouse testis. Immunolocalization and stereology were performed to characterize ability of vectors to target and deliver transgenes to Leydig cells. RESULTS Viral vectors utilized in this study were found to specifically target Leydig cells when delivered interstitially. Transgene expression in lentiviral-targeted Leydig cells was detected for 7 days post-injection before Leydig cells underwent apoptosis. Adenoviral-delivered transgene expression was detected for 10 days post-injection with no evidence of targeted cell apoptosis. We found serotype differences in AAV injected testis with AAV serotype 9 targeting a significant proportion of Leydig cells. Targeting efficiency increased to an average of 59.63% (and a maximum of 80%) of Leydig cells with the addition of neuraminidase during injection. In AAV injected testis sections, transgene expression was detectable for up to 50 days post-injection. DISCUSSION & CONCLUSION Lentivirus, Adenovirus and Adeno-Associated virus delivery to the testis resulted in key variances in targeting efficiency of Leydig cells and in longevity of transgene expression, but identified AAV9 + Neuraminidase as an efficient vector system for transgene delivery and long-term expression. Simple viral delivery procedures and the commercial availability of viral vectors suggests AAV9 + Neuraminidase will be of significant utility to researchers investigating the genetics underpinning Leydig cell function and holds promise to inform the development of novel therapeutics for the treatment of male reproductive disorders.
Collapse
Affiliation(s)
- Annalucia Darbey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Diane Rebourcet
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Michael Curley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Karen Kilcoyne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Nathan Jeffery
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Natalie Reed
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Cornelia Roesl
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Pamela Brown
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
4
|
Pastén-Hidalgo K, Riverón-Negrete L, Sicilia-Argumedo G, Canul-Medina G, Salazar-Anzures T, Tapia-Rodríguez M, Hernández-González EO, Roa-Espitia AL, Cedillo-Peláez C, Fernandez-Mejia C. Dietary Biotin Supplementation Impairs Testis Morphology and Sperm Quality. J Med Food 2019; 23:535-544. [PMID: 31660770 DOI: 10.1089/jmf.2019.0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Supplements containing pharmacological concentrations of biotin are commercially available over the counter. Classical toxicity studies have considered biotin administration as harmless; however, recent investigations have shown that biotin supplementation modifies tissue morphology without changes in toxicity markers, raising concerns about the consequences of morphological changes on tissues' functions and the safety of pharmacological concentrations of the vitamin. Testes are very sensitive to toxicants, and testicular histology is a reliable method to study its function. In this work, we investigated the effects of dietary biotin supplementation on testis morphology and spermatogenesis function using an experimental model, in which we have not observed unfavorable effects on other tissue functions or toxicity markers. Male BALB/cAnNHsd mice were fed a control or a biotin-supplemented diet (1.76 or 97.7 mg biotin/kg diet) for 8 weeks. Compared to the control group, the biotin-supplemented mice presented remarkable testis morphology changes, including increased spermatogonia layers; the cellular mechanism involved is related to increased proliferation. Sperm count and serum testosterone levels were not affected, but spermatozoa motility and morphology were significantly impaired in the biotin-supplemented mice. These results caution against the use of supplements with high concentrations of biotin and indicate that biotin's pharmacological effects on morphology need to be considered in toxicological studies.
Collapse
Affiliation(s)
| | - Leticia Riverón-Negrete
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Gloria Sicilia-Argumedo
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Gustavo Canul-Medina
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Tonatiuh Salazar-Anzures
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Miguel Tapia-Rodríguez
- Unidad de Microscopía, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | | | - Ana Lilia Roa-Espitia
- Departamento de Biología Celular, CINVESTAV-IPN, San Pedro Zacatenco, Mexico City, Mexico
| | - Carlos Cedillo-Peláez
- Departamento de Inmunología Experimental, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Cristina Fernandez-Mejia
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Pediatría, Mexico City, Mexico
| |
Collapse
|
5
|
Wen Q, Tang EI, Lui WY, Lee WM, Wong CKC, Silvestrini B, Cheng CY. Dynein 1 supports spermatid transport and spermiation during spermatogenesis in the rat testis. Am J Physiol Endocrinol Metab 2018; 315:E924-E948. [PMID: 30016153 PMCID: PMC6293164 DOI: 10.1152/ajpendo.00114.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/26/2018] [Accepted: 07/10/2018] [Indexed: 12/22/2022]
Abstract
In the mammalian testis, spermatogenesis is dependent on the microtubule (MT)-specific motor proteins, such as dynein 1, that serve as the engine to support germ cell and organelle transport across the seminiferous epithelium at different stages of the epithelial cycle. Yet the underlying molecular mechanism(s) that support this series of cellular events remain unknown. Herein, we used RNAi to knockdown cytoplasmic dynein 1 heavy chain (Dync1h1) and an inhibitor ciliobrevin D to inactivate dynein in Sertoli cells in vitro and the testis in vivo, thereby probing the role of dynein 1 in spermatogenesis. Both treatments were shown to extensively induce disruption of MT organization across Sertoli cells in vitro and the testis in vivo. These changes also perturbed the transport of spermatids and other organelles (such as phagosomes) across the epithelium. These changes thus led to disruption of spermatogenesis. Interestingly, the knockdown of dynein 1 or its inactivation by ciliobrevin D also perturbed gross disruption of F-actin across the Sertoli cells in vitro and the seminiferous epithelium in vivo, illustrating there are cross talks between the two cytoskeletons in the testis. In summary, these findings confirm the role of cytoplasmic dynein 1 to support the transport of spermatids and organelles across the seminiferous epithelium during the epithelial cycle of spermatogenesis.
Collapse
Affiliation(s)
- Qing Wen
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, New York
| | - Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, New York
| | - Wing-Yee Lui
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Will M Lee
- School of Biological Sciences, University of Hong Kong, Hong Kong, China
| | - Chris K C Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | | | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, New York
| |
Collapse
|
6
|
Hori JI, Koga D, Kakizaki H, Watanabe T. Differential effects of depot formulations of GnRH agonist leuprorelin and antagonist degarelix on the seminiferous epithelium of the rat testis. Biomed Res 2018; 39:197-214. [PMID: 30101840 DOI: 10.2220/biomedres.39.197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Despite their pharmacologically opposite actions, long-acting depot formulations of both GnRH agonists and antagonists have been clinically applied for treatment of androgen-sensitive prostate cancer. Sustained treatment with GnRH analogues commonly suppresses both the synthesis and release of gonadotropins, leading to depletion of testicular testosterone. To clarify the underlying differences in the effects of GnRH agonists and antagonists on spermatogenesis, we compared histological changes in the seminiferous epithelium after administration of depot formulations of GnRH agonist leuprorelin and antagonist degarelix to male rats. Testicular weight had markedly declined by 28 days after administration of both GnRH analogues, although the testicular weight was decreased more promptly by leuprorelin compared with degarelix. Shortly after administration, massive exfoliation of premature spermatids and anomalous multinucleated giant cells was observed in seminiferous tubules of leuprorelin-treated rats, probably via the initial hyperstimulatory effects on the hypothalamic-pituitary-testicular axis, whereas no discernible changes were found in those of degarelix-treated rats. Long term treatment with both types of GnRH analogues similarly induced a marked reduction in the height of the epithelium and deformation of apical cytoplasm in Sertoli cells, resulting in premature detachment of spermatids from the epithelium. Lipid droplets had accumulated progressively in Sertoli cells, especially in those of degarelix-treated rats. These findings clearly demonstrate the differences in the effects of GnRH agonists and antagonists on the spermatogenic process. This study suggests that an appropriate choice of GnRH analogues is necessary to minimize their adverse effects on spermatogenesis when reproductive functions should be preserved in patients.
Collapse
Affiliation(s)
- Jun-Ichi Hori
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University.,Department of Renal and Urologic Surgery, Asahikawa Medical University
| | - Daisuke Koga
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University
| | - Hidehiro Kakizaki
- Department of Renal and Urologic Surgery, Asahikawa Medical University
| | - Tsuyoshi Watanabe
- Department of Microscopic Anatomy and Cell Biology, Asahikawa Medical University
| |
Collapse
|
7
|
Darbey A, Smith LB. Deliverable transgenics & gene therapy possibilities for the testes. Mol Cell Endocrinol 2018; 468:81-94. [PMID: 29191697 DOI: 10.1016/j.mce.2017.11.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 11/30/2022]
Abstract
Male infertility and hypogonadism are clinically prevalent conditions with a high socioeconomic burden and are both linked to an increased risk in cardiovascular-metabolic diseases and earlier mortality. Therefore, there is an urgent need to better understand the causes and develop new treatments for these conditions that affect millions of men. The accelerating advancement in gene editing and delivery technologies promises improvements in both diagnosis as well as affording the opportunity to develop bespoke treatment options which would both prove beneficial for the millions of individuals afflicted with these reproductive disorders. In this review, we summarise the systems developed and utilised for the delivery of gene therapy and discuss how each of these systems could be applied for the development of a gene therapy system in the testis and how they could be of use for the future diagnosis and repair of common male reproductive disorders.
Collapse
Affiliation(s)
- Annalucia Darbey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
8
|
Johnson KJ. Testicular histopathology associated with disruption of the Sertoli cell cytoskeleton. SPERMATOGENESIS 2015; 4:e979106. [PMID: 26413393 DOI: 10.4161/21565562.2014.979106] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/16/2014] [Indexed: 12/19/2022]
Abstract
Testicular histological alterations following Sertoli cell cytoskeleton disruption are numerous. The Sertoli cell cytoskeleton is comprised of intermediate filaments, microtubules, microfilaments and their direct interacting proteins and performs essential functions including structural support of the seminiferous epithelium, apicobasal movement of elongate spermatids, and release of elongate spermatids from the seminiferous epithelium during spermiation. This review summarizes the histological changes occurring after disruption of the Sertoli cell cytoskeleton, including the signature lesion of seminiferous epithelium sloughing. By presenting examples of histological changes after exposure to toxins or toxicants directly affecting the Sertoli cell cytoskeleton or genetic manipulations of this cytoskeleton, the toxicologist observing similar histological changes associated with exposure to novel compounds can use this information to generate hypotheses about a potential mode of action.
Collapse
Affiliation(s)
- Kamin J Johnson
- Toxicology and Environmental Research and Consulting; The Dow Chemical Company ; Midland, MI USA
| |
Collapse
|
9
|
Tang EI, Mok KW, Lee WM, Cheng CY. EB1 regulates tubulin and actin cytoskeletal networks at the sertoli cell blood-testis barrier in male rats: an in vitro study. Endocrinology 2015; 156:680-93. [PMID: 25456071 PMCID: PMC4298315 DOI: 10.1210/en.2014-1720] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
During spermatogenesis, developing germ cells are transported across the seminiferous epithelium. Studies propose that because microtubules (MTs) serve as the tracks for transporting cell organelles, they may also serve a similar function in the transport of developing germ cells. Polarized MTs may provide the tracks along which polarized actin microfilaments, which act as vehicles to transport cargo, such as preleptotene spermatocytes through the blood-testis barrier (BTB) and spermatids across the epithelium. Yet the molecular mechanism(s) underlying these events remain unknown. Using an established in vitro Sertoli cell system to study BTB function, we demonstrated herein that a MT regulatory protein end-binding protein 1 (EB1) regulates the MT- and also the actin-based cytoskeleton of the Sertoli cell BTB in the rat. EB1 serves as a coordinator between the two cytoskeletons by regulating MT polymerization and actin filament bundling to modulate germ cell transport at the Sertoli cell BTB. A knockdown of EB1 by RNA interference was found to perturb the tight junction (TJ)-permeability barrier, as evidenced by mislocalization of junctional proteins critical for barrier function to facilitate spermatocyte transport, which was likely achieved by two coordinated events. First, EB1 knockdown resulted in changes in MT polymerization, thereby perturbing MT organization in Sertoli cells in which polarized MT no longer stretched properly across the cell cytosol to serve as the tracks. Second, EB1 knockdown perturbed actin organization via its effects on the branched actin polymerization-inducing protein called Arp3 (actin-related protein 3), perturbing microfilament bundling capability based on a biochemical assay, thereby causing microfilament truncation and misorganization, disrupting the function of the vehicle. This reduced actin microfilament bundling capability thus perturbed TJ-protein distribution and localization at the BTB, destabilizing the TJ barrier, leading to its remodeling to facilitate spermatocyte transport. In summary, EB1 provides a functional link between tubulin- and actin-based cytoskeletons to confer spermatocyte transport at the BTB.
Collapse
Affiliation(s)
- Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research (E.I.T., K.-W.M., C.Y.C.), Center for Biomedical Research, Population Council, New York, New York 10065; and School of Biological Sciences (W.M.L.), University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
10
|
Differential gene expression profiling of enriched human spermatogonia after short- and long-term culture. BIOMED RESEARCH INTERNATIONAL 2014; 2014:138350. [PMID: 24738045 PMCID: PMC3971551 DOI: 10.1155/2014/138350] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/19/2013] [Indexed: 01/15/2023]
Abstract
This study aimed to provide a molecular signature for enriched adult human stem/progenitor spermatogonia during short-term (<2 weeks) and long-term culture (up to more than 14 months) in comparison to human testicular fibroblasts and human embryonic stem cells. Human spermatogonia were isolated by CD49f magnetic activated cell sorting and collagen(-)/laminin(+) matrix binding from primary testis cultures obtained from ten adult men. For transcriptomic analysis, single spermatogonia-like cells were collected based on their morphology and dimensions using a micromanipulation system from the enriched germ cell cultures. Immunocytochemical, RT-PCR and microarray analyses revealed that the analyzed populations of cells were distinct at the molecular level. The germ- and pluripotency-associated genes and genes of differentiation/spermatogenesis pathway were highly expressed in enriched short-term cultured spermatogonia. After long-term culture, a proportion of cells retained and aggravated the "spermatogonial" gene expression profile with the expression of germ and pluripotency-associated genes, while in the majority of long-term cultured cells this molecular profile, typical for the differentiation pathway, was reduced and more genes related to the extracellular matrix production and attachment were expressed. The approach we provide here to study the molecular status of in vitro cultured spermatogonia may be important to optimize the culture conditions and to evaluate the germ cell plasticity in the future.
Collapse
|
11
|
O'Donnell L, O'Bryan MK. Microtubules and spermatogenesis. Semin Cell Dev Biol 2014; 30:45-54. [PMID: 24440897 DOI: 10.1016/j.semcdb.2014.01.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/04/2014] [Accepted: 01/08/2014] [Indexed: 12/28/2022]
Abstract
Microtubules are dynamic polymers of tubulin subunits that underpin many essential cellular processes, such as cell division and migration. Spermatogenesis is the process by which spermatogenic stem cells undergo mitotic and meiotic division and differentiation to produce streamlined spermatozoa capable of motility and fertilization. This review summarizes the current knowledge of microtubule-based processes in spermatogenesis. We describe the involvement of microtubule dynamics in Sertoli cell shape and function, as well as in the mitotic and meiotic division of germ cells. The roles of microtubules in sperm head shaping, via the development and function of the manchette, and in sperm flagella development are also discussed. The review brings together data from microscopy studies and genetically modified mouse models, and reveals that the regulation of microtubule dynamics is essential for male fertility.
Collapse
Affiliation(s)
- Liza O'Donnell
- MIMR-PHI Institute of Medical Research, Clayton, Victoria 3168, Australia; Department of Anatomy and Developmental Biology, Monash University, Victoria 3800, Australia.
| | - Moira K O'Bryan
- Department of Anatomy and Developmental Biology, Monash University, Victoria 3800, Australia
| |
Collapse
|
12
|
The Cep63 paralogue Deup1 enables massive de novo centriole biogenesis for vertebrate multiciliogenesis. Nat Cell Biol 2013; 15:1434-44. [PMID: 24240477 DOI: 10.1038/ncb2880] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 10/16/2013] [Indexed: 12/15/2022]
Abstract
Dense multicilia in higher vertebrates are important for luminal flow and the removal of thick mucus. To generate hundreds of basal bodies for multiciliogenesis, specialized terminally differentiated epithelial cells undergo massive centriole amplification. In proliferating cells, however, centriole duplication occurs only once per cell cycle. How cells ensure proper regulation of centriole biogenesis in different contexts is poorly understood. We report that the centriole amplification is controlled by two duplicated genes, Cep63 and Deup1. Cep63 regulates mother-centriole-dependent centriole duplication. Deup1 governs deuterosome assembly to mediate large-scale de novo centriole biogenesis. Similarly to Cep63, Deup1 binds to Cep152 and then recruits Plk4 to activate centriole biogenesis. Phylogenetic analyses suggest that Deup1 diverged from Cep63 in a certain ancestor of lobe-finned fishes during vertebrate evolution and was subsequently adopted by tetrapods. Thus, the Cep63 gene duplication has enabled mother-centriole-independent assembly of the centriole duplication machinery to satisfy different requirements for centriole number.
Collapse
|
13
|
Zeng L, Wan Y, Li D, Wu J, Shao M, Chen J, Hui L, Ji H, Zhu X. The m subunit of murine translation initiation factor eIF3 maintains the integrity of the eIF3 complex and is required for embryonic development, homeostasis, and organ size control. J Biol Chem 2013; 288:30087-30093. [PMID: 24003236 DOI: 10.1074/jbc.m113.506147] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mammalian eIF3 is composed of 13 subunits and is the largest eukaryotic initiation factor. eIF3 plays a key role in protein biosynthesis. However, it is not fully understood how different subunits contribute to the structural integrity and function of the eIF3 complex. Whether eIF3 is essential for embryonic development and homeostasis is also not known. Here, we show that eIF3m null embryos are lethal at the peri-implantation stage. Compound heterozygotes (eIF3m(flox)(/-)) or FABP4-Cre-mediated conditional knock-out mice are lethal at mid-gestation stages. Although the heterozygotes are viable, they show markedly reduced organ size and diminished body weight. Acute ablation of eIF3m in adult mouse liver leads to rapidly decreased body weight and death within 2 weeks; these effects are correlated with a severe decline of protein biogenesis in the liver. Protein analyses reveal that eIF3m deficiency significantly impairs the integrity of the eIF3 complex due to down-regulation of multiple other subunits. Two of the subunits, eIF3f and eIF3h, are stabilized by eIF3m through subcomplex formation. Therefore, eIF3m is required for the structural integrity and translation initiation function of eIF3. Furthermore, not only is eIF3m an essential gene, but its expression level is also important for mouse embryonic development and the control of organ size.
Collapse
Affiliation(s)
- Liyong Zeng
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Yihan Wan
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Dan Li
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Jing Wu
- the Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Mengle Shao
- the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| | - Jiong Chen
- the Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Lijian Hui
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Hongbin Ji
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and
| | - Xueliang Zhu
- From the State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, and.
| |
Collapse
|
14
|
Alves MG, Martins AD, Cavaco JE, Socorro S, Oliveira PF. Diabetes, insulin-mediated glucose metabolism and Sertoli/blood-testis barrier function. Tissue Barriers 2013; 1:e23992. [PMID: 24665384 PMCID: PMC3875609 DOI: 10.4161/tisb.23992] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 02/10/2013] [Accepted: 02/12/2013] [Indexed: 02/06/2023] Open
Abstract
Blood testis barrier (BTB) is one of the tightest blood-barriers controlling the entry of substances into the intratubular fluid. Diabetes Mellitus (DM) is an epidemic metabolic disease concurrent with falling fertility rates, which provokes severe detrimental BTB alterations. It induces testicular alterations, disrupting the metabolic cooperation between the cellular constituents of BTB, with dramatic consequences on sperm quality and fertility. As Sertoli cells are involved in the regulation of spermatogenesis, providing nutritional support for germ cells, any metabolic alteration in these cells derived from DM may be responsible for spermatogenesis disruption, playing a crucial role in fertility/subfertility associated with this pathology. These cells have a glucose sensing machinery that reacts to hormonal fluctuations and several mechanisms to counteract hyper/hypoglycemic events. The role of DM on Sertoli/BTB glucose metabolism dynamics and the metabolic molecular mechanisms through which DM and insulin deregulation alter its functioning, affecting male reproductive potential will be discussed.
Collapse
Affiliation(s)
- Marco G. Alves
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - Ana D. Martins
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - José E. Cavaco
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - Pedro F. Oliveira
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| |
Collapse
|
15
|
Su W, Mruk DD, Cheng CY. Regulation of actin dynamics and protein trafficking during spermatogenesis--insights into a complex process. Crit Rev Biochem Mol Biol 2013; 48:153-72. [PMID: 23339542 DOI: 10.3109/10409238.2012.758084] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the mammalian testis, extensive restructuring takes place across the seminiferous epithelium at the Sertoli-Sertoli and Sertoli-germ cell interface during the epithelial cycle of spermatogenesis, which is important to facilitate changes in the cell shape and morphology of developing germ cells. However, precise communications also take place at the cell junctions to coordinate the discrete events pertinent to spermatogenesis, namely spermatogonial renewal via mitosis, cell cycle progression and meiosis, spermiogenesis and spermiation. It is obvious that these cellular events are intimately related to the underlying actin-based cytoskeleton which is being used by different cell junctions for their attachment. However, little is known on the biology and regulation of this cytoskeleton, in particular its possible involvement in endocytic vesicle-mediated trafficking during spermatogenesis, which in turn affects cell adhesive function and communication at the cell-cell interface. Studies in other epithelia in recent years have shed insightful information on the intimate involvement of actin dynamics and protein trafficking in regulating cell adhesion and communications. The goal of this critical review is to provide an updated assessment of the latest findings in the field on how these complex processes are being regulated during spermatogenesis. We also provide a working model based on the latest findings in the field including our laboratory to provide our thoughts on an apparent complicated subject, which also serves as the framework for investigators in the field. It is obvious that this model will be rapidly updated when more data are available in future years.
Collapse
Affiliation(s)
- Wenhui Su
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY 10065, USA
| | | | | |
Collapse
|
16
|
Goldspink DA, Gadsby JR, Bellett G, Keynton J, Tyrrell BJ, Lund EK, Powell PP, Thomas P, Mogensen MM. The microtubule end-binding protein EB2 is a central regulator of microtubule reorganisation in apico-basal epithelial differentiation. J Cell Sci 2013; 126:4000-14. [DOI: 10.1242/jcs.129759] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microtubule end-binding (EB) proteins influence microtubule dynamic instability, a process essential for microtubule reorganisation during apico-basal epithelial differentiation. Here we establish for the first time that EB2, but not EB1, expression is critical for initial microtubule reorganisation during apico-basal epithelial differentiation, and that EB2 downregulation promotes bundle formation. EB2 siRNA knockdown during early stages of apico-basal differentiation prevented microtubule reorganisation, while its downregulation at later stages promoted microtubule stability and bundle formation. Interestingly, while EB1 is not essential for microtubule reorganisation its knockdown prevented apico-basal bundle formation and epithelial elongation. EB2 siRNA depletion in undifferentiated epithelial cells induced formation of straight, less dynamic microtubules with EB1 and ACF7 lattice association and co-alignment with actin filaments, a phenotype that could be rescued by formin inhibition. Importantly, in situ inner ear and intestinal crypt epithelial tissue revealed direct correlations between low level of EB2 expression and presence of apico-basal microtubule bundles, which were absent where EB2 was elevated. EB2 is evidently important for initial microtubule reorganisation during epithelial polarisation, while its downregulation facilitates EB1/ACF7 microtubule lattice association, microtubule-actin filament co-alignment and bundle formation. The spatiotemporal expression of EB2 thus dramatically influences microtubule organisation, EB1/ACF7 deployment and epithelial differentiation.
Collapse
|
17
|
Huang N, Chelliah Y, Shan Y, Taylor CA, Yoo SH, Partch C, Green CB, Zhang H, Takahashi JS. Crystal structure of the heterodimeric CLOCK:BMAL1 transcriptional activator complex. Science 2012; 337:189-94. [PMID: 22653727 PMCID: PMC3694778 DOI: 10.1126/science.1222804] [Citation(s) in RCA: 227] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The circadian clock in mammals is driven by an autoregulatory transcriptional feedback mechanism that takes approximately 24 hours to complete. A key component of this mechanism is a heterodimeric transcriptional activator consisting of two basic helix-loop-helix PER-ARNT-SIM (bHLH-PAS) domain protein subunits, CLOCK and BMAL1. Here, we report the crystal structure of a complex containing the mouse CLOCK:BMAL1 bHLH-PAS domains at 2.3 Å resolution. The structure reveals an unusual asymmetric heterodimer with the three domains in each of the two subunits--bHLH, PAS-A, and PAS-B--tightly intertwined and involved in dimerization interactions, resulting in three distinct protein interfaces. Mutations that perturb the observed heterodimer interfaces affect the stability and activity of the CLOCK:BMAL1 complex as well as the periodicity of the circadian oscillator. The structure of the CLOCK:BMAL1 complex is a starting point for understanding at an atomic level the mechanism driving the mammalian circadian clock.
Collapse
Affiliation(s)
- Nian Huang
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yogarany Chelliah
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yongli Shan
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Clinton A. Taylor
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Molecular Biophysics Graduate Program, Division of Basic Science, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Seung-Hee Yoo
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carrie Partch
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carla B. Green
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hong Zhang
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph S. Takahashi
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
18
|
Smith LB, Milne L, Nelson N, Eddie S, Brown P, Atanassova N, O'Bryan MK, O'Donnell L, Rhodes D, Wells S, Napper D, Nolan P, Lalanne Z, Cheeseman M, Peters J. KATNAL1 regulation of sertoli cell microtubule dynamics is essential for spermiogenesis and male fertility. PLoS Genet 2012; 8:e1002697. [PMID: 22654668 PMCID: PMC3359976 DOI: 10.1371/journal.pgen.1002697] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/20/2012] [Indexed: 11/18/2022] Open
Abstract
Spermatogenesis is a complex process reliant upon interactions between germ cells (GC) and supporting somatic cells. Testicular Sertoli cells (SC) support GCs during maturation through physical attachment, the provision of nutrients, and protection from immunological attack. This role is facilitated by an active cytoskeleton of parallel microtubule arrays that permit transport of nutrients to GCs, as well as translocation of spermatids through the seminiferous epithelium during maturation. It is well established that chemical perturbation of SC microtubule remodelling leads to premature GC exfoliation demonstrating that microtubule remodelling is an essential component of male fertility, yet the genes responsible for this process remain unknown. Using a random ENU mutagenesis approach, we have identified a novel mouse line displaying male-specific infertility, due to a point mutation in the highly conserved ATPase domain of the novel KATANIN p60-related microtubule severing protein Katanin p60 subunit A-like1 (KATNAL1). We demonstrate that Katnal1 is expressed in testicular Sertoli cells (SC) from 15.5 days post-coitum (dpc) and that, consistent with chemical disruption models, loss of function of KATNAL1 leads to male-specific infertility through disruption of SC microtubule dynamics and premature exfoliation of spermatids from the seminiferous epithelium. The identification of KATNAL1 as an essential regulator of male fertility provides a significant novel entry point into advancing our understanding of how SC microtubule dynamics promotes male fertility. Such information will have resonance both for future treatment of male fertility and the development of non-hormonal male contraceptives.
Collapse
Affiliation(s)
- Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lie PPY, Mruk DD, Lee WM, Cheng CY. Cytoskeletal dynamics and spermatogenesis. Philos Trans R Soc Lond B Biol Sci 2010; 365:1581-92. [PMID: 20403871 DOI: 10.1098/rstb.2009.0261] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Different cellular events occur during spermatogenesis, and these include (i) mitosis for self-renewal of spermatogonia, (ii) differentiation of type A spermatogonia into type B and commitment of type B spermatogonia to develop into preleptotene primary spermatocytes, (iii) transit of preleptotene/leptotene spermatocytes across the blood-testis barrier in coordination with germ cell cycle progression and meiosis, (iv) spermiogenesis and spermiation. These events also associate with extensive changes in cell shape and size, and germ cell movement. The cytoskeleton, which comprises actin, microtubules and intermediate filaments, is believed to function in these cellular events. However, few studies have been conducted by investigators in the past decades to unfold the role of the cytoskeleton during spermatogenesis. This review summarizes recent advances in the field relating to cytoskeletal dynamics in the testis, and highlights areas of research that require additional emphasis so that new approaches for male contraception, as well as therapeutic approaches to alleviate environmental toxicant-induced reproductive dysfunction in men, can possibly be developed.
Collapse
Affiliation(s)
- Pearl P Y Lie
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA
| | | | | | | |
Collapse
|
20
|
Han Y, Huang C, Sun X, Xiang B, Wang M, Yeh ETH, Chen Y, Li H, Shi G, Cang H, Sun Y, Wang J, Wang W, Gao F, Yi J. SENP3-mediated de-conjugation of SUMO2/3 from promyelocytic leukemia is correlated with accelerated cell proliferation under mild oxidative stress. J Biol Chem 2010; 285:12906-15. [PMID: 20181954 DOI: 10.1074/jbc.m109.071431] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Small ubiquitin-like modifier (SUMO) 2/3 is known to conjugate to substrates in response to a variety of cellular stresses. However, whether and how SUMO2/3-specific proteases are involved in de-conjugation under cell stress is unclear. Here, we show that low doses of hydrogen peroxide (H(2)O(2)) induce an increase of the SENP3 protein, which removes SUMO2/3 from promyelocytic leukemia (PML). Low dose H(2)O(2) causes SENP3 to co-localize with PML bodies and reduces the number of PML bodies in a SENP3-dependent manner. Furthermore, de-conjugation of SUMO2/3 from PML is responsible for the accelerated cell proliferation caused by low dose H(2)O(2). Knocking down PML promotes basal cell proliferation as expected. This can be reversed by reconstitution with wild-type PML but not its mutant lacking SUMOylation, indicating that only the SUMOylated PML can play an inhibitory role for cell proliferation. Thus, SENP3 appears to be a key mediator in mild oxidative stress-induced cell proliferation via regulation of the SUMOylation status of PML. Furthermore, SENP3 is over-accumulated in a variety of primary human cancers including colon adenocarcinoma in which PML is hypo-SUMOylated. These results reveal an important role of SENP3 and the SUMOylation status of PML in the regulation of cell proliferation under oxidative stress.
Collapse
Affiliation(s)
- Yan Han
- Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
De Groot CO, Jelesarov I, Damberger FF, Bjelić S, Schärer MA, Bhavesh NS, Grigoriev I, Buey RM, Wüthrich K, Capitani G, Akhmanova A, Steinmetz MO. Molecular insights into mammalian end-binding protein heterodimerization. J Biol Chem 2009; 285:5802-14. [PMID: 20008324 DOI: 10.1074/jbc.m109.068130] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microtubule plus-end tracking proteins (+TIPs) are involved in many microtubule-based processes. End binding (EB) proteins constitute a highly conserved family of +TIPs. They play a pivotal role in regulating microtubule dynamics and in the recruitment of diverse +TIPs to growing microtubule plus ends. Here we used a combination of methods to investigate the dimerization properties of the three human EB proteins EB1, EB2, and EB3. Based on Förster resonance energy transfer, we demonstrate that the C-terminal dimerization domains of EBs (EBc) can readily exchange their chains in solution. We further document that EB1c and EB3c preferentially form heterodimers, whereas EB2c does not participate significantly in the formation of heterotypic complexes. Measurements of the reaction thermodynamics and kinetics, homology modeling, and mutagenesis provide details of the molecular determinants of homo- versus heterodimer formation of EBc domains. Fluorescence spectroscopy and nuclear magnetic resonance studies in the presence of the cytoskeleton-associated protein-glycine-rich domains of either CLIP-170 or p150(glued) or of a fragment derived from the adenomatous polyposis coli tumor suppressor protein show that chain exchange of EBc domains can be controlled by binding partners. Extension of these studies of the EBc domains to full-length EBs demonstrate that heterodimer formation between EB1 and EB3, but not between EB2 and the other two EBs, occurs both in vitro and in cells as revealed by live cell imaging. Together, our data provide molecular insights for rationalizing the dominant negative control by C-terminal EB domains and form a basis for understanding the functional role of heterotypic chain exchange by EBs in cells.
Collapse
Affiliation(s)
- Christian O De Groot
- Biomolecular Research, Structural Biology, the Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhang Q, Wang F, Cao J, Shen Y, Huang Q, Bao L, Zhu X. Nudel promotes axonal lysosome clearance and endo-lysosome formation via dynein-mediated transport. Traffic 2009; 10:1337-49. [PMID: 19522757 DOI: 10.1111/j.1600-0854.2009.00945.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Axonal transport is critical for neuronal function and survival. Cytoplasmic dynein and its accessory complex dynactin form a microtubule minus end-directed motor in charge of retrograde transport. In this study, we show that Nudel, a dynein regulator, was highly expressed in dorsal root ganglion (DRG) neurons. Microinjection of anti-Nudel antibody into cultured DRG neurons abolished retrograde transport of membranous organelles in the axon and led to dispersions of Golgi cisternae in the soma. As a result, lysosomes, which are normally enriched in the soma, moved persistently into and thus accumulated in axons. Endo-lysosome formation was also markedly delayed. As anterograde motility of mitochondria was not inhibited, the antibody apparently did not abolish retrograde transport by destructing axonal microtubule tracks. Similar results were obtained by microinjecting N-terminal Nudel, anti-dynein antibody or a p150(Glued) mutant capable of abrogating the dynein-dynactin association. These results indicate a critical role of Nudel in dynein-mediated axonal transport. Moreover, the effects of dynein on endolysosome formation and regional sequestration of lysosomes may contribute to defects in the endocytic pathway seen in neurons of patients or animals with malfunction of dynein.
Collapse
Affiliation(s)
- Qiangge Zhang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Hooley RP, Paterson M, Brown P, Kerr K, Saunders PTK. Intra-testicular injection of adenoviral constructs results in Sertoli cell-specific gene expression and disruption of the seminiferous epithelium. Reproduction 2008; 137:361-70. [PMID: 18955374 DOI: 10.1530/rep-08-0247] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Spermatogenesis is a complex process that cannot be modelled in vitro. The somatic Sertoli cells (SCs) within the seminiferous tubules perform a key role in supporting maturation of germ cells (GCs). Progress has been made in determining what aspects of SC function are critical to maintenance of fertility by developing rodent models based on the Cre/LoxP system; however, this is time-consuming and is only applicable to mice. The aim of the present study was to establish methods for direct injection of adenoviral vectors containing shRNA constructs into the testis as a way of inducing target-selective knock-down in vivo. We describe here a series of experiments using adenovirus expressing a green fluorescent protein (GFP) transgene. Injection via the efferent ductules resulted in SC-specific expression of GFP; expression levels paralleled the amount of infective viral particles injected. At the highest doses of virus seminiferous tubule architecture were grossly disturbed and immune cell invasion noted. At lower concentrations, the expression of GFP was variable/negligible, the seminiferous tubule lumen was maintained but stage-dependent GC loss and development of numerous basal vacuoles was observed. These resembled intercellular dilations of SC junctional complexes previously described in rats and may be a consequence of disturbances in SC function due to interaction of the viral particles with the coxsackie/adenovirus receptor that is a component of the junctional complexes within the blood testis barrier. In conclusion, intra-testicular injection of adenoviral vectors disturbs SC function in vivo and future work will therefore focus on the use of lentiviral delivery systems.
Collapse
Affiliation(s)
- R P Hooley
- MRC Human Reproductive Sciences Unit, The Queen's Medical Research Institute, Edinburgh, UK
| | | | | | | | | |
Collapse
|