1
|
Tilwani S, Gandhi K, Dalal SN. 14-3-3ε conditional knockout mice exhibit defects in the development of the epidermis. FEBS Lett 2024; 598:3005-3020. [PMID: 39511902 DOI: 10.1002/1873-3468.15051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
The epidermis is a stratified epithelium that functions as the first line of defense against pathogenic invasion and acts as a barrier preventing water loss. In this study, we aimed to decipher the role of 14-3-3ε in the development of the epidermis. We report that loss of 14-3-3ε in the epidermis of juvenile and adult mice reduces cell division in the basal layer and increases the percentage of cells with multiple centrosomes, leading to a reduction in the thickness of the basal and stratified layers. We also demonstrate a decrease in the expression of differentiation markers, although no gross morphological defects in the skin or adverse effects on the survival of the mice were observed. These results suggest that loss of 14-3-3ε in the epidermis may lead to defects in proliferation and differentiation.
Collapse
Affiliation(s)
- Sarika Tilwani
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Karan Gandhi
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
2
|
Yamada M, Luo Y, Seandel M. Viral Transduction of Mammalian Spermatogonial Stem Cells. Methods Mol Biol 2023; 2656:211-225. [PMID: 37249874 DOI: 10.1007/978-1-0716-3139-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Lentiviral vectors have been major tools for genetic manipulation of spermatogonial stem cells (SSCs) in vitro. Adeno-associated viral vectors are promising emerging tools for in vivo SSC transduction that are less invasive, compared to lentivirus, since AAV DNA is not integrated into the host genome and the host genome remains intact. In this chapter, we describe protocols using lentiviral and adeno-associated viral vectors to transduce SSCs in vitro and vivo, respectively.
Collapse
Affiliation(s)
- Makiko Yamada
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Yanyun Luo
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Marco Seandel
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Battulin N, Korablev A, Ryzhkova A, Smirnov A, Kabirova E, Khabarova A, Lagunov T, Serova I, Serov O. The human EF1a promoter does not provide expression of the transgene in mice. Transgenic Res 2022; 31:525-535. [PMID: 35960480 PMCID: PMC9372930 DOI: 10.1007/s11248-022-00319-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/03/2022] [Indexed: 12/01/2022]
Abstract
In this work, we set out to create mice susceptible to the SARS-CoV-2 coronavirus. To ensure the ubiquitous expression of the human ACE2 gene we used the human EF1a promoter. Using pronuclear microinjection of the transgene construct, we obtained six founders with the insertion of the EF1a-hACE2 transgene, from which four independent mouse lines were established. Unfortunately, only one line had low levels of hACE2 expression in some organs. In addition, we did not detect the hACE2 protein in primary lung fibroblasts from any of the transgenic lines. Bisulfite sequencing analysis revealed that the EF1a promoter was hypermethylated in the genomes of transgenic animals. Extensive analysis of published works about transgenic animals indicated that EF1a transgenic constructs are frequently inactive. Thus, our case cautions against using the EF1a promoter to generate transgenic animals, as it is prone to epigenetic silencing.
Collapse
Affiliation(s)
- Nariman Battulin
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090. .,Institute of Genetic Technologies, Novosibirsk State University, Novosibirsk, Russia, 630090.
| | - Alexey Korablev
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Anastasia Ryzhkova
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Alexander Smirnov
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Evelyn Kabirova
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Anna Khabarova
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Timofey Lagunov
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Irina Serova
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| | - Oleg Serov
- Laboratory of Developmental Genetics, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia, 630090
| |
Collapse
|
4
|
Raghavan R, Koyande N, Beher R, Chetlangia N, Ramadwar M, Pawade S, Thorat R, van Hengel J, Sklyarova T, van Roy F, Dalal SN. Plakophilin3 loss leads to increased adenoma formation and rectal prolapse in APC min mice. Biochem Biophys Res Commun 2022; 586:14-19. [PMID: 34823217 DOI: 10.1016/j.bbrc.2021.11.071] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
Plakophilin3 (PKP3) loss leads to tumor progression and metastasis of colon cancer cells. The goal of this report was to determine if PKP3 loss led to increased disease progression in mice. We generated a colonocyte-specific knockout of PKP3 in APCmin mice, which led to increased adenoma formation, the formation of rectal prolapse, and a significant decrease in survival. The observed increase in rectal prolapse formation and decrease in survival correlated with an increase in the expression of Lipocalin2 (LCN2). Increased disease progression was observed even upon treatment with 5-fluorouracil (5FU). These results suggest that an increase in LCN2 expression might lead to therapy resistance and that LCN2 might serve as a potential therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Rahul Raghavan
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Navami Koyande
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Rohit Beher
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Neha Chetlangia
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Mukda Ramadwar
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, 400012, India
| | - Shital Pawade
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Jolanda van Hengel
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium; VIB Center of Inflammation Research, VIB, Ghent, Belgium; Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Tetyana Sklyarova
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium; VIB Center of Inflammation Research, VIB, Ghent, Belgium; Laboratory of Molecular Medical Oncology, Oncology Research Centre, Free University of Brussels, Belgium
| | - Frans van Roy
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium; VIB Center of Inflammation Research, VIB, Ghent, Belgium
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India.
| |
Collapse
|
5
|
Tilwani S, Gandhi K, Narayan S, Ainavarapu SRK, Dalal SN. Disruption of desmosome function leads to increased centrosome clustering in 14-3-3γ-knockout cells with supernumerary centrosomes. FEBS Lett 2021; 595:2675-2690. [PMID: 34626438 DOI: 10.1002/1873-3468.14204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/09/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023]
Abstract
14-3-3 proteins are conserved, dimeric, acidic proteins that regulate multiple cellular pathways. Loss of either 14-3-3ε or 14-3-3γ leads to centrosome amplification. However, we find that while the knockout of 14-3-3ε leads to multipolar mitoses, the knockout of 14-3-3γ results in centrosome clustering and pseudo-bipolar mitoses. 14-3-3γ knockouts demonstrate compromised desmosome function and a decrease in keratin levels, leading to decreased cell stiffness and an increase in centrosome clustering. Restoration of desmosome function increased multipolar mitoses, whereas knockdown of either plakoglobin or keratin 5 led to decreased cell stiffness and increased pseudo-bipolar mitoses. These results suggest that the ability of the desmosome to anchor keratin filaments maintains cell stiffness, thus inhibiting centrosome clustering, and that phenotypes observed upon 14-3-3 loss reflect the dysregulation of multiple pathways.
Collapse
Affiliation(s)
- Sarika Tilwani
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - Karan Gandhi
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Satya Narayan
- Department of Chemical Sciences, TIFR, Mumbai, India
| | | | - Sorab Nariman Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Mumbai, India
| |
Collapse
|
6
|
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
|
7
|
Kim BJ, Kim YH, Oh MG, Kim KJ, Jung SE, Jin JH, Kim SU, Min KS, Ryu BY. Direct modification of spermatogonial stem cells using lentivirus vectors in vivo leads to efficient generation of transgenic rats. Asian J Androl 2020; 21:190-195. [PMID: 30319135 PMCID: PMC6413556 DOI: 10.4103/aja.aja_80_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Spermatogonial stem cells (SSCs) transmit genetic information to the next progeny in males. Thus, SSCs are a potential target for germline modifications to generate transgenic animals. In this study, we report a technique for the generation of transgenic rats by in vivo manipulation of SSCs with a high success rate. SSCs in juvenile rats were transduced in vivo with high titers of lentivirus harboring enhanced green fluorescent protein and mated with wild-type females to create founder rats. These founder rats expressed the transgene and passed on the transgene with an overall success rate of 50.0%. Subsequent generations of progeny from the founder rats both expressed and passed on the transgene. Thus, direct modification of SSCs in juvenile rats is an effective means of generating transgenic rats through the male germline. This technology could be adapted to larger animals, in which existing methods for gene modification are inadequate or inapplicable, resulting in the generation of transgenic animals in a variety of species.
Collapse
Affiliation(s)
- Bang-Jin Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi-do 17546, Korea
| | - Yong-Hee Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi-do 17546, Korea
| | - Myeong-Geun Oh
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi-do 17546, Korea
| | - Ki-Jung Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi-do 17546, Korea
| | - Sang-Eun Jung
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi-do 17546, Korea
| | - Ju-Hee Jin
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi-do 17546, Korea
| | - Sun-Uk Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk-do 28116, Korea.,Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Chungcheongbuk-do 28116, Korea
| | - Kwan-Sik Min
- Animal Biotechnology, Graduate School of Future Convergence Technology, Department of Animal Life Science, Institute of Genetic Engineering, Hankyong National University, Anseong, Gyeonggi-do 17579, Korea
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Anseong, Gyeonggi-do 17546, Korea
| |
Collapse
|
8
|
Custer SK, Astroski JW, Li HX, Androphy EJ. Interaction between alpha-COP and SMN ameliorates disease phenotype in a mouse model of spinal muscular atrophy. Biochem Biophys Res Commun 2019; 514:530-537. [PMID: 31060774 DOI: 10.1016/j.bbrc.2019.04.176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 11/25/2022]
Abstract
We report that expression of the α-COP protein rescues disease phenotype in a severe mouse model of Spinal Muscular Atrophy (SMA). Lentiviral particles expressing α-COP were injected directly into the testes of genetically pure mouse strain of interest resulting in infection of the spermatagonial stem cells. α-COP was stably expressed in brain, skeletal muscle, and spinal cord without altering SMN protein levels. SMA mice transgenic for α-COP live significantly longer than their non-transgenic littermates, and showed increased body mass and normal muscle morphology at postnatal day 15. We previously reported that binding between SMN and α-COP is required for restoration of neurite outgrowth in cells lacking SMN, and we report similar finding here. Lentiviral-mediated transgenic expression of SMN where the dilysine domain in exon 2b was mutated was not able to rescue the SMA phenotype despite robust expression of the mutant SMN protein in brain, muscle and spinal cord. These results demonstrate that α-COP is a validated modifier of SMA disease phenotype in a mammalian, vertebrate model and is a potential target for development of future SMN-independent therapeutic interventions.
Collapse
Affiliation(s)
- Sara K Custer
- Indiana University School of Medicine, Dermatology, Indianapolis, IN, USA.
| | - Jacob W Astroski
- Indiana University School of Medicine, Dermatology, Indianapolis, IN, USA
| | - Hong Xia Li
- Indiana University School of Medicine, Dermatology, Indianapolis, IN, USA
| | - Elliot J Androphy
- Indiana University School of Medicine, Dermatology, Indianapolis, IN, USA
| |
Collapse
|
9
|
Rajasekaran S, Thatte J, Periasamy J, Javali A, Jayaram M, Sen D, Krishnagopal A, Jayandharan GR, Sambasivan R. Infectivity of adeno-associated virus serotypes in mouse testis. BMC Biotechnol 2018; 18:70. [PMID: 30384832 PMCID: PMC6211462 DOI: 10.1186/s12896-018-0479-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/18/2018] [Indexed: 01/01/2023] Open
Abstract
Background Recombinant adeno-associated viruses (AAVs) are emerging as favoured transgene delivery vectors for both research applications and gene therapy. In this context, a thorough investigation of the potential of various AAV serotypes to transduce specific cell types is valuable. Here, we rigorously tested the infectivity of a number of AAV serotypes in murine testis by direct testicular injection. Results We report the tropism of serotypes AAV2, 5, 8, 9 and AAVrh10 in mouse testis. We reveal unique infectivity of AAV2 and AAV9, which preferentially target intertubular testosterone-producing Leydig cells. Remarkably, AAV2 TM, a mutant for capsid designed to increase transduction, displayed a dramatic alteration in tropism; it infiltrated seminiferous tubules unlike wildtype AAV2 and transduced Sertoli cells. However, none of the AAVs tested infected spermatogonial cells. Conclusions In spite of direct testicular injection, none of the tested AAVs appeared to infect sperm progenitors as assayed by reporter expression. This lends support to the current view that AAVs are safe gene-therapy vehicles. However, testing the presence of rAAV genomic DNA in germ cells is necessary to assess the risk of individual serotypes. Electronic supplementary material The online version of this article (10.1186/s12896-018-0479-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Jayashree Thatte
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
| | - Jayaprakash Periasamy
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
| | - Alok Javali
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India.,National Centre for Biological Sciences, TIFR, GKVK Campus, Bellary Road, Bengaluru, 560065, India
| | - Manjunath Jayaram
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
| | - Dwaipayan Sen
- Department of Haematology and Centre for Stem Cell Research, Christian Medical College, Vellore, 632004, India.,Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, 632014, India
| | - Akshaya Krishnagopal
- Department of Haematology and Centre for Stem Cell Research, Christian Medical College, Vellore, 632004, India
| | - Giridhara R Jayandharan
- Department of Haematology and Centre for Stem Cell Research, Christian Medical College, Vellore, 632004, India.,Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Ramkumar Sambasivan
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India.
| |
Collapse
|
10
|
Basu S, Chaudhary N, Shah S, Braggs C, Sawant A, Vaz S, Thorat R, Gupta S, Dalal SN. Plakophilin3 loss leads to an increase in lipocalin2 expression, which is required for tumour formation. Exp Cell Res 2018; 369:251-265. [PMID: 29803740 DOI: 10.1016/j.yexcr.2018.05.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022]
Abstract
An increase in tumour formation and metastasis are observed upon plakophilin3 (PKP3) loss. To identify pathways downstream of PKP3 loss that are required for increased tumour formation, a gene expression analysis was performed, which demonstrated that the expression of lipocalin2 (LCN2) was elevated upon PKP3 loss and this is consistent with expression data from human tumour samples suggesting that PKP3 loss correlates with an increase in LCN2 expression. PKP3 loss leads to an increase in invasion, tumour formation and metastasis and these phenotypes were dependent on the increase in LCN2 expression. The increased LCN2 expression was due to an increase in the activation of p38 MAPK in the HCT116 derived PKP3 knockdown clones as LCN2 expression decreased upon inhibition of p38 MAPK. The phosphorylated active form of p38 MAPK is translocated to the nucleus upon PKP3 loss and is dependent on complex formation between p38 MAPK and PKP3. WT PKP3 inhibits LCN2 reporter activity in PKP3 knockdown cells but a PKP3 mutant that fails to form a complex with p38 MAPK cannot suppress LCN2 promoter activity. Further, LCN2 expression is decreased upon loss of p38β, but not p38α, in the PKP3 knockdown cells. These results suggest that PKP3 loss leads to an increase in the nuclear translocation of p38 MAPK and p38β MAPK is required for the increase in LCN2 expression.
Collapse
Affiliation(s)
- Srikanta Basu
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Nazia Chaudhary
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sanket Shah
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Carol Braggs
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Aakanksha Sawant
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Simone Vaz
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Rahul Thorat
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Sanjay Gupta
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sorab N Dalal
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India.
| |
Collapse
|
11
|
Intratesticular injection followed by electroporation allows gene transfer in caprine spermatogenic cells. Sci Rep 2018; 8:3169. [PMID: 29453369 PMCID: PMC5816633 DOI: 10.1038/s41598-018-21558-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/07/2018] [Indexed: 01/09/2023] Open
Abstract
The production of transgenic livestock is constrained due to the limited success of currently available methods for transgenesis. Testis mediated gene transfer (TMGT) is an emerging method that shows a high success rate in generating transgenic mice. In this study, we report a newly developed protocol for electroporation-aided TMGT to produce a transgenic goat. The injectable volume and concentration of the transgene were first standardized, and then electroporation conditions were optimized in vitro. In vivo experiments were performed by injecting a transgenic construct (pIRES2-EGFP; enhanced green fluorescent protein) into the testicular interstitium followed by electroporation. Immunohistochemistry, quantitative real-time PCR (qPCR) and western blotting analyses confirmed the successful transfer of the transgene into seminiferous tubules and testicular cells. Furthermore, chromosomal integration of the transgene and its expression in sperm were evaluated d60 and d120 post-electroporation. Our protocol neither altered the seminal characteristics nor the fertilization capacity of the sperm cells. In vitro fertilization using transgenic sperm generated fluorescent embryos. Finally, natural mating of a pre-founder buck produced a transgenic baby goat. The present study demonstrates the first successful report of an electroporation-aided TMGT method for gene transfer in goats.
Collapse
|
12
|
Gene delivery to Nile tilapia cells for transgenesis and the role of PI3K-c2α in angiogenesis. Sci Rep 2017; 7:44317. [PMID: 28317860 PMCID: PMC5357942 DOI: 10.1038/srep44317] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 02/06/2017] [Indexed: 01/08/2023] Open
Abstract
Microinjection is commonly performed to achieve fish transgenesis; however, due to difficulties associated with this technique, new strategies are being developed. Here we evaluate the potential of lentiviral particles to genetically modify Nile tilapia cells to achieve transgenesis using three different approaches: spermatogonial stem cell (SSC) genetic modification and transplantation (SC), in vivo transduction of gametes (GT), and fertilised egg transduction (ET). The SC protocol using larvae generates animals with sustained production of modified sperm (80% of animals with 77% maximum sperm fluorescence [MSF]), but is a time-consuming protocol (sexual maturity in Nile tilapia is achieved at 6 months of age). GT is a faster technique, but the modified gamete production is temporary (70% of animals with 52% MSF). ET is an easier way to obtain mosaic transgenic animals compared to microinjection of eggs, but non-site-directed integration in the fish genome can be a problem. In this study, PI3Kc2α gene disruption impaired development during the embryo stage and caused premature death. The manipulator should choose a technique based on the time available for transgenic obtainment and if this generation is required to be continuous or not.
Collapse
|
13
|
Sato M, Ohtsuka M, Watanabe S, Gurumurthy CB. Nucleic acids delivery methods for genome editing in zygotes and embryos: the old, the new, and the old-new. Biol Direct 2016; 11:16. [PMID: 27037013 PMCID: PMC4815204 DOI: 10.1186/s13062-016-0115-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/15/2016] [Indexed: 12/31/2022] Open
Abstract
In the recent years, sequence-specific nucleases such as ZFNs, TALENs, and CRISPR/Cas9 have revolutionzed the fields of animal genome editing and transgenesis. However, these new techniques require microinjection to deliver nucleic acids into embryos to generate gene-modified animals. Microinjection is a delicate procedure that requires sophisticated equipment and highly trained and experienced technicians. Though over a dozen alternate approaches for nucleic acid delivery into embryos were attempted during the pre-CRISPR era, none of them became routinely used as microinjection. The addition of CRISPR/Cas9 to the genome editing toolbox has propelled the search for novel delivery approaches that can obviate the need for microinjection. Indeed, some groups have recently developed electroporation-based methods that have the potential to radically change animal transgenesis. This review provides an overview of the old and new delivery methods, and discusses various strategies that were attempted during the last three decades. In addition, several of the methods are re-evaluated with respect to their suitability to deliver genome editing components, particularly CRISPR/Cas9, to embryos.
Collapse
Affiliation(s)
- Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, Kagoshima, 890-8544 Japan
| | - Masato Ohtsuka
- Division of Basic Molecular Science and Molecular Medicine, School of Medicine, Tokai University, Kanagawa, 259 1193 Japan
| | - Satoshi Watanabe
- Animal Genome Research Unit, Division of Animal Science, National Institute of Agrobiological Sciences, Ibaraki, 305-8602 Japan
| | - Channabasavaiah B. Gurumurthy
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
14
|
Chandrashekran A, Casimir C, Dibb N, Readhead C, Winston R. Generating Transgenic Mice by Lentiviral Transduction of Spermatozoa Followed by In Vitro Fertilization and Embryo Transfer. Methods Mol Biol 2016; 1448:95-106. [PMID: 27317176 DOI: 10.1007/978-1-4939-3753-0_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Most transgenic technologies rely on the oocyte as a substrate for genetic modification. Transgenics animals are usually generated by the injection of the gene constructs (including lentiviruses encoding gene constructs or modified embryonic stem cells) into the pronucleus of a fertilized egg followed by the transfer of the injected embryos into the uterus of a foster mother. Male germ cells also have potential as templates for transgenic development. We have previously shown that mature sperm can be utilized as template for lentiviral transduction and as such used to generate transgenic mice efficiently with germ line capabilities. We provide here a detailed protocol that is relatively simple, to establish transgenic mice using lentivirally transduced spermatozoa. This protocol employs a well-established lentiviral gene delivery system (usual for somatic cells) delivering a variety of transgenes to be directly used with sperm, and the subsequent use of these modified sperm in in vitro fertilization studies and embryo transfer into foster female mice, for the establishment of transgenic mice.
Collapse
Affiliation(s)
- Anil Chandrashekran
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Institute of Reproductive and Developmental Biology (IRDB), Du Cane Road, London, W12 0NN, UK.
| | - Colin Casimir
- Department of Natural Sciences, School of Science & Technology, Middlesex University, The Burroughs, London, NW4 4BT, UK
| | - Nick Dibb
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Institute of Reproductive and Developmental Biology (IRDB), Du Cane Road, London, W12 0NN, UK
| | - Carol Readhead
- Translational Imaging Center, University of Southern California, Los Angeles, CA, 90089, USA
| | - Robert Winston
- Department of Surgery and Cancer, Division of Cancer, Imperial College London, Hammersmith Campus, Institute of Reproductive and Developmental Biology (IRDB), Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
15
|
Qin J, Xu H, Zhang P, Zhang C, Zhu Z, Qu R, Qin Y, Zeng W. An efficient strategy for generation of transgenic mice by lentiviral transduction of male germline stem cells in vivo. J Anim Sci Biotechnol 2015; 6:59. [PMID: 26705472 PMCID: PMC4690335 DOI: 10.1186/s40104-015-0058-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/09/2015] [Indexed: 12/27/2022] Open
Abstract
Background Male germline stem cells (MGSCs) are a subpopulation of germ cells in the testis tissue. MGSCs are capable of differentiation into spermatozoa and thus are perfect targets for genomic manipulation to generate transgenic animals. Method The present study was to optimize a protocol of production of transgenic mice through transduction of MGSCs in vivo using lentiviral-based vectors. The recombinant lentiviral vectors with either EF-1 or CMV promoter to drive the expression of enhanced green fluorescent protein (eGFP) transgene were injected into seminiferous tubules or inter-tubular space of 7-day-old and 28-day-old mouse testes. At 5 or 6 wk post-surgery, these pre-founders were mated with wild-type C57BL/6J female mice (1.5 to 2.0-month-old). Results Sixty-seven percent of F1 generation and 55.56 % of F2 offspring were positive for eGFP transgene under the control of EF-1 promoter via PCR analysis. The transgenic pups were generated in an injection site-and age-independent manner. The expression of transgene was displayed in the progeny derived from lentiviral vector containing CMV promoter to drive transgene, but it was silenced or undetectable in the offspring derived from lentiviral vector with transgene under EF-1 promoter. The methylation level of gDNA in the promoter region of transgene was much higher in the samples derived lentiviral vectors with EF-1 promoter than that with CMV promoter, suggesting eGFP transgene was suppressed by DNA methylation in vivo. Conclusion This research reported here an effective strategy for generation of transgenic mice through transduction of MGSCs in vivo using lentivirus vectors with specific promoters, and the transgenic offspring were obtained in an injection site-and age-independent manner. This protocol could be applied to other animal species, leading to advancement of animal transgenesis in agricultural and biomedical fields. Electronic supplementary material The online version of this article (doi:10.1186/s40104-015-0058-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinzhou Qin
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| | - Haixia Xu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| | - Pengfei Zhang
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| | - Conghui Zhang
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| | - Zhendong Zhu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| | - Rongfeng Qu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| | - Yuwei Qin
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| | - Wenxian Zeng
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100 China
| |
Collapse
|
16
|
MMP7 is required to mediate cell invasion and tumor formation upon Plakophilin3 loss. PLoS One 2015; 10:e0123979. [PMID: 25875355 PMCID: PMC4395386 DOI: 10.1371/journal.pone.0123979] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/24/2015] [Indexed: 11/30/2022] Open
Abstract
Plakophilin3 (PKP3) loss results in increased transformation in multiple cell lines in vitro and increased tumor formation in vivo. A microarray analysis performed in the PKP3 knockdown clones, identified an inflammation associated gene signature in cell lines derived from stratified epithelia as opposed to cell lines derived from simple epithelia. However, in contrast to the inflammation associated gene signature, the expression of MMP7 was increased upon PKP3 knockdown in all the cell lines tested. Using vector driven RNA interference, it was demonstrated that MMP7 was required for in-vitro cell migration and invasion and tumor formation in vivo. The increase in MMP7 levels was due to the increase in levels of the Phosphatase of Regenerating Liver3 (PRL3), which is observed upon PKP3 loss. The results suggest that MMP7 over-expression may be one of the mechanisms by which PKP3 loss leads to increased cell invasion and tumor formation.
Collapse
|
17
|
Sehgal L, Mukhopadhyay A, Rajan A, Khapare N, Sawant M, Vishal SS, Bhatt K, Ambatipudi S, Antao N, Alam H, Gurjar M, Basu S, Mathur R, Borde L, Hosing AS, Vaidya MM, Thorat R, Samaniego F, Kolthur-Seetharam U, Dalal SN. 14-3-3γ-Mediated transport of plakoglobin to the cell border is required for the initiation of desmosome assembly in vitro and in vivo. J Cell Sci 2014; 127:2174-88. [PMID: 24610948 DOI: 10.1242/jcs.125807] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The regulation of cell-cell adhesion is important for the processes of tissue formation and morphogenesis. Here, we report that loss of 14-3-3γ leads to a decrease in cell-cell adhesion and a defect in the transport of plakoglobin and other desmosomal proteins to the cell border in HCT116 cells and cells of the mouse testis. 14-3-3γ binds to plakoglobin in a PKCμ-dependent fashion, resulting in microtubule-dependent transport of plakoglobin to cell borders. Transport of plakoglobin to the border is dependent on the KIF5B-KLC1 complex. Knockdown of KIF5B in HCT116 cells, or in the mouse testis, results in a phenotype similar to that observed upon 14-3-3γ knockdown. Our results suggest that loss of 14-3-3γ leads to decreased desmosome formation and a decrease in cell-cell adhesion in vitro, and in the mouse testis in vivo, leading to defects in testis organization and spermatogenesis.
Collapse
Affiliation(s)
- Lalit Sehgal
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | - Anandi Rajan
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Nileema Khapare
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Mugdha Sawant
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Sonali S Vishal
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Khyati Bhatt
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Srikant Ambatipudi
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Noelle Antao
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Hunain Alam
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Mansa Gurjar
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Srikanta Basu
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Rohit Mathur
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Lalit Borde
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai 400005, India
| | - Amol S Hosing
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Milind M Vaidya
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Rahul Thorat
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| | - Felipe Samaniego
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ullas Kolthur-Seetharam
- Department of Biological Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai 400005, India
| | - Sorab N Dalal
- KS215, ACTREC, Tata Memorial Centre Kharghar Node, Navi Mumbai 410210, India
| |
Collapse
|
18
|
Abstract
The protocols in this chapter describe two techniques for the generation of transgenic mice by in vivo manipulation of spermatogonial stem cells (SSCs) with a high rate of success. SSCs in prepubescent animals can either be infected in vivo with recombinant lentiviruses expressing the transgene of interest or DNA can be injected into the testis followed by the application of an electric current resulting in integration of the linearized DNA containing a transgene downstream of the appropriate promoter into SSCs. All male pre-founder mice produced transgenic pups using both protocols with the transgene being heritable. Further, the pre-founder mice could be used in multiple mating experiments resulting in the generation of multiple progeny. These protocols could be extended to perform over-expression/knockdown screens in vivo using bar-coded lentiviruses/plasmid constructs, thus permitting the design of genetic screens in the mouse. Further, these protocols could be adapted to achieve transgenesis in other laboratory animals resulting in the generation of model systems that closely approximate human development and disease.
Collapse
|
19
|
Chandrashekran A, Sarkar R, Thrasher A, Fraser SE, Dibb N, Casimir C, Winston R, Readhead C. Efficient generation of transgenic mice by lentivirus‐mediated modification of spermatozoa. FASEB J 2013; 28:569-76. [DOI: 10.1096/fj.13-233999] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Anil Chandrashekran
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Rupa Sarkar
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Adrian Thrasher
- Molecular Immunology UnitUniversity College London Institute of Child HealthLondonUK
| | - Scott E. Fraser
- Biological Imaging CenterBeckman InstituteCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Nicholas Dibb
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Colin Casimir
- Department of Natural SciencesSchool of Science and TechnologyMiddlesex UniversityLondonUK
| | - Robert Winston
- Department of Surgery and CancerDivision of CancerInstitute of Reproductive and Developmental Biology (IRDB)Imperial College LondonLondonUK
| | - Carol Readhead
- Biological Imaging CenterBeckman InstituteCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| |
Collapse
|
20
|
Li X, Mao Z, Wu M, Xia J. Rescuing infertility of Pick1 knockout mice by generating testis-specific transgenic mice via testicular infection. Sci Rep 2013; 3:2842. [PMID: 24100262 PMCID: PMC3792414 DOI: 10.1038/srep02842] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 09/02/2013] [Indexed: 12/16/2022] Open
Abstract
PICK1 (protein interacting with C-kinase 1) is a peripheral membrane protein with high expression in brain, testis, pancreas and other neuroendocrine tissues. Male Pick1 knockout mice are completely infertile, with a phenotype resembling the human disease globozoospermia. Since PICK1 is expressed in both testis and neuroendocrine tissues, infertility of Pick1 knockout mice may be due to either impaired neuroendocrine function or abnormal spermatogenesis. To distinguish these two possibilities, we restored PICK1's expression in the testis by seminiferous tubule microinjection of PICK1-containing lentivirus. By examining the testis-specific Pick1 transgenic mice, we found that PICK1's expression in testis rescued the spermatogenic abnormalities and male infertility in Pick1 knockout mice. Our results indicate that the infertility is caused by the lack of PICK1 in the testis rather than in other organs. In addition, we found that seminiferous tubule microinjection of lentivirus has a strong preference to produce testis-specific transgenic mice.
Collapse
Affiliation(s)
- Xiumao Li
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zhuo Mao
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Min Wu
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jun Xia
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|