1
|
Vinopal S, Bradke F. Centrosomal and acentrosomal microtubule nucleation during neuronal development. Curr Opin Neurobiol 2025; 92:103016. [PMID: 40147111 DOI: 10.1016/j.conb.2025.103016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025]
Abstract
Neurons rely on the microtubule cytoskeleton to create and maintain their sophisticated cellular architectures. Advances in cryogenic electron microscopy, expansion microscopy, live imaging, and gene editing have enabled novel insights into mechanisms of centrosomal and acentrosomal microtubule nucleation, the key process generating new microtubules. This has paved the way for the functional dissection of distinct microtubule networks that regulate various processes during neuronal development, including neuronal delamination, polarization, migration, maturation, and synapse function. We review recent progress in understanding the molecular concepts of microtubule nucleation, how these concepts underlie neurodevelopmental processes, and pinpoint the open questions. Since microtubules play a pivotal role in axon regeneration within the adult central nervous system, understanding the processes of microtubule nucleation could inform strategies to enhance the regenerative capabilities of neurons in the future.
Collapse
Affiliation(s)
- Stanislav Vinopal
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyne University (UJEP), Usti nad Labem, Czech Republic.
| | - Frank Bradke
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
2
|
Wang L, Bu T, Wu X, Gao S, Yun D, Mao B, Li H, Silvestrini B, Li L, Sun F, Cheng CY. Microtubule-Associated Proteins (MAPs) Are Multifunctional Cytoskeletal Proteins in the Testis That Regulate Spermatogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:411-431. [PMID: 40301267 DOI: 10.1007/978-3-031-82990-1_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Microtubule-associated proteins (MAPs) refer to a large superfamily of proteins that bind to microtubules (MTs) structurally, modulating the rapid transition of MTs from a stable state (polymerized) to shrinkage (or catastrophe) via depolymerization through a meta-stable state. Changes of MTs from an assembled structure as linear protofilaments that are a packed/bundled ultrastructure to disassembled subunits of heterodimers of α-/ß-tubulins (or oligomers) can take place in milliseconds within a living cell. These heterodimers can also be rapidly phosphorylated, becoming GTP-bound, or rapidly polymerized into linear protofilaments of MT again. It is such rapid cyclic changes of MTs that support cellular development, growth, and changes in cell shape in response to changes in development or other physiological phenomena, such as the series of cellular events during spermatogenesis, cell divisions, and in response to environmental toxicants to protect cellular life. In this review, we seek to give a concise update and discussion on MAPs. Particularly, we focus on a specific member of the structural MAPs, namely MAP1a, and its interaction with the microtubule affinity regulatory kinases (MARKs, including MARK1, 2, 3, and 4, all are Ser/Thr protein kinases) in particular MARK4, and how these two MAPs work together to regulate MT dynamics in Sertoli cells to support germ cell development. This information should be helpful to investigators who seek to better understand the role of MAPs in testis biology.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
- Department of AnesthesiologyAffiliated Hospital of Guangdong Medical University Zhanjiang City, Guangdong Province, China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sheng Gao
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Damin Yun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Baiping Mao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huitao Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Karatas E, Gulec A, Korkmaz M, Karaman ZF, Kiraz A, Per H, Dundar M. Brain malformation, neurodevelopmental disorder and epilepsy in a case of two rare genetic diseases: overlapping phenotype. Neurogenetics 2024; 26:16. [PMID: 39724270 DOI: 10.1007/s10048-024-00795-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
In most cases there is a single etiological factor causing neuromotor developmental delay and epilepsy while sometimes more than one gene may be involved. These include the autosomal recessive inherited CAMSAP1 gene, which is associated with cortical developmental malformations such as pachygyria and lissencephaly and the autosomal dominant inherited NBEA gene, which plays crucial roles in vesicle trafficking as well as synapse structure and function. Loss of function of both genes together is a well-known disease mechanism. We report a 7-year-old girl with early-onset epilepsy, severe neuromotor developmental delay and brain malformation. Whole exome analysis of the patient revealed c.1153C > T p.Gln385* nonsense homozygous likely pathogenic variant in CAMSAP1 gene and c.6867G > A p.Trp2289* nonsense heterozygous pathogenic de novo variant in NBEA gene. A small number of cases associated with these genes have been reported. We report the 8th case reported in the CAMSAP1 gene and the overlapping phenotype in these two genes.
Collapse
Affiliation(s)
- Emine Karatas
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Ayten Gulec
- Department of Pediatric Neurology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Maide Korkmaz
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Zehra Filiz Karaman
- Department of Radiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Aslihan Kiraz
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Huseyin Per
- Department of Pediatric Neurology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Munis Dundar
- Department of Medical Genetics, Faculty of Medicine, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
4
|
Duan D, Koleske AJ. Phase separation of microtubule-binding proteins - implications for neuronal function and disease. J Cell Sci 2024; 137:jcs263470. [PMID: 39679446 PMCID: PMC11795294 DOI: 10.1242/jcs.263470] [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] [Indexed: 12/17/2024] Open
Abstract
Protein liquid-liquid phase separation (LLPS) is driven by intrinsically disordered regions and multivalent binding domains, both of which are common features of diverse microtubule (MT) regulators. Many in vitro studies have dissected the mechanisms by which MT-binding proteins (MBPs) regulate MT nucleation, stabilization and dynamics, and investigated whether LLPS plays a role in these processes. However, more recent in vivo studies have focused on how MBP LLPS affects biological functions throughout neuronal development. Dysregulation of MBP LLPS can lead to formation of aggregates - an underlying feature in many neurodegenerative diseases - such as the tau neurofibrillary tangles present in Alzheimer's disease. In this Review, we highlight progress towards understanding the regulation of MT dynamics through the lens of phase separation of MBPs and associated cytoskeletal regulators, from both in vitro and in vivo studies. We also discuss how LLPS of MBPs regulates neuronal development and maintains homeostasis in mature neurons.
Collapse
Affiliation(s)
- Daisy Duan
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Anthony J. Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
- Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
5
|
Li Y, Zhang R, Ren J, Chen W, Zhou Z, Xu H, Li D, Cheng H, Xie Q, Ji W, Feng W, Liang X, Meng W. CAMSAP3 forms dimers via its α-helix domain that directly stabilize non-centrosomal microtubule minus ends. J Cell Sci 2024; 137:jcs263609. [PMID: 39479887 DOI: 10.1242/jcs.263609] [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: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/02/2024] Open
Abstract
Microtubules are vital components of the cytoskeleton. Their plus ends are dynamic and respond to changes in cell morphology, whereas the minus ends are stable and serve a crucial role in microtubule seeding and maintaining spatial organization. In mammalian cells, the calmodulin-regulated spectrin-associated proteins (CAMSAPs), play a key role in directly regulating the dynamics of non-centrosomal microtubules minus ends. However, the molecular mechanisms are not yet fully understood. Our study reveals that CAMSAP3 forms dimers through its C-terminal α-helix; this dimerization not only enhances the microtubule-binding affinity of the CKK domain but also enables the CKK domain to regulate the dynamics of microtubules. Furthermore, CAMSAP3 also specializes in decorating at the minus end of microtubules through the combined action of the microtubule-binding domain (MBD) and the C-terminal α-helix, thereby achieving dynamic regulation of the minus ends of microtubules. These findings are crucial for advancing our understanding and treatment of diseases associated with non-centrosomal microtubules.
Collapse
Affiliation(s)
- Yuejia Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jinqi Ren
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Chen
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhengrong Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- Neuroscience Center, Department of Basic Medical Sciences, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Honglin Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dong Li
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Haisu Cheng
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qi Xie
- Wangjing Hospital of China Academy of Chinese Medical Sciences, Huajiadi Street, Chaoyang District, Beijing 100102, China
| | - Wei Ji
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong 510320, China
| | - Wei Feng
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Liang
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
6
|
Dahawi M, de Sainte Agathe JM, Elmagzoub MS, Ahmed EA, Buratti J, Courtin T, Noé E, Bogoin J, Copin B, Elmugadam FA, Abdelgadir WA, Ahmed AKMA, Daldoum MA, Altayeb RMI, Bashir M, Khalid LM, Gamil S, Baldassari S, Elsayed L, Keren B, Nuel G, Ahmed AE, Leguern E. Genetic heterogeneity in familial forms of genetic generalized epilepsy: from mono- to oligogenism. Hum Genomics 2024; 18:130. [PMID: 39574152 PMCID: PMC11583555 DOI: 10.1186/s40246-024-00659-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 08/16/2024] [Indexed: 11/24/2024] Open
Abstract
Genetic generalized epilepsy (GGE) including childhood absence epilepsy, juvenile absence epilepsy, juvenile myoclonic epilepsy (JME), and GGE with tonic-clonic seizures (TCS) (GGE-TCS), is genetically influenced with a two- to four- fold increased risk in the first-degree relatives of patients. Since large families with GGE are very rare, international studies have focused on sporadic GGE patients using whole exome sequencing, suggesting that GGE are highly genetically heterogeneous and rather involve rare or ultra-rare variants. Moreover, a polygenic mode of inheritance is suspected in most cases. We performed SNP microarrays and whole exome sequencing in 20 families from Sudan, focusing on those with at least four affected members. Standard genetic filters and Endeavour algorithm for functional prioritization of genes selected likely susceptibility variants in FAT1, DCHS1 or ASTN2 genes. FAT1 and DCHS1 are adhesion transmembrane proteins interacting during brain development, while ASTN2 is involved in dendrite development. Our approach on familial forms of GGE is complementary to large-scale collaborative consortia studies of sporadic cases. Our study reinforces the hypothesis that GGE is genetically heterogeneous, even in a relatively limited geographic area, and mainly oligogenic, as supported by the low familial penetrance of GGE and by the Bayesian algorithm that we developed in a large pedigree with JME. Since populations with founder effect and endogamy are appropriate to study autosomal recessive pathologies, they would be also adapted to decipher genetic components of complex diseases, using the reported bayesian model.
Collapse
Affiliation(s)
- Maha Dahawi
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS - Hôpital La Pitié-Salpêtrière, Paris, France.
- Department of Physiology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan.
| | - Jean-Madeleine de Sainte Agathe
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS - Hôpital La Pitié-Salpêtrière, Paris, France
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
- Sorbonne Université, Paris, France
| | - Mohamed S Elmagzoub
- Faculty of Medicine, National Ribat University, Khartoum, Sudan
- Neuroscience Department, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Jubail, Saudi Arabia
| | - Elhami A Ahmed
- Faculty of Dentistry, Shendi University, Shendi, Sudan
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Julien Buratti
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
| | - Thomas Courtin
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS - Hôpital La Pitié-Salpêtrière, Paris, France
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
- Sorbonne Université, Paris, France
| | - Eric Noé
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS - Hôpital La Pitié-Salpêtrière, Paris, France
| | - Julie Bogoin
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
| | - Bruno Copin
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
| | | | - Wasma A Abdelgadir
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, Al-Neelain University, Khartoum, Sudan
| | - Ahmed K M A Ahmed
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mohamed A Daldoum
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
- Division of Neurology, Sudan Medical Council, Khartoum, Sudan
| | | | - Mohamed Bashir
- Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | | | - Sahar Gamil
- Department of Basic Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, AL-Kharj, Saudi Arabia
- Department of Biochemistry, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Sara Baldassari
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS - Hôpital La Pitié-Salpêtrière, Paris, France
| | - Liena Elsayed
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O.Box 84428, 11671, Riyadh, Saudi Arabia
| | - Boris Keren
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
| | - Gregory Nuel
- Stochastics and Biology Group (MAV), Probability and Statistics (LPSM, CNRS 8001), Sorbonne Université, Paris, France
| | - Ammar E Ahmed
- Department of Physiology, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Eric Leguern
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS - Hôpital La Pitié-Salpêtrière, Paris, France
- Department of Medical Genetics, Sorbonne Université, AP-HP Sorbonne Université, Paris, France
- Sorbonne Université, Paris, France
| |
Collapse
|
7
|
Zhou Z, Yang X, Mao A, Xu H, Lin C, Yang M, Hu W, Shao J, Xu P, Li Y, Li W, Lin R, Zhang R, Xie Q, Xu Z, Meng W. Deficiency of CAMSAP2 impairs olfaction and the morphogenesis of mitral cells. EMBO Rep 2024; 25:2861-2877. [PMID: 38839944 PMCID: PMC11239855 DOI: 10.1038/s44319-024-00166-x] [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: 10/06/2023] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 06/07/2024] Open
Abstract
In developing olfactory bulb (OB), mitral cells (MCs) remodel their dendrites to establish the precise olfactory circuit, and these circuits are critical for individuals to sense odors and elicit behaviors for survival. However, how microtubules (MTs) participate in the process of dendritic remodeling remains elusive. Here, we reveal that calmodulin-regulated spectrin-associated proteins (CAMSAPs), a family of proteins that bind to the minus-end of the noncentrosomal MTs, play a crucial part in the development of MC dendrites. We observed that Camsap2 knockout (KO) males are infertile while the reproductive tract is normal. Further study showed that the infertility was due to the severe defects of mating behavior in male mice. Besides, mice with loss-of-function displayed defects in the sense of smell. Furthermore, we found that the deficiency of CAMSAP2 impairs the classical morphology of MCs, and the CAMSAP2-dependent dendritic remodeling process is responsible for this defect. Thus, our findings demonstrate that CAMSAP2 plays a vital role in regulating the development of MCs.
Collapse
Affiliation(s)
- Zhengrong Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China.
- Neuroscience Center, Department of Basic Medical Sciences, Shantou University Medical College, 515041, Shantou, Guangdong, China.
| | - Xiaojuan Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Aihua Mao
- Biology Department, College of Sciences, Shantou University, 515063, Shantou, China
| | - Honglin Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Chunnuan Lin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Mengge Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Weichang Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jinhui Shao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Peipei Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yuejia Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Wenguang Li
- Animal Laboratory Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Ruifan Lin
- Chinese Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Rui Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Qi Xie
- Chinese Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Innovation Academy for Seed Design, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Innovation Academy for Seed Design, Chinese Academy of Sciences, 100101, Beijing, China.
| |
Collapse
|
8
|
Zhang R, Gu L, Chen W, Tanaka N, Zhou Z, Xu H, Xu T, Ji W, Liang X, Meng W. CAMSAP2 and CAMSAP3 localize at microtubule intersections to regulate the spatial distribution of microtubules. J Mol Cell Biol 2024; 15:mjad050. [PMID: 37567766 PMCID: PMC11156519 DOI: 10.1093/jmcb/mjad050] [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: 11/10/2022] [Revised: 03/14/2023] [Accepted: 08/10/2023] [Indexed: 08/13/2023] Open
Abstract
Microtubule networks support many cellular processes and exhibit a highly ordered architecture. However, due to the limited axial resolution of conventional light microscopy, the structural features of these networks cannot be resolved in three-dimensional (3D) space. Here, we used customized ultra-high-resolution interferometric single-molecule localization microscopy to characterize the microtubule networks in Caco2 cells. We found that the calmodulin-regulated spectrin-associated proteins (CAMSAPs) localize at a portion of microtubule intersections. Further investigation showed that depletion of CAMSAP2 and CAMSAP3 leads to the narrowing of the inter-microtubule distance. Mechanistically, CAMSAPs recognize microtubule defects, which often occur near microtubule intersections, and then recruit katanin to remove the damaged microtubules. Therefore, the CAMSAP-katanin complex is a regulatory module for the distance between microtubules. Taken together, our results characterize the architecture of cellular microtubule networks in high resolution and provide molecular insights into how the 3D structure of microtubule networks is controlled.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lusheng Gu
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Center of Optical Imaging and Detection Technology R&D, Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Wei Chen
- IDG/McGovern Institute for Brain Research, Tsinghua–Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Nobutoshi Tanaka
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology/RIKEN Center for Biosystems Dynamics Research, 650-0047 Kobe, Japan
| | - Zhengrong Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Honglin Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tao Xu
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Center of Optical Imaging and Detection Technology R&D, Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Wei Ji
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Innovation Center of Optical Imaging and Detection Technology R&D, Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510320, China
| | - Xin Liang
- IDG/McGovern Institute for Brain Research, Tsinghua–Peking Joint Center for Life Science, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
9
|
Qiao Y, Wang D, Yan G, Yang Z, Tang C. MiR-411-5p Promotes Vascular Smooth Muscle Cell Phenotype Switch by Inhibiting Expression of Calmodulin Regulated Spectrin-Associated Protein-1. Int Heart J 2024; 65:557-565. [PMID: 38825498 DOI: 10.1536/ihj.23-590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
When stimulated, vascular smooth muscle cells (VSMCs) change from a differentiated to a dedifferentiated phenotype. Dedifferentiated VSMCs have a key activity in cardiovascular diseases such as in-stent restenosis. MicroRNAs (miRNAs) have crucial functions in conversion of differentiated VSMCs to a dedifferentiated phenotype. We investigated the activity of miR-411-5p in the proliferation, migration, and phenotype switch of rat VSMCs.Based on a microRNA array assay, miR-411-5p expression was found to be significantly increased in cultured VSMCs stimulated by platelet-derived growth factor-BB (PDGF-BB). A CCK-8 assay, transwell assay, and scratch test were performed to measure the effect of miR-411-5p on the proliferation and migration of PDGF-BB-treated VSMCs. MiR-411-5p promoted expression of dedifferentiated phenotype markers such as osteopontin and tropomyosin 4 in PDGF-BB-treated VSMCs. Using mimics and inhibitors, we identified the target of miR-411-5p in PDGF-BB-treated VSMCs and found that calmodulin-regulated spectrin-associated protein-1 (CAMSAP1) was involved in the phenotypic switch mediated by PDGF-BB.By inhibiting expression of CAMSAP1, miR-411-5p enhanced the proliferation, migration, and phenotype switch of VSMCs.Blockade of miR-411-5p interaction with CAMSAP1 is a promising approach to treat in-stent restenosis.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Becaplermin/pharmacology
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Osteopontin/metabolism
- Osteopontin/genetics
- Phenotype
- Rats, Sprague-Dawley
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
Collapse
Affiliation(s)
- Yong Qiao
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School
| | | | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital of Southeast University Medical School
- Medical School of Southeast University
| |
Collapse
|
10
|
Santhanam A, Shihabeddin E, Wei H, Wu J, O'Brien J. Molecular basis of retinal remodeling in a zebrafish model of retinitis pigmentosa. Cell Mol Life Sci 2023; 80:362. [PMID: 37979052 PMCID: PMC10657301 DOI: 10.1007/s00018-023-05021-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/10/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
A hallmark of inherited retinal degenerative diseases such as retinitis pigmentosa (RP) is progressive structural and functional remodeling of the remaining retinal cells as photoreceptors degenerate. Extensive remodeling of the retina stands as a barrier for the successful implementation of strategies to restore vision. To understand the molecular basis of remodeling, we performed analyses of single-cell transcriptome data from adult zebrafish retina of wild type AB strain (WT) and a P23H mutant rhodopsin transgenic model of RP with continuous degeneration and regeneration. Retinas from both female and male fish were pooled to generate each library, combining data from both sexes. We provide a benchmark atlas of retinal cell type transcriptomes in zebrafish and insight into how each retinal cell type is affected in the P23H model. Oxidative stress is found throughout the retina, with increases in reliance on oxidative metabolism and glycolysis in the affected rods as well as cones, bipolar cells, and retinal ganglion cells. There is also transcriptional evidence for widespread synaptic remodeling and enhancement of glutamatergic transmission in the inner retina. Notably, changes in circadian rhythm regulation are detected in cones, bipolar cells, and retinal pigmented epithelium. We also identify the transcriptomic signatures of retinal progenitor cells and newly formed rods essential for the regenerative process. This comprehensive transcriptomic analysis provides a molecular road map to understand how the retina remodels in the context of chronic retinal degeneration with ongoing regeneration.
Collapse
Affiliation(s)
- Abirami Santhanam
- Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- University of Houston College of Optometry, Houston, TX, 77204, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Eyad Shihabeddin
- Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Haichao Wei
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jiaqian Wu
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - John O'Brien
- Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- University of Houston College of Optometry, Houston, TX, 77204, USA.
- MD Anderson UT Health Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
Hu W, Zhang R, Xu H, Li Y, Yang X, Zhou Z, Huang X, Wang Y, Ji W, Gao F, Meng W. CAMSAP1 role in orchestrating structure and dynamics of manchette microtubule minus-ends impacts male fertility during spermiogenesis. Proc Natl Acad Sci U S A 2023; 120:e2313787120. [PMID: 37903275 PMCID: PMC10636317 DOI: 10.1073/pnas.2313787120] [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: 08/23/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023] Open
Abstract
The manchette is a crucial transient structure involved in sperm development, with its composition and regulation still not fully understood. This study focused on investigating the roles of CAMSAP1 and CAMSAP2, microtubule (MT) minus-end binding proteins, in regulating manchette MTs, spermiogenesis, and male fertility. The loss of CAMSAP1, but not CAMSAP2, disrupts the well-orchestrated process of spermiogenesis, leading to abnormal manchette elongation and delayed removal, resulting in deformed sperm nuclei and tails resembling oligoasthenozoospermia symptoms. We investigated the underlying molecular mechanisms by purifying manchette assemblies and comparing them through proteomic analysis, and results showed that the absence of CAMSAP1 disrupted the proper localization of key proteins (CEP170 and KIF2A) at the manchette minus end, compromising its structural integrity and hindering MT depolymerization. These findings highlight the significance of maintaining homeostasis in manchette MT minus-ends for shaping manchette morphology during late spermiogenesis, offering insights into the molecular mechanisms underlying infertility and sperm abnormalities.
Collapse
Affiliation(s)
- Weichang Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Rui Zhang
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
| | - Honglin Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
| | - Yuejia Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Xiaojuan Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Zhengrong Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing100049, China
| | - Wei Ji
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing100101, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing100049, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong510320, China
| | - Fei Gao
- College of Life Science, University of Chinese Academy of Sciences, Beijing100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing100101, China
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing10019, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing100049, China
- Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing100101, China
| |
Collapse
|
12
|
Gujar MR, Gao Y, Teng X, Ding WY, Lin J, Tan YS, Chew LY, Toyama Y, Wang H. Patronin/CAMSAP promotes reactivation and regeneration of Drosophila quiescent neural stem cells. EMBO Rep 2023; 24:e56624. [PMID: 37440685 PMCID: PMC10481672 DOI: 10.15252/embr.202256624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/06/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The ability of stem cells to switch between quiescent and proliferative states is crucial for maintaining tissue homeostasis and regeneration. Drosophila quiescent neural stem cells (qNSCs) extend a primary protrusion that is enriched in acentrosomal microtubules and can be regenerated upon injury. Arf1 promotes microtubule growth, reactivation (exit from quiescence), and regeneration of qNSC protrusions upon injury. However, how Arf1 is regulated in qNSCs remains elusive. Here, we show that the microtubule minus-end binding protein Patronin/CAMSAP promotes acentrosomal microtubule growth and quiescent NSC reactivation. Patronin is important for the localization of Arf1 at Golgi and physically associates with Arf1, preferentially with its GDP-bound form. Patronin is also required for the regeneration of qNSC protrusion, likely via the regulation of microtubule growth. Finally, Patronin functions upstream of Arf1 and its effector Msps/XMAP215 to target the cell adhesion molecule E-cadherin to NSC-neuropil contact sites during NSC reactivation. Our findings reveal a novel link between Patronin/CAMSAP and Arf1 in the regulation of microtubule growth and NSC reactivation. A similar mechanism might apply to various microtubule-dependent systems in mammals.
Collapse
Affiliation(s)
- Mahekta R Gujar
- Neuroscience and Behavioral Disorders ProgrammeDuke‐NUS Medical SchoolSingaporeSingapore
| | - Yang Gao
- Neuroscience and Behavioral Disorders ProgrammeDuke‐NUS Medical SchoolSingaporeSingapore
| | - Xiang Teng
- Mechanobiology InstituteSingaporeSingapore
| | - Wei Yung Ding
- Neuroscience and Behavioral Disorders ProgrammeDuke‐NUS Medical SchoolSingaporeSingapore
| | - Jiaen Lin
- Neuroscience and Behavioral Disorders ProgrammeDuke‐NUS Medical SchoolSingaporeSingapore
| | - Ye Sing Tan
- Neuroscience and Behavioral Disorders ProgrammeDuke‐NUS Medical SchoolSingaporeSingapore
| | - Liang Yuh Chew
- Neuroscience and Behavioral Disorders ProgrammeDuke‐NUS Medical SchoolSingaporeSingapore
- Present address:
Temasek LifeSciences LaboratorySingaporeSingapore
| | - Yusuke Toyama
- Mechanobiology InstituteSingaporeSingapore
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Hongyan Wang
- Neuroscience and Behavioral Disorders ProgrammeDuke‐NUS Medical SchoolSingaporeSingapore
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Integrative Sciences and Engineering ProgrammeNational University of SingaporeSingaporeSingapore
| |
Collapse
|
13
|
Jaskiewicz K, Maleszka-Kurpiel M, Kabza M, Karolak JA, Gajecka M. Sequence variants contributing to dysregulated inflammatory responses across keratoconic cone surface in adolescent patients with keratoconus. Front Immunol 2023; 14:1197054. [PMID: 37483635 PMCID: PMC10359427 DOI: 10.3389/fimmu.2023.1197054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/09/2023] [Indexed: 07/25/2023] Open
Abstract
Background Keratoconus (KTCN) is the most common corneal ectasia resulting in a conical shape of the cornea. Here, genomic variation in the corneal epithelium (CE) across the keratoconic cone surface in patients with KTCN and its relevance in the functioning of the immune system were assessed. Methods Samples from four unrelated adolescent patients with KTCN and two control individuals were obtained during the CXL and PRK procedures, respectively. Three topographic regions, central, middle, and peripheral, were separated towards the whole-genome sequencing (WGS) study embracing a total of 18 experimental samples. The coding and non-coding sequence variation, including structural variation, was assessed and then evaluated together with the previously reported transcriptomic outcomes for the same CE samples and full-thickness corneas. Results First, pathway enrichment analysis of genes with identified coding variants pointed to "Antigen presentation" and "Interferon alpha/beta signaling" as the most overrepresented pathways, indicating the involvement of inflammatory responses in KTCN. Both coding and non-coding sequence variants were found in genes (or in their close proximity) linked to the previously revealed KTCN-specific cellular components, namely, "Actin cytoskeleton", "Extracellular matrix", "Collagen-containing extracellular matrix", "Focal adhesion", "Hippo signaling pathway", and "Wnt signaling" pathways. No genomic heterogeneity across the corneal surface was found comparing the assessed topographic regions. Thirty-five chromosomal regions enriched in both coding and non-coding KTCN-specific sequence variants were revealed, with a most representative 5q locus previously recognized as involved in KTCN. Conclusion The identified genomic features indicate the involvement of innate and adaptive immune system responses in KTCN pathogenesis.
Collapse
Affiliation(s)
| | - Magdalena Maleszka-Kurpiel
- Optegra Eye Health Care Clinic in Poznan, Poznan, Poland
- Chair of Ophthalmology and Optometry, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Kabza
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Justyna A. Karolak
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marzena Gajecka
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
14
|
Gu X, Jia C, Wang J. Advances in Understanding the Molecular Mechanisms of Neuronal Polarity. Mol Neurobiol 2023; 60:2851-2870. [PMID: 36738353 DOI: 10.1007/s12035-023-03242-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/22/2023] [Indexed: 02/05/2023]
Abstract
The establishment and maintenance of neuronal polarity are important for neural development and function. Abnormal neuronal polarity establishment commonly leads to a variety of neurodevelopmental disorders. Over the past three decades, with the continuous development and improvement of biological research methods and techniques, we have made tremendous progress in the understanding of the molecular mechanisms of neuronal polarity establishment. The activity of positive and negative feedback signals and actin waves are both essential in this process. They drive the directional transport and aggregation of key molecules of neuronal polarity, promote the spatiotemporal regulation of ordered and coordinated interactions of actin filaments and microtubules, stimulate the specialization and growth of axons, and inhibit the formation of multiple axons. In this review, we focus on recent advances in these areas, in particular the important findings about neuronal polarity in two classical models, in vitro primary hippocampal/cortical neurons and in vivo cortical pyramidal neurons, and discuss our current understanding of neuronal polarity..
Collapse
Affiliation(s)
- Xi Gu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Chunhong Jia
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Junhao Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
15
|
Fan Z, Wang S, Xu C, Yang J, Huang X, Xu H, Wang Y, Meng W, Cui B. Fu Fang Gang Liu aqueous extract inhibits the proliferation of HeLa cells by causing deoxyribonucleic acid damage. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:116083. [PMID: 36584921 DOI: 10.1016/j.jep.2022.116083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fu Fang Gang Liu (FFGL) is an effective formula for treating wart proliferation caused by human papillomavirus (HPV) infection and has the potential to treat HPV-related cancers. However, scientific evidence of its anti-tumor activity against cervical cancer, the most common cancer caused by HPV, is lacking. AIM OF THE STUDY To clarify the anti-tumor effect of an FFGL aqueous extract on human cervical cancer and its possible mechanism of cell cycle arrest in HeLa cells. MATERIALS AND METHODS The anti-proliferative effect of FFGL on cervical cancer cells was assessed using the cell counting kit-8 assay. The proportion of apoptotic cells, cell cycle distribution, and cell division rate were determined using flow cytometry. Quantitative proteomics was used to identify differentially expressed proteins after FFGL treatment, and bioinformatics analysis was used to identify key nodal proteins affected by FFGL. Immunofluorescence and western blot analyses were used to explore changes in the expression of related proteins in the cell cycle and DNA damage pathways to elucidate the potential mechanism of action of FFGL against HeLa cell proliferation. RESULTS FFGL inhibited cervical cancer cell proliferation and caused cell cycle arrest. According to quantitative proteomics, CyclinB1 may play an important role in the anti-proliferative effect of FFGL on HeLa cells. Additional experiments showed that FFGL aqueous extract caused ATM-mediated DNA damage, further phosphorylated CHK2, led to the inactivation of Cdc25C, inhibited the activity of the CDK1/CyclinB1 complex, and resulted in cell cycle arrest. CONCLUSIONS FFGL can inhibit cervical cancer cell proliferation. Furthermore, it can increase CDK1 phosphorylation, block the cell cycle by causing DNA damage, and inhibit HeLa cell proliferation.
Collapse
Affiliation(s)
- Zhu Fan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Postdoctoral Research Station, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Shuxin Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Chenchen Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Jiao Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Xiahe Huang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Honglin Xu
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yingchun Wang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wenxiang Meng
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bingnan Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
16
|
Katzenberger RJ, Ganetzky B, Wassarman DA. Lissencephaly-1 mutations enhance traumatic brain injury outcomes in Drosophila. Genetics 2023; 223:iyad008. [PMID: 36683334 PMCID: PMC9991514 DOI: 10.1093/genetics/iyad008] [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: 11/14/2022] [Revised: 11/14/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Traumatic brain injury (TBI) outcomes vary greatly among individuals, but most of the variation remains unexplained. Using a Drosophila melanogaster TBI model and 178 genetically diverse lines from the Drosophila Genetic Reference Panel (DGRP), we investigated the role that genetic variation plays in determining TBI outcomes. Following injury at 20-27 days old, DGRP lines varied considerably in mortality within 24 h ("early mortality"). Additionally, the disparity in early mortality resulting from injury at 20-27 vs 0-7 days old differed among DGRP lines. These data support a polygenic basis for differences in TBI outcomes, where some gene variants elicit their effects by acting on aging-related processes. Our genome-wide association study of DGRP lines identified associations between single nucleotide polymorphisms in Lissencephaly-1 (Lis-1) and Patronin and early mortality following injury at 20-27 days old. Lis-1 regulates dynein, a microtubule motor required for retrograde transport of many cargoes, and Patronin protects microtubule minus ends against depolymerization. While Patronin mutants did not affect early mortality, Lis-1 compound heterozygotes (Lis-1x/Lis-1y) had increased early mortality following injury at 20-27 or 0-7 days old compared with Lis-1 heterozygotes (Lis-1x/+), and flies that survived 24 h after injury had increased neurodegeneration but an unaltered lifespan, indicating that Lis-1 affects TBI outcomes independently of effects on aging. These data suggest that Lis-1 activity is required in the brain to ameliorate TBI outcomes through effects on axonal transport, microtubule stability, and other microtubule proteins, such as tau, implicated in chronic traumatic encephalopathy, a TBI-associated neurodegenerative disease in humans.
Collapse
Affiliation(s)
- Rebeccah J Katzenberger
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Barry Ganetzky
- Department of Genetics, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David A Wassarman
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
17
|
Khalaf-Nazzal R, Fasham J, Inskeep KA, Blizzard LE, Leslie JS, Wakeling MN, Ubeyratna N, Mitani T, Griffith JL, Baker W, Al-Hijawi F, Keough KC, Gezdirici A, Pena L, Spaeth CG, Turnpenny PD, Walsh JR, Ray R, Neilson A, Kouranova E, Cui X, Curiel DT, Pehlivan D, Akdemir ZC, Posey JE, Lupski JR, Dobyns WB, Stottmann RW, Crosby AH, Baple EL. Bi-allelic CAMSAP1 variants cause a clinically recognizable neuronal migration disorder. Am J Hum Genet 2022; 109:2068-2079. [PMID: 36283405 PMCID: PMC9674946 DOI: 10.1016/j.ajhg.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/27/2022] [Indexed: 01/26/2023] Open
Abstract
Non-centrosomal microtubules are essential cytoskeletal filaments that are important for neurite formation, axonal transport, and neuronal migration. They require stabilization by microtubule minus-end-targeting proteins including the CAMSAP family of molecules. Using exome sequencing on samples from five unrelated families, we show that bi-allelic CAMSAP1 loss-of-function variants cause a clinically recognizable, syndromic neuronal migration disorder. The cardinal clinical features of the syndrome include a characteristic craniofacial appearance, primary microcephaly, severe neurodevelopmental delay, cortical visual impairment, and seizures. The neuroradiological phenotype comprises a highly recognizable combination of classic lissencephaly with a posterior more severe than anterior gradient similar to PAFAH1B1(LIS1)-related lissencephaly and severe hypoplasia or absence of the corpus callosum; dysplasia of the basal ganglia, hippocampus, and midbrain; and cerebellar hypodysplasia, similar to the tubulinopathies, a group of monogenic tubulin-associated disorders of cortical dysgenesis. Neural cell rosette lineages derived from affected individuals displayed findings consistent with these phenotypes, including abnormal morphology, decreased cell proliferation, and neuronal differentiation. Camsap1-null mice displayed increased perinatal mortality, and RNAScope studies identified high expression levels in the brain throughout neurogenesis and in facial structures, consistent with the mouse and human neurodevelopmental and craniofacial phenotypes. Together our findings confirm a fundamental role of CAMSAP1 in neuronal migration and brain development and define bi-allelic variants as a cause of a clinically distinct neurodevelopmental disorder in humans and mice.
Collapse
Affiliation(s)
- Reham Khalaf-Nazzal
- Biomedical Sciences Department, Faculty of Medicine, Arab American University of Palestine, Jenin P227, Palestine
| | - James Fasham
- Department of Clinical and Biomedical Science, University of Exeter Faculty of Health and Life Science, RILD building, Barrack Road, Exeter EX2 5DW, UK; Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust (Heavitree Hospital), Gladstone Road, Exeter EX1 2ED, UK
| | - Katherine A Inskeep
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA; Institute for Genomic Medicine at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Lauren E Blizzard
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA
| | - Joseph S Leslie
- Department of Clinical and Biomedical Science, University of Exeter Faculty of Health and Life Science, RILD building, Barrack Road, Exeter EX2 5DW, UK
| | - Matthew N Wakeling
- Department of Clinical and Biomedical Science, University of Exeter Faculty of Health and Life Science, RILD building, Barrack Road, Exeter EX2 5DW, UK
| | - Nishanka Ubeyratna
- Department of Clinical and Biomedical Science, University of Exeter Faculty of Health and Life Science, RILD building, Barrack Road, Exeter EX2 5DW, UK
| | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer L Griffith
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wisam Baker
- Paediatrics Department, Dr. Khalil Suleiman Government Hospital, Jenin, Palestine
| | - Fida' Al-Hijawi
- Paediatrics Community Outpatient Clinics, Palestinian Ministry of Health, Jenin, Palestine
| | - Karen C Keough
- Department of Pediatrics, Dell Medical School, 1400 Barbara Jordan Boulevard, Austin, TX 78723, USA; Child Neurology Consultants of Austin, 7940 Shoal Creek Boulevard, Suite 100, Austin, TX 78757, USA
| | - Alper Gezdirici
- Department of Medical Genetics, Başakşehir Çam and Sakura City Hospital, 34480 Istanbul, Turkey
| | - Loren Pena
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Christine G Spaeth
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Peter D Turnpenny
- Department of Clinical and Biomedical Science, University of Exeter Faculty of Health and Life Science, RILD building, Barrack Road, Exeter EX2 5DW, UK; Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust (Heavitree Hospital), Gladstone Road, Exeter EX1 2ED, UK
| | - Joseph R Walsh
- Department of Neurological Surgery, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA
| | - Randall Ray
- Departments of Pediatrics and Medical Genetics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amber Neilson
- Genome Engineering & Stem Cell Center, Department of Genetics, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA
| | - Evguenia Kouranova
- Genome Engineering & Stem Cell Center, Department of Genetics, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA
| | - Xiaoxia Cui
- Genome Engineering & Stem Cell Center, Department of Genetics, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA
| | - David T Curiel
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in Saint Louis, St. Louis, MO 63130, USA; Division of Cancer Biology, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA; Biologic Therapeutics Center, Department of Radiation Oncology, School of Medicine, Washington University in Saint Louis, St. Louis, MO 63110, USA
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - Zeynep Coban Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| | - William B Dobyns
- Departments of Pediatrics and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Rolf W Stottmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA; Institute for Genomic Medicine at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH 43205, USA; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7016, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Andrew H Crosby
- Department of Clinical and Biomedical Science, University of Exeter Faculty of Health and Life Science, RILD building, Barrack Road, Exeter EX2 5DW, UK
| | - Emma L Baple
- Department of Clinical and Biomedical Science, University of Exeter Faculty of Health and Life Science, RILD building, Barrack Road, Exeter EX2 5DW, UK; Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust (Heavitree Hospital), Gladstone Road, Exeter EX1 2ED, UK.
| |
Collapse
|
18
|
He L, van Beem L, Snel B, Hoogenraad CC, Harterink M. PTRN-1 (CAMSAP) and NOCA-2 (NINEIN) are required for microtubule polarity in Caenorhabditis elegans dendrites. PLoS Biol 2022; 20:e3001855. [PMID: 36395330 PMCID: PMC9714909 DOI: 10.1371/journal.pbio.3001855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 12/01/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
The neuronal microtubule cytoskeleton is key to establish axon-dendrite polarity. Dendrites are characterized by the presence of minus-end out microtubules. However, the mechanisms that organize these microtubules with the correct orientation are still poorly understood. Using Caenorhabditis elegans as a model system for microtubule organization, we characterized the role of 2 microtubule minus-end related proteins in this process, the microtubule minus-end stabilizing protein calmodulin-regulated spectrin-associated protein (CAMSAP/PTRN-1), and the NINEIN homologue, NOCA-2 (noncentrosomal microtubule array). We found that CAMSAP and NINEIN function in parallel to mediate microtubule organization in dendrites. During dendrite outgrowth, RAB-11-positive vesicles localized to the dendrite tip to nucleate microtubules and function as a microtubule organizing center (MTOC). In the absence of either CAMSAP or NINEIN, we observed a low penetrance MTOC vesicles mislocalization to the cell body, and a nearly fully penetrant phenotype in double mutant animals. This suggests that both proteins are important for localizing the MTOC vesicles to the growing dendrite tip to organize microtubules minus-end out. Whereas NINEIN localizes to the MTOC vesicles where it is important for the recruitment of the microtubule nucleator γ-tubulin, CAMSAP localizes around the MTOC vesicles and is cotranslocated forward with the MTOC vesicles upon dendritic growth. Together, these results indicate that microtubule nucleation from the MTOC vesicles and microtubule stabilization are both important to localize the MTOC vesicles distally to organize dendritic microtubules minus-end out.
Collapse
Affiliation(s)
- Liu He
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Lotte van Beem
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Berend Snel
- Theoretical Biology and Bioinformatics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Casper C. Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
- Department of Neuroscience, Genentech, Inc., South San Francisco, California, United States of America
| | - Martin Harterink
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
19
|
Lin CH, Chen YC, Chan SP, Ou CY. TIAM-1 differentially regulates dendritic and axonal microtubule organization in patterning neuronal development through its multiple domains. PLoS Genet 2022; 18:e1010454. [PMID: 36223408 PMCID: PMC9612824 DOI: 10.1371/journal.pgen.1010454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 10/27/2022] [Accepted: 09/29/2022] [Indexed: 12/02/2022] Open
Abstract
Axon and dendrite development require the cooperation of actin and microtubule cytoskeletons. Microtubules form a well-organized network to direct polarized trafficking and support neuronal processes formation with distinct actin structures. However, it is largely unknown how cytoskeleton regulators differentially regulate microtubule organization in axon and dendrite development. Here, we characterize the role of actin regulators in axon and dendrite development and show that the RacGEF TIAM-1 regulates dendritic patterns through its N-terminal domains and suppresses axon growth through its C-terminal domains. TIAM-1 maintains plus-end-out microtubule orientation in posterior dendrites and prevents the accumulation of microtubules in the axon. In somatodendritic regions, TIAM-1 interacts with UNC-119 and stabilizes the organization between actin filaments and microtubules. UNC-119 is required for TIAM-1 to control axon growth, and its expression levels determine axon length. Taken together, TIAM-1 regulates neuronal microtubule organization and patterns axon and dendrite development respectively through its different domains.
Collapse
Affiliation(s)
- Chih-Hsien Lin
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ying-Chun Chen
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shih-Peng Chan
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chan-Yen Ou
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
20
|
Wang W, Zhang J, Wang Y, Xu Y, Zhang S. Non-coding ribonucleic acid-mediated CAMSAP1 upregulation leads to poor prognosis with suppressed immune infiltration in liver hepatocellular carcinoma. Front Genet 2022; 13:916847. [PMID: 36212130 PMCID: PMC9532701 DOI: 10.3389/fgene.2022.916847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Liver hepatocellular carcinoma (LIHC) is well-known for its unfavorable prognosis due to the lack of reliable diagnostic and prognostic biomarkers. Calmodulin-regulated spectrin-associated protein 1 (CAMSAP1) is a non-centrosomal microtubule minus-end binding protein that regulates microtubule dynamics. This study aims to investigate the specific role and mechanisms of CAMSAP1 in LIHC. We performed systematical analyses of CAMSAP1 and demonstrated that differential expression of CAMSAP1 is associated with genetic alteration and DNA methylation, and serves as a potential diagnostic and prognostic biomarker in some cancers, especially LIHC. Further evidence suggested that CAMSAP1 overexpression leads to adverse clinical outcomes in advanced LIHC. Moreover, the AC145207.5/LINC01748-miR-101–3p axis is specifically responsible for CAMSAP1 overexpression in LIHC. In addition to the previously reported functions in the cell cycle and regulation of actin cytoskeleton, CAMSAP1-related genes are enriched in cancer- and immune-associated pathways. As expected, CAMSAP1-associated LIHC is infiltrated in the suppressed immune microenvironment. Specifically, except for immune cell infiltration, it is significantly positively correlated with immune checkpoint genes, especially CD274 (PD-L1), and cancer-associated fibroblasts. Prediction of immune checkpoint blockade therapy suggests that these patients may benefit from therapy. Our study is the first to demonstrate that besides genetic alteration and DNA methylation, AC145207.5/LINC01748-miR-101-3p-mediated CAMSAP1 upregulation in advanced LIHC leads to poor prognosis with suppressed immune infiltration, representing a potential diagnostic and prognostic biomarker as well as a promising immunotherapy target for LIHC.
Collapse
|
21
|
Wan Z, Yang X, Liu X, Sun Y, Yu P, Xu F, Deng H. M2 macrophage-derived exosomal microRNA-411-5p impedes the activation of hepatic stellate cells by targeting CAMSAP1 in NASH model. iScience 2022; 25:104597. [PMID: 35789846 PMCID: PMC9249826 DOI: 10.1016/j.isci.2022.104597] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/29/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is a severe stage of nonalcoholic fatty liver disease (NAFLD), which is closely associated with the activation of hepatic stellate cells (HSCs) and their interaction with macrophages. Exosomes can mediate crosstalk between macrophages and HSCs in NAFLD-associated fibrosis. We found that M2 macrophage-derived exosomes significantly inhibit HSCs activation. RNA-seq studies revealed that miRNA-411-5p was decreased in serum exosomes of nonalcoholic steatohepatitis (NASH) patients as compared with that in healthy controls. Besides, miR-411-5p and M2 macrophage markers are decreased in the liver of the NASH model. We further proved that exosomal miR-411-5p from M2 macrophages inhibit HSCs activation and miR-411-5p directly downregulated the expression of Calmodulin-Regulated Spectrin-Associated Protein 1 (CAMSAP1) to inactivate stellate cells. Importantly, knockdown of CAMSAP1 also inhibited HSCs activation. This study contributes to understanding the underlying mechanism of HSCs activation and indicates CAMSAP1 may serve as a potential therapeutic target for NASH. M2 macrophage markers are decreased in the HFHCD-induced rat model of NASH M2 macrophage-derived exosomes inhibit HSCs activation via miR-411-5p CAMSAP1 is a direct target of miR-411-5p Knockdown of CAMSAP1 inhibits HSCs activation
Collapse
|
22
|
Kim J, Roh JD, Kim S, Kang H, Bae M, Kim E. Slc6a20a Heterozygous and Homozygous Mutant Mice Display Differential Behavioral and Transcriptomic Changes. Front Mol Neurosci 2022; 15:857820. [PMID: 35321029 PMCID: PMC8936588 DOI: 10.3389/fnmol.2022.857820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 01/28/2022] [Indexed: 11/23/2022] Open
Abstract
SLC6A20A is a proline and glycine transporter known to regulate glycine homeostasis and NMDA receptor (NMDAR) function in the brain. A previous study found increases in ambient glycine levels and NMDA receptor-mediated synaptic transmission in the brains of Slc6a20a-haploinsufficient mice, but it remained unknown whether Slc6a20a deficiency leads to disease-related behavioral deficits in mice. Here, we report that Slc6a20a heterozygous and homozygous mutant mice display differential behavioral phenotypes in locomotor, repetitive behavioral, and spatial and fear memory domains. In addition, these mice show differential transcriptomic changes in synapse, ribosome, mitochondria, autism, epilepsy, and neuron-related genes. These results suggest that heterozygous and homozygous Slc6a20a deletions in mice lead to differential changes in behaviors and transcriptomes.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Junyeop Daniel Roh
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Seongbin Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Mihyun Bae
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
- *Correspondence: Eunjoon Kim,
| |
Collapse
|
23
|
Wdr47, Camsaps, and Katanin cooperate to generate ciliary central microtubules. Nat Commun 2021; 12:5796. [PMID: 34608154 PMCID: PMC8490363 DOI: 10.1038/s41467-021-26058-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/10/2021] [Indexed: 02/08/2023] Open
Abstract
The axonemal central pair (CP) are non-centrosomal microtubules critical for planar ciliary beat. How they form, however, is poorly understood. Here, we show that mammalian CP formation requires Wdr47, Camsaps, and microtubule-severing activity of Katanin. Katanin severs peripheral microtubules to produce central microtubule seeds in nascent cilia. Camsaps stabilize minus ends of the seeds to facilitate microtubule outgrowth, whereas Wdr47 concentrates Camsaps into the axonemal central lumen to properly position central microtubules. Wdr47 deficiency in mouse multicilia results in complete loss of CP, rotatory beat, and primary ciliary dyskinesia. Overexpression of Camsaps or their microtubule-binding regions induces central microtubules in Wdr47-/- ependymal cells but at the expense of low efficiency, abnormal numbers, and wrong location. Katanin levels and activity also impact the central microtubule number. We propose that Wdr47, Camsaps, and Katanin function together for the generation of non-centrosomal microtubule arrays in polarized subcellular compartments.
Collapse
|
24
|
Buijs RR, Hummel JJA, Burute M, Pan X, Cao Y, Stucchi R, Altelaar M, Akhmanova A, Kapitein LC, Hoogenraad CC. WDR47 protects neuronal microtubule minus ends from katanin-mediated severing. Cell Rep 2021; 36:109371. [PMID: 34260930 DOI: 10.1016/j.celrep.2021.109371] [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: 09/23/2020] [Revised: 02/17/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023] Open
Abstract
Axons and dendrites are long extensions of neurons that contain arrays of noncentrosomal microtubules. Calmodulin-regulated spectrin-associated proteins (CAMSAPs) bind to and stabilize free microtubule minus ends and are critical for proper neuronal development and function. Previous studies have shown that the microtubule-severing ATPase katanin interacts with CAMSAPs and limits the length of CAMSAP-decorated microtubule stretches. However, how CAMSAP and microtubule minus end dynamics are regulated in neurons is poorly understood. Here, we show that the neuron-enriched protein WDR47 interacts with CAMSAPs and is critical for axon and dendrite development. We find that WDR47 accumulates at CAMSAP2-decorated microtubules, is essential for maintaining CAMSAP2 stretches, and protects minus ends from katanin-mediated severing. We propose a model where WDR47 protects CAMSAP2 at microtubule minus ends from katanin activity to ensure proper stabilization of the neuronal microtubule network.
Collapse
Affiliation(s)
- Robin R Buijs
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | - Jessica J A Hummel
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | - Mithila Burute
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | - Xingxiu Pan
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | - Yujie Cao
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | - Riccardo Stucchi
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands; Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, the Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, the Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, the Netherlands; Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|