1
|
Wei L, Hu S, Gong X, Ahemaiti Y, Li D, Ouyang S, Huang Y, Wang Y, Liang Y, Deng Y, Liu L, Zhao T. Disrupted maxillofacial, cardiovascular, and nervous development in washc5 knockout Zebrafish: Insights into 3C syndrome. Gene 2025; 948:149351. [PMID: 39988189 DOI: 10.1016/j.gene.2025.149351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
3C syndrome features craniofacial, nervous, and cardiovascular malformations. WASHC5 gene mutations may underline this syndrome, but the pathogenicity and underlying mechanism remain undetermined. We analyzed the expression pattern of the washc5 gene in zebrafish using whole-body in situ hybridization and generated a zebrafish model with washc5 gene knockout using CRISPR/Cas9 technology. Homozygous zebrafish exhibited high mortality, retarded growth, lighter stripes, and reduced pigmentation around the pupils. In the maxillofacial region, homozygotes displayed a shortened and tilted maxilla and delayed ossification of bones. In the heart, homozygous zebrafish showed a decreased heart rate, increased ventricular area, disorganized ventricular muscle fibers, mitochondrial swelling, Golgi lysis, and endoplasmic reticulum (ER) lysis in ventricular myocytes. The mRNA levels of nppb and myh7 were significantly increased. In the nervous system, homozygotes displayed bradykinesia and impaired neuronal development. qRT-PCR analysis revealed downregulation of col1a2, col1a1a, col1a1b, sp7, and msx2b (osteogenic factors and regulators of maxillofacial skeletal development) and abnormal expression of alpk2, alpk3b, actc2 (cardiac development factors), as well as tsen54, exosc8, and exosc9 (cerebellar development factors). Enrichment analysis of differentially expressed genes and proteins indicated involvement in ER-related processes. The washc5 knockout zebrafish model exhibits phenotypic similarities to human 3C syndrome, suggesting that mutations of this gene may play a pathogenic role in the syndrome. The mechanism of the washc5 gene in 3C syndrome may be associated with disturbances in ER homeostasis, providing insights into potential gene therapy strategies.
Collapse
Affiliation(s)
- Luyao Wei
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Shijun Hu
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Xueyang Gong
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Yiliya Ahemaiti
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Diwen Li
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Shi Ouyang
- Laboratory of Zebrafish Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081 Hunan, China
| | - Yuyang Huang
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Yongyi Wang
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Yan Liang
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Yun Deng
- Laboratory of Zebrafish Genetics, College of Life Sciences, Hunan Normal University, Changsha 410081 Hunan, China
| | - Lin Liu
- Department of Stomatology, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China
| | - Tianli Zhao
- Department of Cardiovascular Surgery, Second Xiangya Hospital, Central South University, Changsha 410011 Hunan, China.
| |
Collapse
|
2
|
Park H, Heo H, Song Y, Lee MS, Cho Y, Lee JS, Chang J, Lee S. TRIM22 functions as a scaffold protein for autophagy initiation. Anim Cells Syst (Seoul) 2025; 29:296-311. [PMID: 40337095 PMCID: PMC12057787 DOI: 10.1080/19768354.2025.2498926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 04/03/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Tripartite motif (TRIM) family proteins are increasingly recognized as important regulators of autophagy under various physiological and pathological conditions. TRIM22 has been previously shown to mediate autophagosome-lysosome fusion, but its potential role in earlier stages of autophagy remained unexplored. In this study, we investigated the function of TRIM22 in autophagy initiation. Overexpression of TRIM22 increased LC3-II levels and enhanced autophagic flux without affecting mTOR and AMPK activity. We found that TRIM22 interacts with components of both the ULK1 complex and the class III PI3K complex through distinct domains, recruiting them into punctate structures that represent autophagosome formation sites. Domain mapping revealed that the SPRY domain mediates interactions with ATG13 and FIP200, while the N-terminal region interacts with ULK1 and ATG101. The B-box domain of TRIM22 was identified as crucial for its interaction with Beclin-1, a key component of the class III PI3K complex. Deletion of this domain impaired the ability of TRIM22 to assemble the class III PI3K complex and induce autophagic flux. Interestingly, competitive binding assays revealed that Beclin-1 and PLEKHM1 bind to the same region of TRIM22, suggesting a mechanism for coordinating different stages of autophagy. The Alzheimer's disease-associated TRIM22 variant R321K maintained autophagy initiation function in both cell lines and primary neurons. These findings demonstrate that TRIM22 acts as a scaffold protein to promote autophagy initiation, in addition to its previously described role in autophagosome-lysosome fusion. Our study provides new insights into the molecular mechanisms by which TRIM proteins regulate multiple stages of the autophagy process.
Collapse
Affiliation(s)
- Hyungsun Park
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Hansol Heo
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yeongseo Song
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Myung Shin Lee
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yebin Cho
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jae-Seon Lee
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Jaerak Chang
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seongju Lee
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
- Department of Anatomy, College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
3
|
Park H, Park J, Kim T, Heo H, Chang J, Blackstone C, Lee S. A depression-associated protein FKBP5 functions in autophagy initiation through scaffolding the VPS34 complex. Mol Neurobiol 2025:10.1007/s12035-025-04897-3. [PMID: 40175715 DOI: 10.1007/s12035-025-04897-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Common variants in the FKBP5 gene have been implicated in recurrence of major depressive disorder (MDD) and response to antidepressant treatment. Although the relationship between FKBP5 and MDD has been revealed through several studies, the detailed molecular mechanisms by which FKBP5 regulates responsiveness to antidepressants have not been fully understood. Here, we aimed to elucidate the molecular mechanisms of FKBP5 in autophagy initiation and its potential role in the antidepressant response. We found that FKBP5 deficiency impaired the initiation of basal and stress-induced autophagy, accompanied by reduced protein levels of the PIK3C3/VPS34 complex, which is essential for autophagy initiation. Mechanistically, we demonstrated that FKBP5 physically binds to the VPS34 complex components, facilitating their assembly and subsequent autophagy initiation. Particularly, our study revealed that FKBP5 mediates antidepressant-induced autophagy by promoting the VPS34 complex assembly. These findings were consistent in neuronal cells, where FKBP5 depletion resulted in decreased autophagy and impaired the VPS34 complex assembly. Understanding the interplay between FKBP5, autophagy, and MDD may provide new insights into more effective treatments for MDD and related disorders.
Collapse
Affiliation(s)
- Hyungsun Park
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Jisoo Park
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Taewan Kim
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Hansol Heo
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jaerak Chang
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Craig Blackstone
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Seongju Lee
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea.
| |
Collapse
|
4
|
Heo H, Park H, Lee MS, Kim J, Kim J, Jung SY, Kim SK, Lee S, Chang J. TRIM22 facilitates autophagosome-lysosome fusion by mediating the association of GABARAPs and PLEKHM1. Autophagy 2024; 20:1098-1113. [PMID: 38009729 PMCID: PMC11135824 DOI: 10.1080/15548627.2023.2287925] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
Tripartite motif (TRIM) proteins are a large family of E3 ubiquitin ligases implicated in antiviral defense systems, tumorigenesis, and protein quality control. TRIM proteins contribute to protein quality control by regulating the ubiquitin-proteasome system, endoplasmic reticulum-associated degradation, and macroautophagy/autophagy. However, the detailed mechanisms through which various TRIM proteins regulate downstream events have not yet been fully elucidated. Herein, we identified a novel function of TRIM22 in the regulation of autophagy. TRIM22 promotes autophagosome-lysosome fusion by mediating the association of GABARAP family proteins with PLEKHM1, thereby inducing the autophagic clearance of protein aggregates, independent of its E3 ubiquitin ligase activity. Furthermore, a TRIM22 variant associated with early-onset familial Alzheimer disease interferes with autophagosome-lysosome fusion and autophagic clearance. These findings suggest TRIM22 as a critical autophagic regulator that orchestrates autophagosome-lysosome fusion by scaffolding autophagy-related proteins, thus representing a potential therapeutic target in neurodegenerative diseases.Abbreviations: AD: Alzheimer disease; ADAOO: AD age of onset; AICD: APP intracellular domain; APP: amyloid beta precursor protein; BSA: bovine serum albumin; cDNAs: complementary DNAs; CQ: chloroquine; CTF: carboxyl-terminal fragment; EBSS: Earle's balanced salt solution; GABARAP: GABA type A receptor-associated protein; GST: glutathione S-transferase; HA: hemagglutinin; HOPS: homotypic fusion and protein sorting; IFN: interferon; IL1A/IL-1α: interleukin 1 alpha; KO: knockout; MTORC1: mechanistic target of rapamycin kinase complex 1; NFKBIA/IκBα: NFKB inhibitor alpha; NFE2L2/NRF2: NFE2 like bZIP transcription factor; PBS: phosphate-buffered saline; PI3K: class I phosphoinositide 3-kinase; PLA: proximity ligation assay; PLEKHM1: pleckstrin homology and RUN domain containing M1; PSEN1: presenilin 1; SEM: standard errors of the means; SNAREs: soluble N-ethylmaleimide-sensitive factor attachment protein receptors; SNCA: synuclein alpha; SNP: single nucleotide polymorphism; TBS: tris-buffered saline; TNF/TNF-α: tumor necrosis factor; TRIM: tripartite motif; ULK1: unc-51 like autophagy activating kinase 1; WT: wild-type.
Collapse
Affiliation(s)
- Hansol Heo
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyungsun Park
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Myung Shin Lee
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jongyoon Kim
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Juyeong Kim
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Soon-Young Jung
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sun Kyeon Kim
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Seongju Lee
- Department of Anatomy, College of Medicine, and Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Jaerak Chang
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
5
|
Shiraishi T, Bono K, Hiraki H, Manome Y, Oka H, Iguchi Y, Okano HJ. The impact of VPS35 D620N mutation on alternative autophagy and its reversal by estrogen in Parkinson's disease. Cell Mol Life Sci 2024; 81:103. [PMID: 38409392 PMCID: PMC10896810 DOI: 10.1007/s00018-024-05123-4] [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: 10/17/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/28/2024]
Abstract
VPS35 plays a key role in neurodegenerative processes in Alzheimer's disease and Parkinson's disease (PD). Many genetic studies have shown a close relationship between autophagy and PD pathophysiology, and specifically, the PD-causing D620N mutation in VPS35 has been shown to impair autophagy. However, the molecular mechanisms underlying neuronal cell death and impaired autophagy in PD are debated. Notably, increasing evidence suggests that Rab9-dependent "alternative" autophagy, which is driven by a different molecular mechanism that driving ATG5-dependent "conventional" autophagy, also contributes to neurodegenerative process. In this study, we investigated the relationship between alternative autophagy and VPS35 D620N mutant-related PD pathogenesis. We isolated iPSCs from the blood mononuclear cell population of two PD patients carrying the VPS35 D620N mutant. In addition, we used CRISPR-Cas9 to generate SH-SY5Y cells carrying the D620N variant of VPS35. We first revealed that the number of autophagic vacuoles was significantly decreased in ATG5-knockout Mouse Embryonic Fibroblast or ATG5-knockdown patient-derived dopaminergic neurons carrying the VPS35 D620N mutant compared with that of the wild type VPS35 control cells. Furthermore, estrogen, which activates alternative autophagy pathways, increased the number of autophagic vacuoles in ATG5-knockdown VPS35 D620N mutant dopaminergic neurons. Estrogen induces Rab9 phosphorylation, mediated through Ulk1 phosphorylation, ultimately regulating alternative autophagy. Moreover, estrogen reduced the apoptosis rate of VPS35 D620N neurons, and this effect of estrogen was diminished under alternative autophagy knockdown conditions. In conclusion, alternative autophagy might be important for maintaining neuronal homeostasis and may be associated with the neuroprotective effect of estrogen in PD with VPS35 D620N.
Collapse
Affiliation(s)
- Tomotaka Shiraishi
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Keiko Bono
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Hiromi Hiraki
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Yoko Manome
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan
| | - Hisayoshi Oka
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 105‑8461, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3‑25‑8 Nishi‑Shinbashi, Minato‑ku, Tokyo, 1058461, Japan.
| |
Collapse
|
6
|
Gao SY, Liu YX, Dong Y, Fan LL, Ding Q, Liu L. Case report: A novel WASHC5 variant altering mRNA splicing causes spastic paraplegia in a patient. Front Genet 2023; 14:1205052. [PMID: 38028608 PMCID: PMC10644772 DOI: 10.3389/fgene.2023.1205052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Hereditary spastic paraplegia (HSP) is a progressive upper-motor neurodegenerative disease. Mutations in the WASHC5 gene are associated with autosomal dominant HSP, spastic paraplegia 8 (SPG8). However, due to the small number of reported cases, the exact mechanism remains unclear. Method: We report a Chinese family with HSP. The proband was referred to our hospital due to restless leg syndrome and insomnia. The preliminary clinical diagnosis of the proband was spastic paraplegia. Whole-exome sequencing (WES) and RNA splicing analysis were conducted to evaluate the genetic cause of the disease in this family. Results: A novel splice-altering variant (c.712-2A>G) in the WASHC5 gene was detected and further verified by RNA splicing analysis and Sanger sequencing. Real-time qPCR analysis showed that the expression of genes involved in the Wiskott-Aldrich syndrome protein and SCAR homolog (WASH) complex and endosomal and lysosomal systems was altered due to this variant. Conclusion: A novel heterozygous splice-altering variant (c.712-2A>G) in the WASHC5 gene was detected in a Chinese family with HSP. Our study provided data for genetic counseling to this family and offered evidence that this splicing variant in the WASHC5 gene is significant in causing HSP.
Collapse
Affiliation(s)
- Shan-Yu Gao
- Department of Neurology, Changshu No. 2 People’s Hospital, Changshu, China
- Department of Cell Biology, School of Life Science, Central South University, Changsha, China
- Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Xing Liu
- Department of Cell Biology, School of Life Science, Central South University, Changsha, China
- Department of Nephrology, Xiangya Hospital Central South University, Changsha, China
| | - Yi Dong
- Department of Cell Biology, School of Life Science, Central South University, Changsha, China
| | - Liang-Liang Fan
- Department of Cell Biology, School of Life Science, Central South University, Changsha, China
| | - Qi Ding
- Department of Neurology, Changshu No. 2 People’s Hospital, Changshu, China
| | - Lv Liu
- Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
7
|
Martinello C, Panza E, Orlacchio A. Hereditary spastic paraplegias proteome: common pathways and pathogenetic mechanisms. Expert Rev Proteomics 2023; 20:171-188. [PMID: 37788157 DOI: 10.1080/14789450.2023.2260952] [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: 06/12/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
INTRODUCTION Hereditary spastic paraplegias (HSPs) are a group of inherited neurodegenerative disorders characterized by progressive spasticity and weakness of the lower limbs. These conditions are caused by lesions in the neuronal pyramidal tract and exhibit clinical and genetic variability. Ongoing research focuses on understanding the underlying mechanisms of HSP onset, which ultimately lead to neuronal degeneration. Key molecular mechanisms involved include axonal transport, cytoskeleton dynamics, myelination abnormalities, membrane trafficking, organelle morphogenesis, ER homeostasis, mitochondrial dysfunction, and autophagy deregulation. AREAS COVERED This review aims to provide an overview of the shared pathogenetic mechanisms in various forms of HSPs. By examining disease-causing gene products and their associated functional pathways, this understanding could lead to the discovery of new therapeutic targets and the development of treatments to modify the progression of the disease. EXPERT OPINION Investigating gene functionality is crucial for identifying shared pathogenetic pathways underlying different HSP subtypes. Categorizing protein function and identifying pathways aids in finding biomarkers, predicting early onset, and guiding treatment for a better quality of life. Targeting shared mechanisms enables efficient and cost-effective therapies. Prospects involve identifying new disease-causing genes, refining molecular processes, and implementing findings in diagnosis, key for advancing HSP understanding and developing effective treatments.
Collapse
Affiliation(s)
- Chiara Martinello
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Emanuele Panza
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
- Unità di Genetica Medica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Antonio Orlacchio
- Laboratorio di Neurogenetica, Centro Europeo di Ricerca sul Cervello (CERC), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| |
Collapse
|
8
|
Lim SH, Shin S, Lee NY, Min SS, Kim NS, Lee DY, Lee JR. Strumpellin/WASHC5 regulates the structural plasticity of cortical neurons involved in gait coordination. Biochem Biophys Res Commun 2023; 673:169-174. [PMID: 37392480 DOI: 10.1016/j.bbrc.2023.06.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Strumpellin/Wiskott-Aldrich syndrome protein and SCAR homologue (WASH) complex subunit 5 (WASHC5) is a core component of the WASH complex, and its mutations confer pathogenicity for hereditary spastic paraplegia (HSP) type SPG8, a rare neurodegenerative gait disorder. WASH complex activates actin-related protein-2/3-mediated actin polymerization and plays a pivotal role in intracellular membrane trafficking in endosomes. In this study, we examined the role of strumpellin in the regulation of structural plasticity of cortical neurons involved in gait coordination. Administration of a lentivirus containing a strumpellin-targeting short hairpin RNA (shRNA) to cortical motor neurons lead to abnormal motor coordination in mice. Strumpellin knockdown using shRNA attenuated dendritic arborization and synapse formation in cultured cortical neurons, and this effect was rescued by wild-type strumpellin expression. Compared with the wild-type, strumpellin mutants N471D or V626F identified in patients with SPG8 exhibited no differences in rescuing the defects. Moreover, the number of F-actin clusters in neuronal dendrites was decreased by strumpellin knockdown and rescued by strumpellin expression. In conclusion, our results indicate that strumpellin regulates the structural plasticity of cortical neurons via actin polymerization.
Collapse
Affiliation(s)
- So-Hee Lim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea
| | - Sangyep Shin
- Department of Physiology and Biophysics, School of Medicine, Eulji University, Daejeon, 34824, South Korea
| | - Na-Yoon Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea; Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, South Korea
| | - Sun Seek Min
- Department of Physiology and Biophysics, School of Medicine, Eulji University, Daejeon, 34824, South Korea
| | - Nam-Soon Kim
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea
| | - Da Yong Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea.
| | - Jae-Ran Lee
- Rare Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea.
| |
Collapse
|
9
|
Schurr Y, Reil L, Spindler M, Nieswandt B, Machesky LM, Bender M. The WASH-complex subunit Strumpellin regulates integrin αIIbβ3 trafficking in murine platelets. Sci Rep 2023; 13:9526. [PMID: 37308549 PMCID: PMC10260982 DOI: 10.1038/s41598-023-36387-8] [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: 02/03/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023] Open
Abstract
The platelet specific integrin αIIbβ3 mediates platelet adhesion, aggregation and plays a central role in thrombosis and hemostasis. In resting platelets, αIIbβ3 is expressed on the membrane surface and in intracellular compartments. Upon activation, the number of surface-expressed αIIbβ3 is increased by the translocation of internal granule pools to the plasma membrane. The WASH complex is the major endosomal actin polymerization-promoting complex and has been implicated in the generation of actin networks involved in endocytic trafficking of integrins in other cell types. The role of the WASH complex and its subunit Strumpellin in platelet function is still unknown. Here, we report that Strumpellin-deficient murine platelets display an approximately 20% reduction in integrin αIIbβ3 surface expression. While exposure of the internal αIIbβ3 pool after platelet activation was unaffected, the uptake of the αIIbβ3 ligand fibrinogen was delayed. The number of platelet α-granules was slightly but significantly increased in Strumpellin-deficient platelets. Quantitative proteome analysis of isolated αIIbβ3-positive vesicular structures revealed an enrichment of protein markers, which are associated with the endoplasmic reticulum, Golgi complex and early endosomes in Strumpellin-deficient platelets. These results point to a so far unidentified role of the WASH complex subunit Strumpellin in integrin αIIbβ3 trafficking in murine platelets.
Collapse
Affiliation(s)
- Yvonne Schurr
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Lucy Reil
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Markus Spindler
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Laura M Machesky
- Department of Biochemistry, University of Cambridge, Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Markus Bender
- Institute of Experimental Biomedicine-Chair I, University Hospital and Rudolf Virchow Center, Josef-Schneider-Str. 2, 97080, Würzburg, Germany.
| |
Collapse
|
10
|
Mahale R, Arunachal G, Davuluri A, Padmanabha H, Mailankody P. Spastic Paraplegia Type 8: A First Report from India. Ann Indian Acad Neurol 2023; 26:189-190. [PMID: 37179679 PMCID: PMC10170995 DOI: 10.4103/aian.aian_901_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Rohan Mahale
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Gautam Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Anudeep Davuluri
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Hansashree Padmanabha
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Pooja Mailankody
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
11
|
Kramer DA, Piper HK, Chen B. WASP family proteins: Molecular mechanisms and implications in human disease. Eur J Cell Biol 2022; 101:151244. [PMID: 35667337 PMCID: PMC9357188 DOI: 10.1016/j.ejcb.2022.151244] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 02/08/2023] Open
Abstract
Proteins of the Wiskott-Aldrich syndrome protein (WASP) family play a central role in regulating actin cytoskeletal dynamics in a wide range of cellular processes. Genetic mutations or misregulation of these proteins are tightly associated with many diseases. The WASP-family proteins act by transmitting various upstream signals to their conserved WH2-Central-Acidic (WCA) peptide sequence at the C-terminus, which in turn binds to the Arp2/3 complex to stimulate the formation of branched actin networks at membranes. Despite this common feature, the regulatory mechanisms and cellular functions of distinct WASP-family proteins are very different. Here, we summarize and clarify our current understanding of WASP-family proteins and how disruption of their functions is related to human disease.
Collapse
Affiliation(s)
- Daniel A Kramer
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Hannah K Piper
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA
| | - Baoyu Chen
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, 2437 Pammel Drive, Ames, IA 50011, USA.
| |
Collapse
|
12
|
Retromer dependent changes in cellular homeostasis and Parkinson's disease. Essays Biochem 2021; 65:987-998. [PMID: 34528672 PMCID: PMC8709886 DOI: 10.1042/ebc20210023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/18/2022]
Abstract
To date, mechanistic treatments targeting the initial cause of Parkinson's disease (PD) are limited due to the underlying biological cause(s) been unclear. Endosomes and their associated cellular homeostasis processes have emerged to have a significant role in the pathophysiology associated with PD. Several variants within retromer complex have been identified and characterised within familial PD patients. The retromer complex represents a key sorting platform within the endosomal system that regulates cargo sorting that maintains cellular homeostasis. In this review, we summarise the current understandings of how PD-associated retromer variants disrupt cellular trafficking and how the retromer complex can interact with other PD-associated genes to contribute to the disease progression.
Collapse
|
13
|
Clemen CS, Schmidt A, Winter L, Canneva F, Wittig I, Becker L, Coras R, Berwanger C, Hofmann A, Eggers B, Marcus K, Gailus-Durner V, Fuchs H, de Angelis MH, Krüger M, von Hörsten S, Eichinger L, Schröder R. N471D WASH complex subunit strumpellin knock-in mice display mild motor and cardiac abnormalities and BPTF and KLHL11 dysregulation in brain tissue. Neuropathol Appl Neurobiol 2021; 48:e12750. [PMID: 34312900 DOI: 10.1111/nan.12750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/12/2021] [Indexed: 11/30/2022]
Abstract
AIMS We investigated N471D WASH complex subunit strumpellin (Washc5) knock-in and Washc5 knock-out mice as models for hereditary spastic paraplegia type 8 (SPG8). METHODS We generated heterozygous and homozygous N471D Washc5 knock-in mice and subjected them to a comprehensive clinical, morphological and laboratory parameter screen, and gait analyses. Brain tissue was used for proteomic analysis. Furthermore, we generated heterozygous Washc5 knock-out mice. WASH complex subunit strumpellin expression was determined by qPCR and immunoblotting. RESULTS Homozygous N471D Washc5 knock-in mice showed mild dilated cardiomyopathy, decreased acoustic startle reactivity, thinner eye lenses, increased alkaline phosphatase and potassium levels and increased white blood cell counts. Gait analyses revealed multiple aberrations indicative of locomotor instability. Similarly, the clinical chemistry, haematology and gait parameters of heterozygous mice also deviated from the values expected for healthy animals, albeit to a lesser extent. Proteomic analysis of brain tissue depicted consistent upregulation of BPTF and downregulation of KLHL11 in heterozygous and homozygous knock-in mice. WASHC5-related protein interaction partners and complexes showed no change in abundancies. Heterozygous Washc5 knock-out mice showing normal WASHC5 levels could not be bred to homozygosity. CONCLUSIONS While biallelic ablation of Washc5 was prenatally lethal, expression of N471D mutated WASHC5 led to several mild clinical and laboratory parameter abnormalities, but not to a typical SPG8 phenotype. The consistent upregulation of BPTF and downregulation of KLHL11 suggest mechanistic links between the expression of N471D mutated WASHC5 and the roles of both proteins in neurodegeneration and protein quality control, respectively.
Collapse
Affiliation(s)
- Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany.,Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany.,Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andreas Schmidt
- Center for Molecular Medicine and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Cologne, Germany
| | - Lilli Winter
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Fabio Canneva
- Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Center, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ilka Wittig
- Functional Proteomics, Medical School, Goethe University, Frankfurt, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Roland Coras
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Carolin Berwanger
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | | | - Andreas Hofmann
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Britta Eggers
- Medical Proteome Center, Medical Faculty, and Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Katrin Marcus
- Medical Proteome Center, Medical Faculty, and Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,TUM School of Life Sciences (SoLS), Technical University of Munich, Freising, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus Krüger
- Center for Molecular Medicine and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Cologne, Germany
| | - Stephan von Hörsten
- Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Center, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ludwig Eichinger
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
14
|
Toupenet Marchesi L, Leblanc M, Stevanin G. Current Knowledge of Endolysosomal and Autophagy Defects in Hereditary Spastic Paraplegia. Cells 2021; 10:cells10071678. [PMID: 34359848 PMCID: PMC8307360 DOI: 10.3390/cells10071678] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 12/25/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) refers to a group of neurological disorders involving the degeneration of motor neurons. Due to their clinical and genetic heterogeneity, finding common effective therapeutics is difficult. Therefore, a better understanding of the common pathological mechanisms is necessary. The role of several HSP genes/proteins is linked to the endolysosomal and autophagic pathways, suggesting a functional convergence. Furthermore, impairment of these pathways is particularly interesting since it has been linked to other neurodegenerative diseases, which would suggest that the nervous system is particularly sensitive to the disruption of the endolysosomal and autophagic systems. In this review, we will summarize the involvement of HSP proteins in the endolysosomal and autophagic pathways in order to clarify their functioning and decipher some of the pathological mechanisms leading to HSP.
Collapse
Affiliation(s)
- Liriopé Toupenet Marchesi
- Institut du Cerveau—Paris Brain Institute—ICM, INSERM, CNRS, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 75013 Paris, France; (L.T.M.); (M.L.)
- Neurogenetics Team, EPHE, Paris Sciences Lettres Research University, 75000 Paris, France
| | - Marion Leblanc
- Institut du Cerveau—Paris Brain Institute—ICM, INSERM, CNRS, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 75013 Paris, France; (L.T.M.); (M.L.)
- Neurogenetics Team, EPHE, Paris Sciences Lettres Research University, 75000 Paris, France
| | - Giovanni Stevanin
- Institut du Cerveau—Paris Brain Institute—ICM, INSERM, CNRS, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 75013 Paris, France; (L.T.M.); (M.L.)
- Neurogenetics Team, EPHE, Paris Sciences Lettres Research University, 75000 Paris, France
- Correspondence:
| |
Collapse
|
15
|
Chen YJ, Zhang ZQ, Wang MW, Qiu YS, Yuan RY, Dong EL, Zhao Z, Zhou HT, Wang N, Chen WJ, Lin X. Novel Compound Missense and Intronic Splicing Mutation in ALDH18A1 Causes Autosomal Recessive Spastic Paraplegia. Front Neurol 2021; 12:627531. [PMID: 34093392 PMCID: PMC8170465 DOI: 10.3389/fneur.2021.627531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Hereditary spastic paraplegia (HSP) caused by mutations in ALDH18A1 have been reported as spastic paraplegia 9 (SPG9), with autosomal dominant and autosomal recessive transmission (SPG9A and SPG9B). SPG9 is rare and has shown phenotypic and genotypic heterogeneity in previous reports. Methods: This study screened ALDH18A1 mutations in autosomal recessive HSP patients using combined whole exome sequencing and RNA splicing analysis. We conducted in silico investigations, co-segregation analysis, and ELISA-based analysis of P5CS (Δ1-pyrroline-5-carboxylate synthetase; encoded by ALDH18A1) concentration to validate the pathogenicity of the detected ALDH18A1 variants. All previously reported bi-allelic ALDH18A1 mutations and cases were reviewed to summarize the genetic and clinical features of ALDH18A1-related HSP. Results: A novel missense mutation c.880T>C, p.S294P and an intronic splicing mutation c.-28-13A>G were both detected in ALDH18A1 in an autosomal recessive family presenting with a complicated form HSP. ELISA assays revealed significantly decreased P5CS concentration in the proband's plasma compared with that in the healthy controls. Moreover, review of previously reported recessive cases showed that SPG9B patients in our cohort presented with milder symptoms, i.e., later age at onset and without cognitive impairment. Conclusion: The present study expands the genetic and clinical spectrum of SPG9B caused by ALDH18A1 mutation. Our work defines new genetic variants to facilitate future diagnoses, in addition to demonstrating the highly informative value of splicing mutation prediction in the characterization of disease-related intronic variants.
Collapse
Affiliation(s)
- Yi-Jun Chen
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zai-Qiang Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Meng-Wen Wang
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yu-Sen Qiu
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ru-Ying Yuan
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - En-Lin Dong
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhe Zhao
- Department of Neuromuscular Disorders, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hai-Tao Zhou
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Ning Wang
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Wan-Jin Chen
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Xiang Lin
- Department of Neurology and Institute of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
16
|
Edmison D, Wang L, Gowrishankar S. Lysosome Function and Dysfunction in Hereditary Spastic Paraplegias. Brain Sci 2021; 11:152. [PMID: 33498913 PMCID: PMC7911997 DOI: 10.3390/brainsci11020152] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/15/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
Hereditary Spastic Paraplegias (HSPs) are a genetically diverse group of inherited neurological diseases with over 80 associated gene loci. Over the last decade, research into mechanisms underlying HSPs has led to an emerging interest in lysosome dysfunction. In this review, we highlight the different classes of HSPs that have been linked to lysosome defects: (1) a subset of complex HSPs where mutations in lysosomal genes are causally linked to the diseases, (2) other complex HSPs where mutation in genes encoding membrane trafficking adaptors lead to lysosomal defects, and (3) a subset of HSPs where mutations affect genes encoding proteins whose function is primarily linked to a different cellular component or organelle such as microtubule severing and Endoplasmic Reticulum-shaping, while also altering to lysosomes. Interestingly, aberrant axonal lysosomes, associated with the latter two subsets of HSPs, are a key feature observed in other neurodegenerative diseases such as Alzheimer's disease. We discuss how altered lysosome function and trafficking may be a critical contributor to HSP pathology and highlight the need for examining these features in the cortico-spinal motor neurons of HSP mutant models.
Collapse
Affiliation(s)
| | | | - Swetha Gowrishankar
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (D.E.); (L.W.)
| |
Collapse
|
17
|
Shirane M. Lipid Transfer-Dependent Endosome Maturation Mediated by Protrudin and PDZD8 in Neurons. Front Cell Dev Biol 2020; 8:615600. [PMID: 33385000 PMCID: PMC7769939 DOI: 10.3389/fcell.2020.615600] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
Endosome maturation refers to the conversion of early endosomes (EEs) to late endosomes (LEs) for subsequent fusion with lysosomes. It is an incremental process that involves a combination of endosome fusion and fission and which occurs at contact sites between endosomes and the endoplasmic reticulum (ER), with knowledge of the underlying mechanisms having increased greatly in recent years. Protrudin is an ER-resident protein that was originally shown to regulate neurite formation by promoting endosome trafficking, whereas PDZD8 is a mammalian paralog of a subunit of the yeast ERMES (ER-mitochondrial encounter structure) complex that possesses lipid transfer activity. A complex of protrudin and PDZD8 was recently found to promote endosome maturation by mediating lipid transfer at ER-endosome membrane contact sites. This review focuses on the roles of the protrudin-PDZD8 complex in tethering of endosomes to the ER, in mediating lipid transfer at such contact sites, and in regulating endosome dynamics, especially in neuronal cells. It also addresses the physiological contribution of endosome maturation mediated by this complex to neuronal polarity and integrity.
Collapse
Affiliation(s)
- Michiko Shirane
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
18
|
Shirane M, Shoji H, Hashimoto Y, Katagiri H, Kobayashi S, Manabe T, Miyakawa T, Nakayama KI. Protrudin-deficient mice manifest depression-like behavior with abnormalities in activity, attention, and cued fear-conditioning. Mol Brain 2020; 13:146. [PMID: 33172474 PMCID: PMC7654181 DOI: 10.1186/s13041-020-00693-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/03/2020] [Indexed: 01/02/2023] Open
Abstract
Protrudin is a protein that resides in the membrane of the endoplasmic reticulum and is highly expressed in the nervous system. Although mutations in the human protrudin gene (ZFYVE27, also known as SPG33) give rise to hereditary spastic paraplegia (HSP), the physiological role of the encoded protein has been largely unclear. We therefore generated mice deficient in protrudin and subjected them to a battery of behavioral tests designed to examine their intermediate phenotypes. The protrudin-deficient mice were found to have a reduced body size and to manifest pleiotropic behavioral abnormalities, including hyperactivity, depression-like behavior, and deficits in attention and fear-conditioning memory. They exhibited no signs of HSP, however, consistent with the notion that HSP-associated mutations of protrudin may elicit neural degeneration, not as a result of a loss of function, but rather as a result of a gain of toxic function. Overall, our results suggest that protrudin might play an indispensable role in normal neuronal development and behavior.
Collapse
Affiliation(s)
- Michiko Shirane
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan.
| | - Hirotaka Shoji
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Yutaka Hashimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan
| | - Hiroyuki Katagiri
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shizuka Kobayashi
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Toshiya Manabe
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tsuyoshi Miyakawa
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Fukuoka, Japan.
| |
Collapse
|
19
|
Protrudin and PDZD8 contribute to neuronal integrity by promoting lipid extraction required for endosome maturation. Nat Commun 2020; 11:4576. [PMID: 32917905 PMCID: PMC7486383 DOI: 10.1038/s41467-020-18413-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/21/2020] [Indexed: 01/08/2023] Open
Abstract
Endosome maturation depends on membrane contact sites (MCSs) formed between endoplasmic reticulum (ER) and endolysosomes (LyLEs). The mechanism underlying lipid supply for this process and its pathophysiological relevance remains unclear, however. Here, we identify PDZD8-the mammalian ortholog of a yeast ERMES subunit-as a protein that interacts with protrudin, which is located at ER-LyLE MCSs. Protrudin and PDZD8 promote the formation of ER-LyLE MCSs, and PDZD8 shows the ability to extract various lipids from the ER. Overexpression of both protrudin and PDZD8 in HeLa cells, as well as their depletion in mouse primary neurons, impairs endosomal homeostasis by inducing the formation of abnormal large vacuoles reminiscent of those apparent in spastin- or REEP1-deficient neurons. The protrudin-PDZD8 system is also essential for the establishment of neuronal polarity. Our results suggest that protrudin and PDZD8 cooperatively promote endosome maturation by mediating ER-LyLE tethering and lipid extraction at MCSs, thereby maintaining neuronal polarity and integrity.
Collapse
|
20
|
Park H, Kang JH, Lee S. Autophagy in Neurodegenerative Diseases: A Hunter for Aggregates. Int J Mol Sci 2020; 21:ijms21093369. [PMID: 32397599 PMCID: PMC7247013 DOI: 10.3390/ijms21093369] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
Cells have developed elaborate quality-control mechanisms for proteins and organelles to maintain cellular homeostasis. Such quality-control mechanisms are maintained by conformational folding via molecular chaperones and by degradation through the ubiquitin-proteasome or autophagy-lysosome system. Accumulating evidence suggests that impaired autophagy contributes to the accumulation of intracellular inclusion bodies consisting of misfolded proteins, which is a hallmark of most neurodegenerative diseases. In addition, genetic mutations in core autophagy-related genes have been reported to be linked to neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. Conversely, the pathogenic proteins, such as amyloid β and α-synuclein, are detrimental to the autophagy pathway. Here, we review the recent advances in understanding the relationship between autophagic defects and the pathogenesis of neurodegenerative diseases and suggest autophagy induction as a promising strategy for the treatment of these conditions.
Collapse
Affiliation(s)
- Hyungsun Park
- Department of Anatomy, College of Medicine, Inha University, Incheon 22212, Korea;
- Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea;
| | - Ju-Hee Kang
- Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea;
- Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea
| | - Seongju Lee
- Department of Anatomy, College of Medicine, Inha University, Incheon 22212, Korea;
- Hypoxia-related Disease Research Center, College of Medicine, Inha University, Incheon 22212, Korea;
- Correspondence: ; Tel.: +82-32-860-9891
| |
Collapse
|