1
|
Piao X, Li D, Liu H, Guo Q, Yu Y. Advances in gene and cellular therapeutic approaches for Huntington's disease. Protein Cell 2025; 16:307-337. [PMID: 39121016 PMCID: PMC12120246 DOI: 10.1093/procel/pwae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/24/2024] [Indexed: 08/11/2024] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by the abnormal expansion of CAG trinucleotide repeats in the Huntingtin gene (HTT) located on chromosome 4. It is transmitted in an autosomal dominant manner and is characterized by motor dysfunction, cognitive decline, and emotional disturbances. To date, there are no curative treatments for HD have been developed; current therapeutic approaches focus on symptom relief and comprehensive care through coordinated pharmacological and nonpharmacological methods to manage the diverse phenotypes of the disease. International clinical guidelines for the treatment of HD are continually being revised in an effort to enhance care within a multidisciplinary framework. Additionally, innovative gene and cell therapy strategies are being actively researched and developed to address the complexities of the disorder and improve treatment outcomes. This review endeavours to elucidate the current and emerging gene and cell therapy strategies for HD, offering a detailed insight into the complexities of the disorder and looking forward to future treatment paradigms. Considering the complexity of the underlying mechanisms driving HD, a synergistic treatment strategy that integrates various factors-such as distinct cell types, epigenetic patterns, genetic components, and methods to improve the cerebral microenvironment-may significantly enhance therapeutic outcomes. In the future, we eagerly anticipate ongoing innovations in interdisciplinary research that will bring profound advancements and refinements in the treatment of HD.
Collapse
Affiliation(s)
- Xuejiao Piao
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Dan Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Hui Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Qing Guo
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
2
|
González-Fernández M, Perry C, Gerhards NM, Francica P, Rottenberg S. Docetaxel response in BRCA1,p53-deficient mammary tumor cells is affected by Huntingtin and BAP1. Proc Natl Acad Sci U S A 2024; 121:e2402849121. [PMID: 39705313 DOI: 10.1073/pnas.2402849121] [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: 02/14/2024] [Accepted: 10/30/2024] [Indexed: 12/22/2024] Open
Abstract
Taxanes are frequently used anticancer drugs known to kill tumor cells by inducing mitotic aberrations and segregation defects. A defining feature of specific cancers, notably triple-negative breast cancer (TNBC) and particularly those deficient in BRCA1, is chromosomal instability (CIN). Here, we focused on understanding the mechanisms of docetaxel-induced cytotoxicity, especially in the context of BRCA1-deficient TNBC. Using functional genetic screens in CIN+ cells, we identified genes that mediate docetaxel response and found an interaction between Huntingtin (HTT) and BRCA1-associated protein-1 (BAP1). We employed Brca1-/-;p53-/- mammary tumor cells, derived from genetically engineered mouse tumors that closely mimic the human disease, to investigate the role of these genes in CIN+ BRCA1-deficient cells. Specifically, we observed that loss of HTT sensitizes CIN+ BRCA1-deficient mammary tumor cells to docetaxel by shortening mitotic spindle poles and increasing spindle multipolarity. In contrast, BAP1 depletion protected cells against these spindle aberrations by restoring spindle length and enhancing mitotic clustering of the extra centrosomes. In conclusion, our findings shed light on the roles of HTT and BAP1 in controlling mitotic spindle multipolarity and centrosome clustering, specifically in the absence of BRCA1. This affects the response to microtubule-targeting agents and suggests that further studies of the interaction of these genes with the mitotic spindle may provide useful insights into how to target CIN+ cells, particularly in the challenging therapeutic landscape of BRCA1-deficient TNBC.
Collapse
Affiliation(s)
- Martín González-Fernández
- Department of Infectious Diseases and Pathobiology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, Department of Biomedical Research, University of Bern, 3012 Bern, Switzerland
| | - Carmen Perry
- Department of Infectious Diseases and Pathobiology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, Department of Biomedical Research, University of Bern, 3012 Bern, Switzerland
| | - Nora Merete Gerhards
- Department of Infectious Diseases and Pathobiology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
| | - Paola Francica
- Department of Infectious Diseases and Pathobiology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, Department of Biomedical Research, University of Bern, 3012 Bern, Switzerland
| | - Sven Rottenberg
- Department of Infectious Diseases and Pathobiology, Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012 Bern, Switzerland
- Bern Center for Precision Medicine, Department of Biomedical Research, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
3
|
Zeng P, Shu LZ, Zhou YH, Huang HL, Wei SH, Liu WJ, Deng H. Stem Cell Division and Its Critical Role in Mammary Gland Development and Tumorigenesis: Current Progress and Remaining Challenges. Stem Cells Dev 2024; 33:449-467. [PMID: 38943275 DOI: 10.1089/scd.2024.0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024] Open
Abstract
The origin of breast cancer (BC) has traditionally been a focus of medical research. It is widely acknowledged that BC originates from immortal mammary stem cells and that these stem cells participate in two division modes: symmetric cell division (SCD) and asymmetrical cell division (ACD). Although both of these modes are key to the process of breast development and their imbalance is closely associated with the onset of BC, the molecular mechanisms underlying these phenomena deserve in-depth exploration. In this review, we first outline the molecular mechanisms governing ACD/SCD and analyze the role of ACD/SCD in various stages of breast development. We describe that the changes in telomerase activity, the role of polar proteins, and the stimulation of ovarian hormones subsequently lead to two distinct consequences: breast development or carcinogenesis. Finally, gene mutations, abnormalities in polar proteins, modulation of signal-transduction pathways, and alterations in the microenvironment disrupt the balance of BC stem cell division modes and cause BC. Important regulatory factors such as mammalian Inscuteable mInsc, Numb, Eya1, PKCα, PKCθ, p53, and IL-6 also play significant roles in regulating pathways of ACD/SCD and may constitute key targets for future research on stem cell division, breast development, and tumor therapy.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Animals
- Mammary Glands, Human/growth & development
- Mammary Glands, Human/pathology
- Mammary Glands, Human/cytology
- Mammary Glands, Human/metabolism
- Carcinogenesis/pathology
- Carcinogenesis/metabolism
- Carcinogenesis/genetics
- Stem Cells/metabolism
- Stem Cells/cytology
- Cell Division
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/pathology
- Mammary Glands, Animal/metabolism
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Peng Zeng
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Lin-Zhen Shu
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yu-Hong Zhou
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Hai-Lin Huang
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Shu-Hua Wei
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Wen-Jian Liu
- Department of Breast Surgery, Jiangxi Armed Police Corps Hospital, Nanchang, China
| | - Huan Deng
- Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Tumor Immunology Institute, Nanchang University, Nanchang, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Tong H, Yang T, Xu S, Li X, Liu L, Zhou G, Yang S, Yin S, Li XJ, Li S. Huntington's Disease: Complex Pathogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:3845. [PMID: 38612657 PMCID: PMC11011923 DOI: 10.3390/ijms25073845] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Huntington's disease (HD) arises from the abnormal expansion of CAG repeats in the huntingtin gene (HTT), resulting in the production of the mutant huntingtin protein (mHTT) with a polyglutamine stretch in its N-terminus. The pathogenic mechanisms underlying HD are complex and not yet fully elucidated. However, mHTT forms aggregates and accumulates abnormally in neuronal nuclei and processes, leading to disruptions in multiple cellular functions. Although there is currently no effective curative treatment for HD, significant progress has been made in developing various therapeutic strategies to treat HD. In addition to drugs targeting the neuronal toxicity of mHTT, gene therapy approaches that aim to reduce the expression of the mutant HTT gene hold great promise for effective HD therapy. This review provides an overview of current HD treatments, discusses different therapeutic strategies, and aims to facilitate future therapeutic advancements in the field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (H.T.); (T.Y.); (S.X.); (X.L.); (L.L.); (G.Z.); (S.Y.); (S.Y.)
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (H.T.); (T.Y.); (S.X.); (X.L.); (L.L.); (G.Z.); (S.Y.); (S.Y.)
| |
Collapse
|
5
|
Khoshnan A. Gut Microbiota as a Modifier of Huntington's Disease Pathogenesis. J Huntingtons Dis 2024; 13:133-147. [PMID: 38728199 PMCID: PMC11307070 DOI: 10.3233/jhd-240012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2024] [Indexed: 05/12/2024]
Abstract
Huntingtin (HTT) protein is expressed in most cell lineages, and the toxicity of mutant HTT in multiple organs may contribute to the neurological and psychiatric symptoms observed in Huntington's disease (HD). The proteostasis and neurotoxicity of mutant HTT are influenced by the intracellular milieu and responses to environmental signals. Recent research has highlighted a prominent role of gut microbiota in brain and immune system development, aging, and the progression of neurological disorders. Several studies suggest that mutant HTT might disrupt the homeostasis of gut microbiota (known as dysbiosis) and impact the pathogenesis of HD. Dysbiosis has been observed in HD patients, and in animal models of the disease it coincides with mutant HTT aggregation, abnormal behaviors, and reduced lifespan. This review article aims to highlight the potential toxicity of mutant HTT in organs and pathways within the microbiota-gut-immune-central nervous system (CNS) axis. Understanding the functions of Wild-Type (WT) HTT and the toxicity of mutant HTT in these organs and the associated networks may elucidate novel pathogenic pathways, identify biomarkers and peripheral therapeutic targets for HD.
Collapse
Affiliation(s)
- Ali Khoshnan
- Keck School of Medicine, Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
6
|
Sahito JZA, Deng S, Qin L, Xiao L, Zhang D, Huang B. CeRNA Network Reveals the Circular RNA Characterization in Goat Ear Fibroblasts Reprogramming into Mammary Epithelial Cells. Genes (Basel) 2023; 14:1831. [PMID: 37895180 PMCID: PMC10606430 DOI: 10.3390/genes14101831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 10/29/2023] Open
Abstract
Circular RNAs (circRNAs) are a type of non-coding RNA that play a crucial role in the development and lactation of mammary glands in mammals. A total of 107 differentially expressed circRNAs (DE circRNAs) were found, of which 52 were up-regulated and 55 were down-regulated. We also found that DE circRNA host genes were mainly involved in GO terms related to the development process of mammary epithelial cells and KEGG pathways were mostly related to mammary epithelial cells, lactation, and gland development. Protein network analysis found that DE circRNAs can competitively bind to miRNAs as key circRNAs by constructing a circRNA-miRNA-mRNA network. CircRNAs competitively bind to miRNAs (miR-10b-3p, miR-671-5p, chi-miR-200c, chi-miR-378-3p, and chi-miR-30e-5p) involved in goat mammary gland development, mammary epithelial cells, and lactation, affecting the expression of core genes (CDH2, MAPK1, ITGB1, CAMSAP2, and MAPKAPK5). Here, we generated CiMECs and systematically explored the differences in the transcription profile for the first time using whole-transcriptome sequencing. We also analyzed the interaction among mRNA, miRNA, and cirRNA and predicted that circRNA plays an important role in the maintenance of mammary epithelial cells.
Collapse
Affiliation(s)
- Jam Zaheer Ahmed Sahito
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.Z.A.S.); (S.D.); (L.Q.); (L.X.); (D.Z.)
| | - Shan Deng
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.Z.A.S.); (S.D.); (L.Q.); (L.X.); (D.Z.)
| | - Liangshan Qin
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.Z.A.S.); (S.D.); (L.Q.); (L.X.); (D.Z.)
| | - Lianggui Xiao
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.Z.A.S.); (S.D.); (L.Q.); (L.X.); (D.Z.)
| | - Dandan Zhang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.Z.A.S.); (S.D.); (L.Q.); (L.X.); (D.Z.)
- Guangxi Key Laboratory of Eye Health, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Ben Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (J.Z.A.S.); (S.D.); (L.Q.); (L.X.); (D.Z.)
- Guangxi Key Laboratory of Eye Health, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| |
Collapse
|
7
|
Fankhaenel M, Hashemi FSG, Mourao L, Lucas E, Hosawi MM, Skipp P, Morin X, Scheele CLGJ, Elias S. Annexin A1 is a polarity cue that directs mitotic spindle orientation during mammalian epithelial morphogenesis. Nat Commun 2023; 14:151. [PMID: 36631478 PMCID: PMC9834401 DOI: 10.1038/s41467-023-35881-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Oriented cell divisions are critical for the formation and maintenance of structured epithelia. Proper mitotic spindle orientation relies on polarised anchoring of force generators to the cell cortex by the evolutionarily conserved protein complex formed by the Gαi subunit of heterotrimeric G proteins, the Leucine-Glycine-Asparagine repeat protein (LGN) and the nuclear mitotic apparatus protein. However, the polarity cues that control cortical patterning of this ternary complex remain largely unknown in mammalian epithelia. Here we identify the membrane-associated protein Annexin A1 (ANXA1) as an interactor of LGN in mammary epithelial cells. Annexin A1 acts independently of Gαi to instruct the accumulation of LGN and nuclear mitotic apparatus protein at the lateral cortex to ensure cortical anchoring of Dynein-Dynactin and astral microtubules and thereby planar alignment of the mitotic spindle. Loss of Annexin A1 randomises mitotic spindle orientation, which in turn disrupts epithelial architecture and luminogenesis in three-dimensional cultures of primary mammary epithelial cells. Our findings establish Annexin A1 as an upstream cortical cue that regulates LGN to direct planar cell divisions during mammalian epithelial morphogenesis.
Collapse
Affiliation(s)
- Maria Fankhaenel
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Farahnaz S Golestan Hashemi
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Larissa Mourao
- VIB-KULeuven Center for Cancer Biology, Herestraat 49, 3000, Leuven, Belgium
| | - Emily Lucas
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Manal M Hosawi
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| | - Paul Skipp
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.,Centre for Proteomic Research, University of Southampton, Southampton, SO17 1BJ, UK
| | - Xavier Morin
- Ecole Normale Supérieure, CNRS, Inserm, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), PSL Research University, Paris, France
| | | | - Salah Elias
- School of Biological Sciences, University of Southampton, Southampton, SO17 1BJ, UK. .,Insitute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
8
|
Zhao X, Sun Y, Wang Z, Chen L, Li S, Li XJ. Huntingtin exon 1 deletion does not alter the subcellular distribution of huntingtin and gene transcription in mice. Front Cell Neurosci 2022; 16:1021592. [PMID: 36439204 PMCID: PMC9684630 DOI: 10.3389/fncel.2022.1021592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/25/2022] [Indexed: 04/29/2025] Open
Abstract
Huntington disease (HD) is caused by the expansion of CAG triplet repeats in exon 1 of the huntingtin (HTT) gene, which also encodes the first 17 amino acids (N-17) that can modulate the toxicity of the expanded polyQ repeat. N-17 are conserved in a wide range of species and are found to influence the subcellular distribution of mutant Htt. Moreover, N-17 is subject to many posttranslational modifications that may regulate the function, stability, and distribution of HTT. However, the function of Htt exon 1 and its influence on the normal Htt remains to be fully investigated. By investigating a knock-in mouse model that lacks Htt exon1, we found that deletion of Htt exon1 does not affect the survival of mice and differentiation of cultured mouse neurons. Furthermore, the lack of Htt exon 1 does not alter the subcellular distribution of Htt, autophagy protein expression, and global gene transcription in the mouse brain. These results suggest that removing the entire exon 1 of Htt could be a therapeutic approach to eliminate expanded polyQ toxicity.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| |
Collapse
|
9
|
He Z, Ghorayeb R, Tan S, Chen K, Lorentzian AC, Bottyan J, Aalam SMM, Pujana MA, Lange PF, Kannan N, Eaves CJ, Maxwell CA. Pathogenic BRCA1 variants disrupt PLK1-regulation of mitotic spindle orientation. Nat Commun 2022; 13:2200. [PMID: 35459234 PMCID: PMC9033786 DOI: 10.1038/s41467-022-29885-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 04/04/2022] [Indexed: 11/09/2022] Open
Abstract
Preneoplastic mammary tissues from human female BRCA1 mutation carriers, or Brca1-mutant mice, display unexplained abnormalities in luminal differentiation. We now study the division characteristics of human mammary cells purified from female BRCA1 mutation carriers or non-carrier donors. We show primary BRCA1 mutant/+ cells exhibit defective BRCA1 localization, high radiosensitivity and an accelerated entry into cell division, but fail to orient their cell division axis. We also analyse 15 genetically-edited BRCA1 mutant/+ human mammary cell-lines and find that cells carrying pathogenic BRCA1 mutations acquire an analogous defect in their division axis accompanied by deficient expression of features of mature luminal cells. Importantly, these alterations are independent of accumulated DNA damage, and specifically dependent on elevated PLK1 activity induced by reduced BRCA1 function. This essential PLK1-mediated role of BRCA1 in controlling the cell division axis provides insight into the phenotypes expressed during BRCA1 tumorigenesis.
Collapse
Affiliation(s)
- Zhengcheng He
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ryan Ghorayeb
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Susanna Tan
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ke Chen
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amanda C Lorentzian
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jack Bottyan
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Syed Mohammed Musheer Aalam
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Miguel Angel Pujana
- ProCURE, Catalan Institute of Oncology, Oncobell, Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Catalonia, Spain
| | - Philipp F Lange
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Cuccione Childhood Cancer Research Program, British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Nagarajan Kannan
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Cancer Center, Mayo Clinic, Rochester, MN, USA
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Research Institute, Vancouver, British Columbia, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher A Maxwell
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
- Michael Cuccione Childhood Cancer Research Program, British Columbia Children's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
10
|
Sawant N, Morton H, Kshirsagar S, Reddy AP, Reddy PH. Mitochondrial Abnormalities and Synaptic Damage in Huntington's Disease: a Focus on Defective Mitophagy and Mitochondria-Targeted Therapeutics. Mol Neurobiol 2021; 58:6350-6377. [PMID: 34519969 DOI: 10.1007/s12035-021-02556-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022]
Abstract
Huntington's disease (HD) is a fatal and pure genetic disease with a progressive loss of medium spiny neurons (MSN). HD is caused by expanded polyglutamine repeats in the exon 1 of HD gene. Clinically, HD is characterized by chorea, seizures, involuntary movements, dystonia, cognitive decline, intellectual impairment, and emotional disturbances. Several years of intense research revealed that multiple cellular changes, including defective axonal transport, protein-protein interactions, defective bioenergetics, calcium dyshomeostasis, NMDAR activation, synaptic damage, mitochondrial abnormalities, and selective loss of medium spiny neurons are implicated in HD. Recent research on mutant huntingtin (mHtt) and mitochondria has found that mHtt interacts with the mitochondrial division protein, dynamin-related protein 1 (DRP1), enhances GTPase DRP1 enzymatic activity, and causes excessive mitochondrial fragmentation and abnormal distribution, leading to defective axonal transport of mitochondria and selective synaptic degeneration. Recent research also revealed that failure to remove dead and/or dying mitochondria is an early event in the disease progression. Currently, efforts are being made to reduce abnormal protein interactions and enhance synaptic mitophagy as therapeutic strategies for HD. The purpose of this article is to discuss recent research in HD progression. This article also discusses recent developments of cell and mouse models, cellular changes, mitochondrial abnormalities, DNA damage, bioenergetics, oxidative stress, mitophagy, and therapeutics strategies in HD.
Collapse
Affiliation(s)
- Neha Sawant
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hallie Morton
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neurology, Department of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Internal Medicine, Cell Biology & Biochemistry, Public Health and School of Health Professions, Texas Tech University Health Sciences Center, Neuroscience & Pharmacology3601 4th Street, NeurologyLubbock, TX, 79430, USA.
| |
Collapse
|
11
|
Kumar V, Singh C, Singh A. Zebrafish an experimental model of Huntington's disease: molecular aspects, therapeutic targets and current challenges. Mol Biol Rep 2021; 48:8181-8194. [PMID: 34665402 DOI: 10.1007/s11033-021-06787-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022]
Abstract
Huntington disease (HD) is a lethal autosomal dominant neurodegenerative disease whose exact causative mechanism is still unknown. It can transform from one generation to another generation. The CAG triplet expansion on polyglutamine (PolyQ) tract on Huntingtin protein primarily contributes in HD pathogenesis. Apart from this some another molecular mechanisms are also involved in HD pathology such as loss of Brain derived neurotrophic factor in medium spiny neurons, mitochondrial dysfunction, and alterations in synaptic plasticity are briefly discussed in this review. However, several chemicals (3-nitropropionic acid, and Quinolinic acid) and genetic (mHTT-ΔN17-97Q over expression) experimental models are used to explore the exact pathogenic mechanism and finding of new drug targets for the development of novel therapeutic approaches. The zebrafish (Danio rerio) is widely used in in-vivo screening of several central nervous system (CNS) diseases such as HD, Alzheimer's disease (AD), Parkinson's disease (PD), and in memory deficits. Thus, this makes zebrafish as an excellent animal model for the development of new therapeutic strategies against various CNS disorders. We had reviewed several publications utilizing zebrafish and rodents to explore the disease pathology. Studies suggested that zebrafish genes and their human homologues have conserved functions. Zebrafish advantages and their characteristics over the other experimental animals make it an excellent tool for the disease study. This review explains the possible pathogenic mechanism of HD and also discusses about possible treatment therapies, apart from this we also discussed about possible potential therapeutic targets which will helps in designing of novel therapeutic approaches to overcome the disease progression. Diagrammatic depiction shows prevention of HD pathogenesis through attenuation of various biochemical alterations.
Collapse
Affiliation(s)
- Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Charan Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
12
|
Lechler T, Mapelli M. Spindle positioning and its impact on vertebrate tissue architecture and cell fate. Nat Rev Mol Cell Biol 2021; 22:691-708. [PMID: 34158639 PMCID: PMC10544824 DOI: 10.1038/s41580-021-00384-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2021] [Indexed: 12/18/2022]
Abstract
In multicellular systems, oriented cell divisions are essential for morphogenesis and homeostasis as they determine the position of daughter cells within the tissue and also, in many cases, their fate. Early studies in invertebrates led to the identification of conserved core mechanisms of mitotic spindle positioning centred on the Gαi-LGN-NuMA-dynein complex. In recent years, much has been learnt about the way this complex functions in vertebrate cells. In particular, studies addressed how the Gαi-LGN-NuMA-dynein complex dynamically crosstalks with astral microtubules and the actin cytoskeleton, and how it is regulated to orient the spindle according to cellular and tissue-wide cues. We have also begun to understand how dynein motors and actin regulators interact with mechanosensitive adhesion molecules sensing extracellular mechanical stimuli, such as cadherins and integrins, and with signalling pathways so as to respond to extracellular cues instructing the orientation of the division axis in vivo. In this Review, with the focus on epithelial tissues, we discuss the molecular mechanisms of mitotic spindle orientation in vertebrate cells, and how this machinery is regulated by epithelial cues and extracellular signals to maintain tissue cohesiveness during mitosis. We also outline recent knowledge of how spindle orientation impacts tissue architecture in epithelia and its emerging links to the regulation of cell fate decisions. Finally, we describe how defective spindle orientation can be corrected or its effects eliminated in tissues under physiological conditions, and the pathological implications associated with spindle misorientation.
Collapse
Affiliation(s)
- Terry Lechler
- Department of Dermatology, Duke University Medical Center, Durham, NC, USA.
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
13
|
Galgoczi S, Ruzo A, Markopoulos C, Yoney A, Phan-Everson T, Li S, Haremaki T, Metzger JJ, Etoc F, Brivanlou AH. Huntingtin CAG expansion impairs germ layer patterning in synthetic human 2D gastruloids through polarity defects. Development 2021; 148:272380. [PMID: 34608934 PMCID: PMC8513611 DOI: 10.1242/dev.199513] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/20/2021] [Indexed: 11/20/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of the CAG repeats in the huntingtin gene (HTT). Although HD has been shown to have a developmental component, how early during human embryogenesis the HTT-CAG expansion can cause embryonic defects remains unknown. Here, we demonstrate a specific and highly reproducible CAG length-dependent phenotypic signature in a synthetic model for human gastrulation derived from human embryonic stem cells (hESCs). Specifically, we observed a reduction in the extension of the ectodermal compartment that is associated with enhanced activin signaling. Surprisingly, rather than a cell-autonomous effect, tracking the dynamics of TGFβ signaling demonstrated that HTT-CAG expansion perturbs the spatial restriction of activin response. This is due to defects in the apicobasal polarization in the context of the polarized epithelium of the 2D gastruloid, leading to ectopic subcellular localization of TGFβ receptors. This work refines the earliest developmental window for the prodromal phase of HD to the first 2 weeks of human development, as modeled by our 2D gastruloids. Summary: 2D gastruloids of isogenic human embryonic stem cells modeling Huntington's Disease reveal that huntingtin CAG expansion perturbs the spatial restriction of the activin response in the context of the polarized epithelium.
Collapse
Affiliation(s)
- Szilvia Galgoczi
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Albert Ruzo
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Christian Markopoulos
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Yoney
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Tien Phan-Everson
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Shu Li
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Tomomi Haremaki
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Jakob J Metzger
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Fred Etoc
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Laboratory of condensed matter physics, The Rockefeller University, New York, NY 10065, USA
| | - Ali H Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
14
|
Fields E, Vaughan E, Tripu D, Lim I, Shrout K, Conway J, Salib N, Lee Y, Dhamsania A, Jacobsen M, Woo A, Xue H, Cao K. Gene targeting techniques for Huntington's disease. Ageing Res Rev 2021; 70:101385. [PMID: 34098113 PMCID: PMC8373677 DOI: 10.1016/j.arr.2021.101385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 02/08/2023]
Abstract
Huntington's disease (HD) is an autosomal neurodegenerative disorder caused by extended trinucleotide CAG repetition in the HTT gene. Wild-type huntingtin protein (HTT) is essential, involved in a variety of crucial cellular functions such as vesicle transportation, cell division, transcription regulation, autophagy, and tissue maintenance. The mutant HTT (mHTT) proteins in the body interfere with HTT's normal cellular functions and cause additional detrimental effects. In this review, we discuss multiple approaches targeting DNA and RNA to reduce mHTT expression. These approaches are categorized into non-allele-specific silencing and allele-specific-silencing using Single Nucleotide Polymorphisms (SNPs) and haplogroup analysis. Additionally, this review discusses a potential application of recent CRISPR prime editing technology in targeting HD.
Collapse
Affiliation(s)
- Eric Fields
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Erik Vaughan
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Deepika Tripu
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Isabelle Lim
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Katherine Shrout
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Jessica Conway
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Nicole Salib
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Yubin Lee
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Akash Dhamsania
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Michael Jacobsen
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Ashley Woo
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States
| | - Huijing Xue
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, United States
| | - Kan Cao
- Gemstone Honors Program, University of Maryland, College Park, MD 20742, United States; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
15
|
Edamakanti CR, Opal P. Developmental Alterations in Adult-Onset Neurodegenerative Disorders: Lessons from Polyglutamine Diseases. Mov Disord 2021; 36:1548-1552. [PMID: 34014004 DOI: 10.1002/mds.28657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 01/22/2023] Open
Affiliation(s)
| | - Puneet Opal
- Davee Department of Neurology, Northwestern University Feinberg School of Medicine Chicago, Chicago, IL, USA
| |
Collapse
|
16
|
Katow H, Katow T, Yoshida H, Kiyomoto M. Involvement of Huntingtin in Development and Ciliary Beating Regulation of Larvae of the Sea Urchin, Hemicentrotus pulcherrimus. Int J Mol Sci 2021; 22:5116. [PMID: 34066037 PMCID: PMC8151597 DOI: 10.3390/ijms22105116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022] Open
Abstract
The multiple functions of the wild type Huntington's disease protein of the sea urchin Hemicentrotus pulcherrimus (Hp-Htt) have been examined using the anti-Hp-Htt antibody (Ab) raised against synthetic oligopeptides. According to immunoblotting, Hp-Htt was detected as a single band at around the 350 kDa region at the swimming blastula stage to the prism larva stage. From the 2-arm pluteus stage (2aPL), however, an additional smaller band at the 165 kDa region appeared. Immunohistochemically, Hp-Htt was detected in the nuclei and the nearby cytoplasm of the ectodermal cells from the swimming blastula stage, and the blastocoelar cells from the mid-gastrula stage. The Ab-positive signal was converged to the ciliary band-associated strand (CBAS). There, it was accompanied by several CBAS-marker proteins in the cytoplasm, such as glutamate decarboxylase. Application of Hp-Htt morpholino (Hp-Htt-MO) has resulted in shortened larval arms, accompanied by decreased 5-bromo-2-deoxyuridin (BrdU) incorporation by the ectodermal cells of the larval arms. Hp-Htt-MO also resulted in lowered ciliary beating activity, accompanied by a disordered swirling pattern formation around the body. These Hp-Htt-MO-induced deficiencies took place after the onset of CBAS system formation at the larval arms. Thus, Hp-Htt is involved in cell proliferation and the ciliary beating pattern regulation signaling system in pluteus larvae.
Collapse
Affiliation(s)
- Hideki Katow
- Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan; (H.Y.); (M.K.)
- Research Center for Marine Biology, Tohoku University, Aomori 039-3501, Japan;
| | - Tomoko Katow
- Research Center for Marine Biology, Tohoku University, Aomori 039-3501, Japan;
| | - Hiromi Yoshida
- Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan; (H.Y.); (M.K.)
| | - Masato Kiyomoto
- Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan; (H.Y.); (M.K.)
- Marine and Coastal Research Center, Ochanomizu University, Chiba 294-0301, Japan
| |
Collapse
|
17
|
Proteotoxicity and Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21165646. [PMID: 32781742 PMCID: PMC7460676 DOI: 10.3390/ijms21165646] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/01/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases are a major burden for our society, affecting millions of people worldwide. A main goal of past and current research is to enhance our understanding of the mechanisms underlying proteotoxicity, a common theme among these incurable and debilitating conditions. Cell proteome alteration is considered to be one of the main driving forces that triggers neurodegeneration, and unraveling the biological complexity behind the affected molecular pathways constitutes a daunting challenge. This review summarizes the current state on key processes that lead to cellular proteotoxicity in Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, providing a comprehensive landscape of recent literature. A foundational understanding of how proteotoxicity affects disease etiology and progression may provide essential insight towards potential targets amenable of therapeutic intervention.
Collapse
|
18
|
Taran AS, Shuvalova LD, Lagarkova MA, Alieva IB. Huntington's Disease-An Outlook on the Interplay of the HTT Protein, Microtubules and Actin Cytoskeletal Components. Cells 2020; 9:E1514. [PMID: 32580314 PMCID: PMC7348758 DOI: 10.3390/cells9061514] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/17/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease is a severe and currently incurable neurodegenerative disease. An autosomal dominant mutation in the Huntingtin gene (HTT) causes an increase in the polyglutamine fragment length at the protein N-terminus. The consequence of the mutation is the death of neurons, mostly striatal neurons, leading to the occurrence of a complex of motor, cognitive and emotional-volitional personality sphere disorders in carriers. Despite intensive studies, the functions of both mutant and wild-type huntingtin remain poorly understood. Surprisingly, there is the selective effect of the mutant form of HTT even on nervous tissue, whereas the protein is expressed ubiquitously. Huntingtin plays a role in cell physiology and affects cell transport, endocytosis, protein degradation and other cellular and molecular processes. Our experimental data mining let us conclude that a significant part of the Huntingtin-involved cellular processes is mediated by microtubules and other cytoskeletal cell structures. The review attempts to look at unresolved issues in the study of the huntingtin and its mutant form, including their functions affecting microtubules and other components of the cell cytoskeleton.
Collapse
Affiliation(s)
- Aleksandra S. Taran
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1–73, Leninsky Gory, 119992 Moscow, Russia; (A.S.T.); (L.D.S.)
| | - Lilia D. Shuvalova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 1–73, Leninsky Gory, 119992 Moscow, Russia; (A.S.T.); (L.D.S.)
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
| | - Maria A. Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
| | - Irina B. Alieva
- Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 1a Malaya Pirogovskaya St., 119435 Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 1–40, Leninsky Gory, 119992 Moscow, Russia
| |
Collapse
|
19
|
Abstract
Huntingtin (HTT) is a scaffold protein mostly known because it gives rise to the severe and incurable inherited neurological disorder Huntington’s disease (HD) when mutated. The Huntingtin gene (HTT) carries a polymorphic trinucleotide expansion of CAGs in exon 1 that ranges from 9 to 35 in the non-HD affected population. However, if it exceeds 35 CAG repeats, the altered protein is referred to as mutant HTT and leads to the development of HD. Given the wide spectrum of severe symptoms developed by HD individuals, wild-type and mutant HTT have been mostly studied in the context of this disorder. However, HTT expression is ubiquitous and several peripheral symptoms in HD have been described, suggesting that HTT is of importance, not only in the central nervous system (CNS), but also in peripheral organs. Accordingly, HTT and mutant HTT may interfere with non-brain-related diseases. Correlative studies have highlighted a decreased cancer incidence in the HD population and both wild-type and mutant HTT have been implicated in tumor progression. In this review, we describe the current evidence linking wild-type and mutant HTT to cancer and discuss how CAG polymorphism, HTT function, and partners may influence carcinogenesis and metastatic progression.
Collapse
Affiliation(s)
- Morgane Sonia Thion
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris Cedex 05, France
| | - Sandrine Humbert
- Grenoble Institut des Neurosciences, GIN, Univ. Grenoble Alpes, Grenoble, France.,INSERM, U1216, Grenoble, France
| |
Collapse
|
20
|
Segatto I, Zompit MDM, Citron F, D'Andrea S, Vinciguerra GLR, Perin T, Berton S, Mungo G, Schiappacassi M, Marchini C, Amici A, Vecchione A, Baldassarre G, Belletti B. Stathmin Is Required for Normal Mouse Mammary Gland Development and Δ16HER2-Driven Tumorigenesis. Cancer Res 2018; 79:397-409. [PMID: 30478213 DOI: 10.1158/0008-5472.can-18-2488] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/17/2018] [Accepted: 11/19/2018] [Indexed: 11/16/2022]
Abstract
Postnatal development of the mammary gland relies on the maintenance of oriented cell division and apicobasal polarity, both of which are often deregulated in cancer. The microtubule (MT) network contributes to control these processes; however, very little is known about the impact of altered MT dynamics in the development of a complex organ and on the role played by MT-interacting proteins such as stathmin. In this study, we report that female stathmin knock-out (STM KO) mice are unable to nurse their litters due to frank impairment of mammary gland development. In mouse mammary epithelial cells, loss of stathmin compromised the trafficking of polarized proteins and the achievement of proper apicobasal polarity. In particular, prolactin receptor internalization and localization was altered in STM KO mammary epithelial cells, leading to decreased protein stability and downmodulation of the Prl/PrlR/STAT5 signaling pathway. Absence of stathmin induced alterations in mitotic spindle orientation, accumulation of mitotic defects, and apoptosis, overall contributing to tissue disorganization and further decreasing the expansion of the mammary epithelial compartment. Loss of stathmin in MMTV-Δ16HER2 transgenic mice decreased the incidence and increased the latency of these very aggressive mammary carcinomas. Collectively, these data identify the essential mammary protein stathmin as protumorigenic and suggest it may serve as a potential therapeutic target in breast cancer. SIGNIFICANCE: Stathmin expression is critical to maintain oriented cell division and apicobasal polarity in normal mammary glands and to establish a protumorigenic program that eventually sustains HER2-positive breast cancer formation in mice.
Collapse
Affiliation(s)
- Ilenia Segatto
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Mara De Marco Zompit
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Francesca Citron
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Sara D'Andrea
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Gian Luca Rampioni Vinciguerra
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.,Faculty of Medicine and Psychology, Department of Clinical and Molecular Medicine, University of Rome "Sapienza" Sant'Andrea Hospital, Rome, Italy
| | - Tiziana Perin
- Unit of Pathology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Stefania Berton
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Giorgia Mungo
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Monica Schiappacassi
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy
| | - Cristina Marchini
- Department of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Augusto Amici
- Department of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Andrea Vecchione
- Faculty of Medicine and Psychology, Department of Clinical and Molecular Medicine, University of Rome "Sapienza" Sant'Andrea Hospital, Rome, Italy
| | - Gustavo Baldassarre
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.
| | - Barbara Belletti
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
21
|
He Z, Kannan N, Nemirovsky O, Chen H, Connell M, Taylor B, Jiang J, Pilarski LM, Fleisch MC, Niederacher D, Pujana MA, Eaves CJ, Maxwell CA. BRCA1 controls the cell division axis and governs ploidy and phenotype in human mammary cells. Oncotarget 2018; 8:32461-32475. [PMID: 28427147 PMCID: PMC5464802 DOI: 10.18632/oncotarget.15688] [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: 02/08/2017] [Accepted: 02/16/2017] [Indexed: 11/25/2022] Open
Abstract
BRCA1 deficiency may perturb the differentiation hierarchy present in the normal mammary gland and is associated with the genesis of breast cancers that are genomically unstable and typically display a basal-like transcriptome. Oriented cell division is a mechanism known to regulate cell fates and to restrict tumor formation. We now show that the cell division axis is altered following shRNA-mediated BRCA1 depletion in immortalized but non-tumorigenic, or freshly isolated normal human mammary cells with graded consequences in progeny cells that include aneuploidy, perturbation of cell polarity in spheroid cultures, and a selective loss of cells with luminal features. BRCA1 depletion stabilizes HMMR abundance and disrupts cortical asymmetry of NUMA-dynein complexes in dividing cells such that polarity cues provided by cell-matrix adhesions were not able to orient division. We also show that immortalized mammary cells carrying a mutant BRCA1 allele (BRCA1 185delAG/+) reproduce many of these effects but in this model, oriented divisions were maintained through cues provided by CDH1+ cell-cell junctions. These findings reveal a previously unknown effect of BRCA1 suppression on mechanisms that regulate the cell division axis in proliferating, non-transformed human mammary epithelial cells and consequent downstream effects on the mitotic integrity and phenotype control of their progeny.
Collapse
Affiliation(s)
- Zhengcheng He
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nagarajan Kannan
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Laboratory Medicine and Pathology, Division of Experimental Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | - Oksana Nemirovsky
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Helen Chen
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian Taylor
- Department of Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Jihong Jiang
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Linda M Pilarski
- Department of Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Markus C Fleisch
- Department of Obstetrics and Gynaecology, Landesfrauenklinik, HELIOS University Medical Center, Wuppertal, Germany
| | - Dieter Niederacher
- Department of Gynaecology and Obstetrics, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Germany
| | - Miguel Angel Pujana
- Breast Cancer and Systems Biology Unit, Program Against Cancer Therapeutic Resistance (ProCure), Catalan Institute of Oncology, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher A Maxwell
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
22
|
Abstract
Asymmetric cell divisions balance stem cell proliferation and differentiation to sustain tissue morphogenesis and homeostasis. During asymmetric divisions, fate determinants and niche contacts segregate unequally between daughters, but little is known on how this is achieved mechanistically. In Drosophila neuroblasts and murine mammary stem cells, the association of the spindle orientation protein LGN with the stem cell adaptor Inscuteable has been connected to asymmetry. Here we report the crystal structure of Drosophila LGN in complex with the asymmetric domain of Inscuteable, which reveals a tetrameric arrangement of intertwined molecules. We show that Insc:LGN tetramers constitute stable cores of Par3–Insc-LGN-GαiGDP complexes, which cannot be dissociated by NuMA. In mammary stem cells, the asymmetric domain of Insc bound to LGN:GαiGDP suffices to drive asymmetric fate, and reverts aberrant symmetric divisions induced by p53 loss. We suggest a novel role for the Insc-bound pool of LGN acting independently of microtubule motors to promote asymmetric fate specification. During asymmetric divisions fate determinants and niche contacts segregate unequally between daughter cells, but the mechanism is unclear. Here the authors show that Insc:LGN tetramers promote assembly of Par3-Insc-LGN-GαiGDP complexes and asymmetric fate specification independently of microtubule motors.
Collapse
|
23
|
Ruzo A, Croft GF, Metzger JJ, Galgoczi S, Gerber LJ, Pellegrini C, Wang H, Fenner M, Tse S, Marks A, Nchako C, Brivanlou AH. Chromosomal instability during neurogenesis in Huntington's disease. Development 2018; 145:145/2/dev156844. [PMID: 29378824 DOI: 10.1242/dev.156844] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 12/03/2017] [Indexed: 12/21/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disease caused by expansion of CAG repeats in the Huntingtin gene (HTT). Neither its pathogenic mechanisms nor the normal functions of HTT are well understood. To model HD in humans, we engineered a genetic allelic series of isogenic human embryonic stem cell (hESC) lines with graded increases in CAG repeat length. Neural differentiation of these lines unveiled a novel developmental HD phenotype: the appearance of giant multinucleated telencephalic neurons at an abundance directly proportional to CAG repeat length, generated by a chromosomal instability and failed cytokinesis over multiple rounds of DNA replication. We conclude that disrupted neurogenesis during development is an important, unrecognized aspect of HD pathogenesis. To address the function of normal HTT protein we generated HTT+/- and HTT-/- lines. Surprisingly, the same phenotype emerged in HTT-/- but not HTT+/- lines. We conclude that HD is a developmental disorder characterized by chromosomal instability that impairs neurogenesis, and that HD represents a genetic dominant-negative loss of function, contrary to the prevalent gain-of-toxic-function hypothesis. The consequences of developmental alterations should be considered as a new target for HD therapies.
Collapse
Affiliation(s)
- Albert Ruzo
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Gist F Croft
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Jakob J Metzger
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA.,Center for Studies in Physics and Biology, The Rockefeller University, New York, NY 10065, USA
| | - Szilvia Galgoczi
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Lauren J Gerber
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Cecilia Pellegrini
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Hanbin Wang
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Maria Fenner
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Stephanie Tse
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Adam Marks
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Corbyn Nchako
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| | - Ali H Brivanlou
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
24
|
Ahmed MI, Elias S, Mould AW, Bikoff EK, Robertson EJ. The transcriptional repressor Blimp1 is expressed in rare luminal progenitors and is essential for mammary gland development. Development 2017; 143:1663-73. [PMID: 27190036 PMCID: PMC4874485 DOI: 10.1242/dev.136358] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/11/2016] [Indexed: 01/26/2023]
Abstract
Mammary gland morphogenesis depends on a tight balance between cell proliferation, differentiation and apoptosis, to create a defined functional hierarchy within the epithelia. The limited availability of stem cell/progenitor markers has made it challenging to decipher lineage relationships. Here, we identify a rare subset of luminal progenitors that express the zinc finger transcriptional repressor Blimp1, and demonstrate that this subset of highly clonogenic luminal progenitors is required for mammary gland development. Conditional inactivation experiments using K14-Cre and WAPi-Cre deleter strains revealed essential functions at multiple developmental stages. Thus, Blimp1 regulates proliferation, apoptosis and alveolar cell maturation during puberty and pregnancy. Loss of Blimp1 disrupts epithelial architecture and lumen formation both in vivo and in three-dimensional (3D) primary cell cultures. Collectively, these results demonstrate that Blimp1 is required to maintain a highly proliferative luminal subset necessary for mammary gland development and homeostasis. Highlighted article: In the mouse mammary gland, Blimp1 marks a rare progenitor population, and is required for cell proliferation and polarity as well as efficient milk production.
Collapse
Affiliation(s)
- Mohammed I Ahmed
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Salah Elias
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Arne W Mould
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Elizabeth K Bikoff
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | |
Collapse
|
25
|
Yu MS, Tanese N. Huntingtin Is Required for Neural But Not Cardiac/Pancreatic Progenitor Differentiation of Mouse Embryonic Stem Cells In vitro. Front Cell Neurosci 2017; 11:33. [PMID: 28270748 PMCID: PMC5318384 DOI: 10.3389/fncel.2017.00033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 02/06/2017] [Indexed: 12/25/2022] Open
Abstract
Mutation in the huntingtin (HTT) gene causes Huntington's disease (HD). It is an autosomal dominant trinucleotide-repeat expansion disease in which CAG repeat sequence expands to >35. This results in the production of mutant HTT protein with an increased stretch of glutamines near the N-terminus. The wild type HTT gene encodes a 350 kD protein whose function remains elusive. Mutant HTT protein has been implicated in transcription, axonal transport, cytoskeletal structure/function, signal transduction, and autophagy. HD is characterized by the appearance of nuclear inclusions and degeneration of the striatum. Although HTT protein is expressed early in embryos, most patients develop symptoms in mid-life. It is also unclear why the ubiquitously expressed mutant HTT specifically causes striatal atrophy. Wild type Htt is essential for development as Htt knockout mice die at day E7.5. Increasing evidence suggests mutant Htt may alter neurogenesis and development of striatal neurons resulting in neuronal loss. Using a mouse embryonic stem cell model, we examined the role of Htt in neural differentiation. We found cells lacking Htt inefficient in generating neural stem cells. In contrast differentiation into progenitors of mesoderm and endoderm lineages was not affected. The data suggests Htt is essential for neural but not cardiac/pancreatic progenitor differentiation of embryonic stem cells in vitro.
Collapse
Affiliation(s)
- Man Shan Yu
- Department of Microbiology, New York University School of Medicine, New York NY, USA
| | - Naoko Tanese
- Department of Microbiology, New York University School of Medicine, New York NY, USA
| |
Collapse
|
26
|
Tadenev ALD, Tarchini B. The Spindle Orientation Machinery Beyond Mitosis: When Cell Specialization Demands Polarization. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:209-225. [DOI: 10.1007/978-3-319-57127-0_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
27
|
Santoro A, Vlachou T, Carminati M, Pelicci PG, Mapelli M. Molecular mechanisms of asymmetric divisions in mammary stem cells. EMBO Rep 2016; 17:1700-1720. [PMID: 27872203 DOI: 10.15252/embr.201643021] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/04/2016] [Accepted: 10/25/2016] [Indexed: 01/16/2023] Open
Abstract
Stem cells have the remarkable ability to undergo proliferative symmetric divisions and self-renewing asymmetric divisions. Balancing of the two modes of division sustains tissue morphogenesis and homeostasis. Asymmetric divisions of Drosophila neuroblasts (NBs) and sensory organ precursor (SOP) cells served as prototypes to learn what we consider now principles of asymmetric mitoses. They also provide initial evidence supporting the notion that aberrant symmetric divisions of stem cells could correlate with malignancy. However, transferring the molecular knowledge of circuits underlying asymmetry from flies to mammals has proven more challenging than expected. Several experimental approaches have been used to define asymmetry in mammalian systems, based on daughter cell fate, unequal partitioning of determinants and niche contacts, or proliferative potential. In this review, we aim to provide a critical evaluation of the assays used to establish the stem cell mode of division, with a particular focus on the mammary gland system. In this context, we will discuss the genetic alterations that impinge on the modality of stem cell division and their role in breast cancer development.
Collapse
Affiliation(s)
- Angela Santoro
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Thalia Vlachou
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Manuel Carminati
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | | | - Marina Mapelli
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
28
|
Affiliation(s)
- Morgane S Thion
- École Normale Supérieure, Institut de Biologie de l'École Normale Supérieure (IBENS), Inserm U1024, CNRS UMR 8797, 46, rue d'Ulm, 75230 Paris Cedex 05, France
| | - Sandrine Humbert
- Université Grenoble Alpes, Grenoble Institut des Neurosciences GIN, chemin Fortuné Ferrini, 38000 Grenoble, France - Inserm U1216, chemin Fortuné Ferrini, 38000 Grenoble, France
| |
Collapse
|
29
|
di Pietro F, Echard A, Morin X. Regulation of mitotic spindle orientation: an integrated view. EMBO Rep 2016; 17:1106-30. [PMID: 27432284 DOI: 10.15252/embr.201642292] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/17/2016] [Indexed: 12/18/2022] Open
Abstract
Mitotic spindle orientation is essential for cell fate decisions, epithelial maintenance, and tissue morphogenesis. In most animal cell types, the dynein motor complex is anchored at the cell cortex and exerts pulling forces on astral microtubules to position the spindle. Early studies identified the evolutionarily conserved Gαi/LGN/NuMA complex as a key regulator that polarizes cortical force generators. In recent years, a combination of genetics, biochemistry, modeling, and live imaging has contributed to decipher the mechanisms of spindle orientation. Here, we highlight the dynamic nature of the assembly of this complex and discuss the molecular regulation of its localization. Remarkably, a number of LGN-independent mechanisms were described recently, whereas NuMA remains central in most pathways involved in recruiting force generators at the cell cortex. We also describe the emerging role of the actin cortex in spindle orientation and discuss how dynamic astral microtubule formation is involved. We further give an overview on instructive external signals that control spindle orientation in tissues. Finally, we discuss the influence of cell geometry and mechanical forces on spindle orientation.
Collapse
Affiliation(s)
- Florencia di Pietro
- Cell Division and Neurogenesis Laboratory, Ecole Normale Supérieure CNRS Inserm Institut de Biologie de l'Ecole Normale Supérieure (IBENS) PSL Research University, Paris, France Institute of Doctoral Studies (IFD), Sorbonne Universités Université Pierre et Marie Curie-Université Paris 6, Paris, France
| | - Arnaud Echard
- Membrane Traffic and Cell Division Laboratory, Cell Biology and Infection Department, Institut Pasteur, Paris, France Centre National de la Recherche Scientifique, Unité Mixte de Recherche 3691, Paris, France
| | - Xavier Morin
- Cell Division and Neurogenesis Laboratory, Ecole Normale Supérieure CNRS Inserm Institut de Biologie de l'Ecole Normale Supérieure (IBENS) PSL Research University, Paris, France
| |
Collapse
|
30
|
Chang NC, Chevalier FP, Rudnicki MA. Satellite Cells in Muscular Dystrophy - Lost in Polarity. Trends Mol Med 2016; 22:479-496. [PMID: 27161598 PMCID: PMC4885782 DOI: 10.1016/j.molmed.2016.04.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/21/2022]
Abstract
Recent findings employing the mdx mouse model for Duchenne muscular dystrophy (DMD) have revealed that muscle satellite stem cells play a direct role in contributing to disease etiology and progression of DMD, the most common and severe form of muscular dystrophy. Lack of dystrophin expression in DMD has critical consequences in satellite cells including an inability to establish cell polarity, abrogation of asymmetric satellite stem-cell divisions, and failure to enter the myogenic program. Thus, muscle wasting in dystrophic mice is not only caused by myofiber fragility but is exacerbated by intrinsic satellite cell dysfunction leading to impaired regeneration. Despite intense research and clinical efforts, there is still no effective cure for DMD. In this review we highlight recent research advances in DMD and discuss the current state of treatment and, importantly, how we can incorporate satellite cell-targeted therapeutic strategies to correct satellite cell dysfunction in DMD.
Collapse
Affiliation(s)
- Natasha C Chang
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Fabien P Chevalier
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
31
|
N-terminal Huntingtin Knock-In Mice: Implications of Removing the N-terminal Region of Huntingtin for Therapy. PLoS Genet 2016; 12:e1006083. [PMID: 27203582 PMCID: PMC4874551 DOI: 10.1371/journal.pgen.1006083] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/04/2016] [Indexed: 02/07/2023] Open
Abstract
The Huntington’s disease (HD) protein, huntingtin (HTT), is a large protein consisting of 3144 amino acids and has conserved N-terminal sequences that are followed by a polyglutamine (polyQ) repeat. Loss of Htt is known to cause embryonic lethality in mice, whereas polyQ expansion leads to adult neuronal degeneration. Whether N-terminal HTT is essential for neuronal development or contributes only to late-onset neurodegeneration remains unknown. We established HTT knock-in mice (N160Q-KI) expressing the first 208 amino acids of HTT with 160Q, and they show age-dependent HTT aggregates in the brain and neurological phenotypes. Importantly, the N-terminal mutant HTT also preferentially accumulates in the striatum, the brain region most affected in HD, indicating the importance of N-terminal HTT in selective neuropathology. That said, homozygous N160Q-KI mice are also embryonic lethal, suggesting that N-terminal HTT alone is unable to support embryonic development. Using Htt knockout neurons, we found that loss of Htt selectively affects the survival of developing neuronal cells, but not astrocytes, in culture. This neuronal degeneration could be rescued by a truncated HTT lacking the first 237 amino acids, but not by N-terminal HTT (1–208 amino acids). Also, the rescue effect depends on the region in HTT known to be involved in intracellular trafficking. Thus, the N-terminal HTT region may not be essential for the survival of developing neurons, but when carrying a large polyQ repeat, can cause selective neuropathology. These findings imply a possible therapeutic benefit of removing the N-terminal region of HTT containing the polyQ repeat to treat the neurodegeneration in HD. The 17 amino acids in the N-terminal region of huntingtin (HTT) are conserved in a wide range of species and are followed by a polyglutamine repeat whose expansion causes selective neurodegeneration in Huntington’s disease (HD). Loss of Htt can affect developing neurons and early embryonic development in mice. Whether N-terminal HTT is important for the survival of developing neurons or contributes mainly to a gain of toxic function in HD remains unknown. In the current study, we generated N-terminal mutant HTT knock-in mice and found that N-terminal HTT with an expanded polyQ repeat is unable to support the early development of mice, but can cause age-dependent neurological phenotypes. Further, we show that a truncated HTT without the N-terminal region can rescue the Htt loss-mediated degeneration of developing neurons. Our studies suggest that removal of the N-terminal region of mutant HTT could be a strategy to abolish the neuronal toxicity of mutant HTT.
Collapse
|
32
|
Thion MS, Tézenas du Montcel S, Golmard JL, Vacher S, Barjhoux L, Sornin V, Cazeneuve C, Bièche I, Sinilnikova O, Stoppa-Lyonnet D, Durr A, Humbert S. CAG repeat size in Huntingtin alleles is associated with cancer prognosis. Eur J Hum Genet 2016; 24:1310-5. [PMID: 26980106 DOI: 10.1038/ejhg.2016.13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 01/14/2016] [Accepted: 02/01/2016] [Indexed: 12/26/2022] Open
Abstract
The abnormal expansion of a ≥36 CAG unit tract in the Huntingtin gene (HTT) leads to Huntington's disease (HD), but has also been associated with cancer: the incidence of cancer is lower in HD patients than in age-matched controls, but HD-causing variants of HTT accelerate the progression of breast tumors and the development of metastases in mouse models of breast cancer. To investigate the relationship between HTT CAGs and cancer, data concerning 2407 women with BRCA1 or BRCA2 mutations that predispose to breast and ovarian cancers and 431 patients with breast cancer without family histories were studied; the size of the CAG expansions on both HTT alleles was determined in each subject. The proportion of individuals carrying a CAG expansion in a pathological range for HD was 10 times more frequent than previously reported in the literature. In carriers of BRCA2 mutations, the length of the HTT CAG tract was correlated with lower incidence of ovarian cancer. Among carriers of BRCA1 mutations who developed a breast cancer, its onset occurred 2.4 years earlier in individuals with intermediate HTT alleles (≥27) than in those with a CAG tract <27. Finally, in patients with sporadic HER2 breast cancer, metastasis increased by a factor of 11.10 per 10 additional CAG repeats in HTT. We concluded that whereas long CAG length could be associated with lower cancer incidence, it could also be paradoxically associated with cancer severity (age of apparition and metastasis development).
Collapse
Affiliation(s)
- Morgane Sonia Thion
- Institut Curie, Paris, France.,CNRS UMR 3306, Orsay, France.,INSERM U1005, Orsay, France.,University Paris Sud 11, Orsay, France
| | - Sophie Tézenas du Montcel
- Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris, Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France.,INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France
| | - Jean-Louis Golmard
- Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Laure Barjhoux
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Valérie Sornin
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Cécile Cazeneuve
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, INSERM U975, CNRS UMR7225, UPMC Paris VI UMR S975, Paris, France.,Assistance Publique-Hôpitaux de Paris, Département de Génétique, Centre Hospitalier Universitaire Pitié-Salpêtrière, Paris, France
| | - Ivan Bièche
- Institut Curie, Paris, France.,University Paris Descartes, Sorbonne Paris Cité, France
| | - Olga Sinilnikova
- Unité Mixte de Génétique Constitutionnelle des Cancers Fréquents, Centre Hospitalier Universitaire de Lyon, Centre Léon Bérard, Lyon, France
| | | | - Alexandra Durr
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, INSERM U975, CNRS UMR7225, UPMC Paris VI UMR S975, Paris, France.,Assistance Publique-Hôpitaux de Paris, Département de Génétique, Centre Hospitalier Universitaire Pitié-Salpêtrière, Paris, France
| | - Sandrine Humbert
- University of Grenoble Alpes, Grenoble Institut des Neurosciences, INSERM U1216, Grenoble, France.,INSERM, U1216, Grenoble, France
| |
Collapse
|
33
|
Faraldo MM, Glukhova MA. Regulating the regulator: Numb acts upstream of p53 to control mammary stem and progenitor cell. J Cell Biol 2016; 211:737-9. [PMID: 26598611 PMCID: PMC4657175 DOI: 10.1083/jcb.201510104] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
In this issue, Tosoni et al. (2015. J. Cell Biol.http://dx.doi.org/10.1083/jcb.201505037) report that cell fate determinant and tumor suppressor Numb imposes asymmetric cell divisions in mammary stem cells by regulating p53. Numb thereby restricts mammary stem cell expansion and controls the proliferation and lineage-specific characteristics of their progeny.
Collapse
Affiliation(s)
- Marisa M Faraldo
- Institut Curie, PSL Research University, F-75248 Paris, France Centre National de la Recherche Scientifique, UMR144, F-75248 Paris, France Institut National de la Santé et de la Recherche Médicale, F-75013 Paris, France
| | - Marina A Glukhova
- Institut Curie, PSL Research University, F-75248 Paris, France Centre National de la Recherche Scientifique, UMR144, F-75248 Paris, France Institut National de la Santé et de la Recherche Médicale, F-75013 Paris, France
| |
Collapse
|
34
|
|
35
|
Lopes C, Aubert S, Bourgois-Rocha F, Barnat M, Rego AC, Déglon N, Perrier AL, Humbert S. Dominant-Negative Effects of Adult-Onset Huntingtin Mutations Alter the Division of Human Embryonic Stem Cells-Derived Neural Cells. PLoS One 2016; 11:e0148680. [PMID: 26863614 PMCID: PMC4749329 DOI: 10.1371/journal.pone.0148680] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/20/2016] [Indexed: 01/30/2023] Open
Abstract
Mutations of the huntingtin protein (HTT) gene underlie both adult-onset and juvenile forms of Huntington’s disease (HD). HTT modulates mitotic spindle orientation and cell fate in mouse cortical progenitors from the ventricular zone. Using human embryonic stem cells (hESC) characterized as carrying mutations associated with adult-onset disease during pre-implantation genetic diagnosis, we investigated the influence of human HTT and of an adult-onset HD mutation on mitotic spindle orientation in human neural stem cells (NSCs) derived from hESCs. The RNAi-mediated silencing of both HTT alleles in neural stem cells derived from hESCs disrupted spindle orientation and led to the mislocalization of dynein, the p150Glued subunit of dynactin and the large nuclear mitotic apparatus (NuMA) protein. We also investigated the effect of the adult-onset HD mutation on the role of HTT during spindle orientation in NSCs derived from HD-hESCs. By combining SNP-targeting allele-specific silencing and gain-of-function approaches, we showed that a 46-glutamine expansion in human HTT was sufficient for a dominant-negative effect on spindle orientation and changes in the distribution within the spindle pole and the cell cortex of dynein, p150Glued and NuMA in neural cells. Thus, neural derivatives of disease-specific human pluripotent stem cells constitute a relevant biological resource for exploring the impact of adult-onset HD mutations of the HTT gene on the division of neural progenitors, with potential applications in HD drug discovery targeting HTT-dynein-p150Glued complex interactions.
Collapse
Affiliation(s)
- Carla Lopes
- Grenoble Institut des Neurosciences, Grenoble, France
- INSERM U836, Grenoble, France
- Grenoble Alpes University, Grenoble, France
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research of the University of Coimbra (IIIUC), Coimbra, Portugal
| | | | - Fany Bourgois-Rocha
- Inserm U861, I-STEM, AFM, Corbeil-Essonnes, France
- UEVE U861, I-STEM, AFM, Evry, France
| | - Monia Barnat
- Grenoble Institut des Neurosciences, Grenoble, France
- INSERM U836, Grenoble, France
- Grenoble Alpes University, Grenoble, France
| | - Ana Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Nicole Déglon
- Lausanne University Hospital (CHUV), Department of Clinical Neurosciences (DNC), Laboratory of Cellular and Molecular Neurotherapies (LNCM), Lausanne, Switzerland
| | - Anselme L. Perrier
- Inserm U861, I-STEM, AFM, Corbeil-Essonnes, France
- UEVE U861, I-STEM, AFM, Evry, France
- * E-mail: (ALP); (SH)
| | - Sandrine Humbert
- Grenoble Institut des Neurosciences, Grenoble, France
- INSERM U836, Grenoble, France
- Grenoble Alpes University, Grenoble, France
- * E-mail: (ALP); (SH)
| |
Collapse
|
36
|
Carminati M, Gallini S, Pirovano L, Alfieri A, Bisi S, Mapelli M. Concomitant binding of Afadin to LGN and F-actin directs planar spindle orientation. Nat Struct Mol Biol 2016; 23:155-63. [DOI: 10.1038/nsmb.3152] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 11/26/2015] [Indexed: 12/19/2022]
|
37
|
Ballard MS, Zhu A, Iwai N, Stensrud M, Mapps A, Postiglione MP, Knoblich JA, Hinck L. Mammary Stem Cell Self-Renewal Is Regulated by Slit2/Robo1 Signaling through SNAI1 and mINSC. Cell Rep 2015; 13:290-301. [PMID: 26440891 PMCID: PMC4606466 DOI: 10.1016/j.celrep.2015.09.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/03/2015] [Accepted: 09/01/2015] [Indexed: 01/12/2023] Open
Abstract
Tissue homeostasis requires somatic stem cell maintenance; however, mechanisms regulating this process during organogenesis are not well understood. Here, we identify asymmetrically renewing basal and luminal stem cells in the mammary end bud. We demonstrate that SLIT2/ROBO1 signaling regulates the choice between self-renewing asymmetric cell divisions (ACDs) and expansive symmetric cell divisions (SCDs) by governing Inscuteable (mInsc), a key member of the spindle orientation machinery, through the transcription factor Snail (SNAI1). Loss of SLIT2/ROBO1 signaling increases SNAI1 in the nucleus. Overexpression of SNAI1 increases mInsc expression, an effect that is inhibited by SLIT2 treatment. Increased mInsc does not change cell proliferation in the mammary gland (MG) but instead causes more basal cap cells to divide via SCD, at the expense of ACD, leading to more stem cells and larger outgrowths. Together, our studies provide insight into how the number of mammary stem cells is regulated by the extracellular cue SLIT2.
Collapse
Affiliation(s)
- Mimmi S Ballard
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Anna Zhu
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Naomi Iwai
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Michael Stensrud
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Department of Biology, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Aurelia Mapps
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Maira Pia Postiglione
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Juergen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Lindsay Hinck
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
38
|
Thion MS, McGuire JR, Sousa CM, Fuhrmann L, Fitamant J, Leboucher S, Vacher S, du Montcel ST, Bièche I, Bernet A, Mehlen P, Vincent-Salomon A, Humbert S. Unraveling the Role of Huntingtin in Breast Cancer Metastasis. J Natl Cancer Inst 2015; 107:djv208. [PMID: 26293574 DOI: 10.1093/jnci/djv208] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 07/06/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Huntingtin (HTT) is mutated in Huntington's disease but is ubiquitously expressed, and mutant HTT influences cancer progression. We investigated wild-type HTT function during breast cancer. METHODS We analyzed HTT and ZO1 expression as well as the HTT phosphoserine 421-activated form (S421-P-HTT) in human breast cancer tissues by quantitative reverse transcription polymerase chain reaction and immunohistochemistry. We performed in vitro migration and invasion assays as well as in vivo tail vein injections of the metastatic 4T1 cells in BALB/c mice (n = 11 per group). We analyzed tumor progression in knock-in mice with modified S421 crossed with the MMTV-PyVT mammary cancer model (at least n = 12 per group). Data were analyzed with unpaired t tests, analysis of variance, Pearson or Spearman correlation, and Mann Whitney or Kruskal-Wallis tests. All statistical tests were two-sided. RESULTS Levels of HTT and of S421-P-HTT are abnormally low in poorly differentiated and metastatic human breast cancers. HTT expression is downregulated in invasive compared with in situ carcinoma (P < .001). In BALB/c mice, silencing of HTT promotes lung colonization by a metastatic mammary cancer cell line (P = .005) and S421-unphosphorylatable-HTT accelerates cancer progression. HTT interacts with ZO1 and regulates both its expression and its localization to tight junctions. In human breast tumors, the patterns of HTT and ZO1 expression are similar (Pearson correlation coefficient = 0.66, P < .001). CONCLUSIONS HTT may inhibit breast tumor dissemination through maintenance of ZO1 at tight junctions. Downregulation of HTT transcript and protein levels is a prognostic factor for poor prognosis and metastasis development.
Collapse
Affiliation(s)
- Morgane S Thion
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - John R McGuire
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Cristovao M Sousa
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Laetitia Fuhrmann
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Julien Fitamant
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Sophie Leboucher
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Sophie Vacher
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Sophie Tezenas du Montcel
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Ivan Bièche
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Agnès Bernet
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Patrick Mehlen
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Anne Vincent-Salomon
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH)
| | - Sandrine Humbert
- Institut Curie, Paris, France (MST, JRM, CMS, LF, SL, SV, IB, AVS, SH); CNRS UMR 3306, Orsay, France (MST, JRM, CMS, SL, SH); INSERM U1005, Orsay, France (MST, JRM, CMS, SL, SH); University Paris Sud 11, Orsay, France (MST); Apoptosis, Cancer and Development Laboratory, Equipe labellisée 'La Ligue', Centre National de la Recherche Scientifique UMR5238, Université de Lyon, Lyon, France (JF, AB, PM); Department of Biostatistics and Medical Informatics, Assistance Publique-Hôpitaux de Paris (STdM); Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); INSERM, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France (STdM); University Paris Descartes, Sorbonne Paris Cité, France (IB); Department of Pathology, Institut Curie, Paris, France (AVS); Univ. Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Grenoble, France (SH); INSERM U836, Grenoble, France (SH).
| |
Collapse
|
39
|
Srivastava D, Chakrabarti O. Ubiquitin in regulation of spindle apparatus and its positioning: implications in development and disease. Biochem Cell Biol 2015; 93:273-81. [PMID: 26110206 DOI: 10.1139/bcb-2015-0011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Emerging data implicates ubiquitination, a post-translational modification, in regulating essential cellular events, one of them being mitosis. In this review we discuss how various E3 ligases modulate the cortical proteins such as dynein, LGN, NuMa, Gα, along with polymerization, stability, and integrity of spindles. These are responsible for regulating symmetric cell division. Some of the ubiquitin ligases regulating these proteins include PARK2, BRCA1/BARD1, MGRN1, SMURF2, and SIAH1; these play a pivotal role in the correct positioning of the spindle apparatus. A direct connection between developmental or various pathological disorders and the ubiquitination mediated cortical regulation is rather speculative, though deletions or mutations in them lead to developmental disorders and disease conditions.
Collapse
Affiliation(s)
- Devika Srivastava
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-1, Block-AF, Bidhannagar, Kolkata, West Bengal 700064, India
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-1, Block-AF, Bidhannagar, Kolkata, West Bengal 700064, India
| | - Oishee Chakrabarti
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-1, Block-AF, Bidhannagar, Kolkata, West Bengal 700064, India
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Sector-1, Block-AF, Bidhannagar, Kolkata, West Bengal 700064, India
| |
Collapse
|
40
|
Huntingtin is required for ciliogenesis and neurogenesis during early Xenopus development. Dev Biol 2015; 408:305-15. [PMID: 26192473 DOI: 10.1016/j.ydbio.2015.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/09/2015] [Accepted: 07/13/2015] [Indexed: 11/21/2022]
Abstract
Huntington's Disease (HD) is a neurodegenerative disorder that results from the abnormal expansion of poly-glutamine (polyQ) repeats in the Huntingtin (HTT) gene. Although HTT has been linked to a variety of cellular events, it is still not clear what the physiological functions of the protein are. Because of its critical role during mouse embryonic mouse development, we investigated the functions of Htt during early Xenopus embryogenesis. We find that reduction of Htt levels affects cilia polarity and function and causes whole body paralysis. Moreover, Htt loss of function leads to abnormal development of trigeminal and motor neurons. Interestingly, these phenotypes are partially rescued by either wild-type or expanded HTT. These results show that the Htt activity is required for normal embryonic development, and highlight the usefulness of the Xenopus system for investigating proteins involved in human diseases.
Collapse
|
41
|
Elias S, McGuire JR, Yu H, Humbert S. Huntingtin Is Required for Epithelial Polarity through RAB11A-Mediated Apical Trafficking of PAR3-aPKC. PLoS Biol 2015; 13:e1002142. [PMID: 25942483 PMCID: PMC4420272 DOI: 10.1371/journal.pbio.1002142] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/24/2015] [Indexed: 11/19/2022] Open
Abstract
The establishment of apical-basolateral polarity is important for both normal development and disease, for example, during tumorigenesis and metastasis. During this process, polarity complexes are targeted to the apical surface by a RAB11A-dependent mechanism. Huntingtin (HTT), the protein that is mutated in Huntington disease, acts as a scaffold for molecular motors and promotes microtubule-based dynamics. Here, we investigated the role of HTT in apical polarity during the morphogenesis of the mouse mammary epithelium. We found that the depletion of HTT from luminal cells in vivo alters mouse ductal morphogenesis and lumen formation. HTT is required for the apical localization of PAR3-aPKC during epithelial morphogenesis in virgin, pregnant, and lactating mice. We show that HTT forms a complex with PAR3, aPKC, and RAB11A and ensures the microtubule-dependent apical vesicular translocation of PAR3-aPKC through RAB11A. We thus propose that HTT regulates polarized vesicular transport, lumen formation and mammary epithelial morphogenesis. Huntingtin—the protein that is aberrant in Huntington Disease—regulates apical vesicular trafficking to help establish apical-basolateral polarity during the development of mammary epithelia. In the adult mammary gland, tissue architecture is maintained through the regulation of the polarity of epithelial cells, which organize around a central cavity called the lumen. The mammary epithelium comprises a basal layer, which contains myoepithelial contractile cells and so-called mammary stem cells, and a luminal layer of cells organized around the lumen. The establishment of apical-basolateral polarity in luminal cells allows the separation of the apical and basolateral membranes and the maturation of cell–cell junctions. The protein complex composed of PAR3, PAR6, and aPKC regulates apical polarity in several tissues, including the mammary epithelium, and it is known that the loss of PAR3 and aPKC interferes with mammary gland development and promotes mammary tumor metastasis. RAB11A, a protein that regulates intracellular trafficking, coordinates apical translocation of PAR3-PAR6-aPKC. Huntingtin (HTT), the protein mutated in Huntington disease, modulates RAB11A activity and also regulates the microtubule-based vesicular trafficking in neurons. Using MCF10A, MDCK 2-D and 3-D cell cultures, and mouse models, we demonstrate here that HTT coordinates the apical vesicular trafficking of PAR3-PAR6-aPKC through RAB11A. We show that loss of HTT in luminal cells alters apical polarity, tissue architecture and the maturation of luminal cells during pregnancy and lactation in the mouse. Together, these findings uncover HTT-mediated vesicular trafficking as a new pathway in the establishment of epithelial apical polarity, with potential implications for health and disease.
Collapse
Affiliation(s)
- Salah Elias
- Institut Curie, Orsay, France
- CNRS UMR 3306, Orsay, France
- INSERM U1005, Orsay, France
| | - John Russel McGuire
- Institut Curie, Orsay, France
- CNRS UMR 3306, Orsay, France
- INSERM U1005, Orsay, France
| | - Hua Yu
- Institut Curie, Orsay, France
- CNRS UMR 3306, Orsay, France
- INSERM U1005, Orsay, France
| | - Sandrine Humbert
- Institut Curie, Orsay, France
- CNRS UMR 3306, Orsay, France
- INSERM U1005, Orsay, France
- Grenoble Institut des Neurosciences, University Grenoble Alpes, Grenoble, France
- INSERM U836, Grenoble, France
- * E-mail:
| |
Collapse
|
42
|
Fan Y, Zhao HC, Liu J, Tan T, Ding T, Li R, Zhao Y, Yan J, Sun X, Yu Y, Qiao J. Aberrant expression of maternal Plk1 and Dctn3 results in the developmental failure of human in-vivo- and in-vitro-matured oocytes. Sci Rep 2015; 5:8192. [PMID: 25645239 PMCID: PMC4314639 DOI: 10.1038/srep08192] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 01/12/2015] [Indexed: 12/31/2022] Open
Abstract
Fertilisation is the first step in embryonic development, and dynamic changes of key genes may potentially improve assisted reproduction techniques efficiency during this process. Here, we analysed genes that were differentially expressed between oocytes and zygotes and focused on cytokinesis-related genes. Plk1 and Dctn3 were identified as showing dramatic changes in expression during fertilisation and were suggested to play a key role in inducing aneuploidy in zygotes and 8-cell embryos. Moreover, we found that maternal Plk1 and Dctn3 were expressed at lower levels in in vitro matured oocytes, which may have contributed to the high ratio of resulting embryos with abnormal Plk1 and Dctn3 expression levels, thereby reducing the developmental competence of the resulting embryos. Furthermore, the overexpression of Dctn3 can silence Plk1 expression, which suggests a potential regulation mechanism. In conclusion, our present study showed that aberrant expression of Plk1 and Dctn3 increases embryo aneuploidy and developmental failure, particularly in in vitro matured oocytes. Our results facilitate a better understanding of the effects of oocyte maternal gene expression on embryonic development and can be used to improve the outcome of assisted reproduction techniques.
Collapse
Affiliation(s)
- Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Hong-Cui Zhao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Jianqiao Liu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Tao Tan
- Yunnan Key Laboratory of Primate Biomedical Research and Kunming Biomed International and National Engineering Research Center of Biomedicine and Animal Science, Kunming, 650500, China
| | - Ting Ding
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Rong Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Yue Zhao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Jie Yan
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yang Yu
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Jie Qiao
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| |
Collapse
|
43
|
Mutant huntingtin affects cortical progenitor cell division and development of the mouse neocortex. J Neurosci 2014; 34:10034-40. [PMID: 25057205 DOI: 10.1523/jneurosci.0715-14.2014] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A polyglutamine expansion in huntingtin (HTT) causes the specific death of adult neurons in Huntington's disease (HD). Most studies have thus focused on mutant HTT (mHTT) toxicity in adulthood, and its developmental effects have been largely overlooked. We found that mHTT caused mitotic spindle misorientation in cultured cells by altering the localization of dynein, NuMA, and the p150(Glued) subunit of dynactin to the spindle pole and cell cortex and of CLIP170 and p150(Glued) to microtubule plus-ends. mHTT also affected spindle orientation in dividing mouse cortical progenitors, altering the thickness of the developing cortex. The serine/threonine kinase Akt, which regulates HTT function, rescued the spindle misorientation caused by the mHTT, by serine 421 (S421) phosphorylation, in cultured cells and in mice. Thus, cortical development is affected in HD, and this early defect can be rescued by HTT phosphorylation at S421.
Collapse
|
44
|
Saadaoui M, Machicoane M, di Pietro F, Etoc F, Echard A, Morin X. Dlg1 controls planar spindle orientation in the neuroepithelium through direct interaction with LGN. ACTA ACUST UNITED AC 2014; 206:707-17. [PMID: 25202028 PMCID: PMC4164945 DOI: 10.1083/jcb.201405060] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dlg1 recruits LGN to the cortex of cells in the chick neuroepithelium and may provide instructive cues that drive planar spindle orientation. Oriented cell divisions are necessary for the development of epithelial structures. Mitotic spindle orientation requires the precise localization of force generators at the cell cortex via the evolutionarily conserved LGN complex. However, polarity cues acting upstream of this complex in vivo in the vertebrate epithelia remain unknown. In this paper, we show that Dlg1 is localized at the basolateral cell cortex during mitosis and is necessary for planar spindle orientation in the chick neuroepithelium. Live imaging revealed that Dlg1 is required for directed spindle movements during metaphase. Mechanistically, we show that direct interaction between Dlg1 and LGN promotes cortical localization of the LGN complex. Furthermore, in human cells dividing on adhesive micropatterns, homogenously localized Dlg1 recruited LGN to the mitotic cortex and was also necessary for proper spindle orientation. We propose that Dlg1 acts primarily to recruit LGN to the cortex and that Dlg1 localization may additionally provide instructive cues for spindle orientation.
Collapse
Affiliation(s)
- Mehdi Saadaoui
- Institut de Biologie de l'École Normale Supérieure, Ecole Normale Supérieure, F-75005 Paris, France Institut National de la Santé et de la Recherche Medicale, U1024, F-75005 Paris, France Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8197, F-75005 Paris, France
| | - Mickaël Machicoane
- Membrane Traffic and Cell Division Laboratory, Institut Pasteur, F-75015 Paris, France Centre National de la Recherche Scientifique, Unité de Recherche Associée 2582, F-75015 Paris, France Cellule Pasteur-Université Pierre et Marie Curie, Université Pierre et Marie Curie, F-75015 Paris, France
| | - Florencia di Pietro
- Institut de Biologie de l'École Normale Supérieure, Ecole Normale Supérieure, F-75005 Paris, France Institut National de la Santé et de la Recherche Medicale, U1024, F-75005 Paris, France Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8197, F-75005 Paris, France Institute of Doctoral Studies (IFD), Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 6, F-75252 Paris, France
| | - Fred Etoc
- Institut de Biologie de l'École Normale Supérieure, Ecole Normale Supérieure, F-75005 Paris, France Institut National de la Santé et de la Recherche Medicale, U1024, F-75005 Paris, France Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8197, F-75005 Paris, France
| | - Arnaud Echard
- Membrane Traffic and Cell Division Laboratory, Institut Pasteur, F-75015 Paris, France Centre National de la Recherche Scientifique, Unité de Recherche Associée 2582, F-75015 Paris, France
| | - Xavier Morin
- Institut de Biologie de l'École Normale Supérieure, Ecole Normale Supérieure, F-75005 Paris, France Institut National de la Santé et de la Recherche Medicale, U1024, F-75005 Paris, France Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8197, F-75005 Paris, France
| |
Collapse
|