1
|
Tazir M, Nouioua S. Distal hereditary motor neuropathies. Rev Neurol (Paris) 2024; 180:1031-1036. [PMID: 38702287 DOI: 10.1016/j.neurol.2023.09.005] [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: 08/02/2022] [Revised: 07/30/2023] [Accepted: 09/29/2023] [Indexed: 05/06/2024]
Abstract
Distal hereditary motor neuropathies (dHMN) are a group of heterogeneous hereditary disorders characterized by a slowly progressive distal pure motor neuropathy. Electrophysiology, with normal motor and sensory conduction velocities, can suggest the diagnosis of dHMN and guide the genetic study. More than thirty genes are currently associated with HMNs, but around 60 to 70% of cases of dHMN remain uncharacterized genetically. Recent cohort studies showed that HSPB1, GARS, BICB2 and DNAJB2 are among the most frequent dHMN genes and that the prevalence of the disease was calculated as 2.14 and 2.3 per 100,000. The determination of the different genes involved in dHMNs made it possible to observe a genotypic overlap with some other neurogenetic disorders and other hereditary neuropathies such as CMT2, mainly with the HSPB1, HSPB8, BICD2 and TRPV4 genes of AD-inherited transmission and recently observed with SORD gene of AR transmission which seems relatively frequent and potentially curable. Distal hereditary motor neuropathy that predominates in the upper limbs is linked mainly to three genes: GARS, BSCL2 and REEP1, whereas dHMN with vocal cord palsy is associated with SLC5A7, DCTN1 and TRPV4 genes. Among the rare AR forms of dHMN like IGHMBP2 and DNAJB2, the SIGMAR1 gene mutations as well as VRK1 variants are associated with a motor neuropathy phenotype often associated with upper motoneuron involvement. The differential diagnosis of these latter arises with juvenile forms of amyotrophic lateral sclerosis, that could be caused also by variations of these genes, as well as hereditary spastic paraplegia. A differential diagnosis of dHMN related to Brown Vialetto Van Laere syndrome due to riboflavin transporter deficiency is important to consider because of the therapeutic possibility.
Collapse
Affiliation(s)
- Meriem Tazir
- Department of Neurology, University Hospital Mustapha Bacha, Algiers, Algeria; Neurosciences Laboratory, University Benyoucef Benkhedda, Algiers, Algeria.
| | - Sonia Nouioua
- Neurosciences Laboratory, University Benyoucef Benkhedda, Algiers, Algeria; Department of Neurology, EHS El Maham, Cherchell,Tipaza, Algeria
| |
Collapse
|
2
|
van Eyll J, Prior R, Celanire S, Van Den Bosch L, Rombouts F. Therapeutic indications for HDAC6 inhibitors in the peripheral and central nervous disorders. Expert Opin Ther Targets 2024; 28:719-737. [PMID: 39305025 DOI: 10.1080/14728222.2024.2404571] [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: 04/16/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Inhibition of the enzymatic function of HDAC6 is currently being explored in clinical trials ranging from peripheral neuropathies to cancers. Advances in selective HDAC6 inhibitor discovery allowed studying highly efficacious brain penetrant and peripheral restrictive compounds for treating PNS and CNS indications. AREAS COVERED This review explores the multifactorial role of HDAC6 in cells, the common pathological hallmarks of PNS and CNS disorders, and how HDAC6 modulates these mechanisms. Pharmacological inhibition of HDAC6 and genetic knockout/knockdown studies as a therapeutic strategy in PNS and CNS indications were analyzed. Furthermore, we describe the recent developments in HDAC6 PET tracers and their utility in CNS indications. Finally, we explore the advancements and challenges with HDAC6 inhibitor compounds, such as hydroxamic acid, fluoromethyl oxadiazoles, HDAC6 degraders, and thiol-based inhibitors. EXPERT OPINION Based on extensive preclinical evidence, pharmacological inhibition of HDAC6 is a promising approach for treating both PNS and CNS disorders, given its involvement in neurodegeneration and aging-related cellular processes. Despite the progress in the development of selective HDAC6 inhibitors, safety concerns remain regarding their chronic administration in PNS and CNS indications, and the development of novel compound classes and modalities inhibiting HDAC6 function offer a way to mitigate some of these safety concerns.
Collapse
Affiliation(s)
| | | | - Sylvain Celanire
- Augustine Therapeutics, Research and Development, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | | |
Collapse
|
3
|
Fernandez-Fuente G, Farrugia MA, Peng Y, Schneider A, Svaren J, Puglielli L. Spatial selectivity of ATase inhibition in mouse models of Charcot-Marie-Tooth disease. Brain Commun 2024; 6:fcae232. [PMID: 39035418 PMCID: PMC11258571 DOI: 10.1093/braincomms/fcae232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/30/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
The endoplasmic reticulum acetylation machinery has emerged as a new branch of the larger endoplasmic reticulum quality control system. It regulates the selection of correctly folded polypeptides as well as reticulophagy-mediated removal of toxic protein aggregates with the former being a particularly important aspect of the proteostatic functions of endoplasmic reticulum acetylation. Essential to this function is the Nε-lysine acetyltransferase activity of acetyltransferase 1 and acetyltransferase 2, which regulates the induction of endoplasmic reticulum-specific autophagy through the acetylation of the autophagy-related protein 9A. Here, we used three mouse models of Charcot-Marie-Tooth disease, peripheral myelin protein 22/Tr-J, C3-peripheral myelin protein 22 and myelin protein zero/ttrr, to study spatial and translational selectivity of endoplasmic reticulum acetyltransferase inhibitors. The results show that inhibition of the endoplasmic reticulum acetyltransferases selectively targets misfolding/pro-aggregating events occurring in the lumen of the organelle. Therefore, they establish acetyltransferase 1 and acetyltransferase 2 as the first proven targets for disease-causing proteotoxic states that initiate within the lumen of the endoplasmic reticulum/secretory pathway.
Collapse
Affiliation(s)
- Gonzalo Fernandez-Fuente
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mark A Farrugia
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yajing Peng
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Andrew Schneider
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Luigi Puglielli
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI 53705, USA
| |
Collapse
|
4
|
van Asperen JV, Kotaich F, Caillol D, Bomont P. Neurofilaments: Novel findings and future challenges. Curr Opin Cell Biol 2024; 87:102326. [PMID: 38401181 DOI: 10.1016/j.ceb.2024.102326] [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: 09/30/2023] [Accepted: 01/07/2024] [Indexed: 02/26/2024]
Abstract
Neurofilaments (NFs) are abundant cytoskeletal proteins that emerge as a critical hub for cell signalling within neurons. As we start to uncover essential roles of NFs in regulating microtubule and organelle dynamics, nerve conduction and neurotransmission, novel discoveries are expected to arise in genetics, with NFs identified as causal genes for various neurodegenerative diseases. This review will discuss how the latest advances in fundamental and translational research illuminate our understanding of NF biology, particularly their assembly, organisation, transport and degradation. We will emphasise the notion that filaments are not one entity and that future challenges will be to apprehend their diverse composition and structural heterogeneity and to scrutinize how this regulates signalling, sustains neuronal physiology and drives pathophysiology in disease.
Collapse
Affiliation(s)
- Jessy V van Asperen
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| | - Farah Kotaich
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| | - Damien Caillol
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| | - Pascale Bomont
- ERC Team, NeuroMyoGene Insitute, INMG-PGNM, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France.
| |
Collapse
|
5
|
Tran VK, Cao MH, Nguyen TTH, Le PT, Tran HA, Vu DC, Nguyen HT, Nguyen MTP, Bui TH, Nguyen TB, Ta TV, Tran TH. A novel IGHMBP2 variant and clinical diversity in Vietnamese SMARD1 and CMT2S patients. Front Pediatr 2024; 12:1165492. [PMID: 38415210 PMCID: PMC10896978 DOI: 10.3389/fped.2024.1165492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 01/26/2024] [Indexed: 02/29/2024] Open
Abstract
Background Pathogenic variants in the IGHMBP2 gene are associated with two distinct autosomal recessive neuromuscular disorders: spinal muscular atrophy with respiratory distress type 1 (SMARD1; OMIM #604320) and Charcot-Marie-Tooth type 2S (CMT2S; OMIM #616155). SMARD1 is a severe and fatal condition characterized by infantile-onset respiratory distress, diaphragmatic palsy, and distal muscular weakness, while CMT2S follows a milder clinical course, with slowly progressive distal muscle weakness and sensory loss, without manifestations of respiratory disorder. Methods Whole-exome sequencing of the IGHMBP2 gene was performed for eight Vietnamese patients with IGHMBP2-related neuromuscular disorders including five patients with SMARD1 and the others with CMT2S. Results We identified one novel IGHMBP2 variant c.1574T > C (p.Leu525Pro) in a SMARD1 patient. Besides that, two patients shared the same pathogenic variants (c.1235 + 3A > G/c.1334A > C) but presented completely different clinical courses: one with SMARD1 who deceased at 8 months of age, the other with CMT2S was alive at 3 years old without any respiratory distress. Conclusion This study is the first to report IGHMBP-2-related neuromuscular disorders in Vietnam. A novel IGHMBP2 variant c.1574T > C (p.Leu525Pro) expressing SMARD1 phenotype was detected. The presence of three patients with the same genotype but distinct clinical outcomes suggested the interaction of variants and other factors including relating modified genes in the mechanism of various phenotypes.
Collapse
Affiliation(s)
- Van Khanh Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Department of Molecular Pathology, Faculty of Medical Laboratory Technology, Hanoi Medical University, Hanoi, Vietnam
| | - My Ha Cao
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | | | - Phuong Thi Le
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Hai Anh Tran
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Dung Chi Vu
- Department of Endocrinology, Metabolism and Genetics, National Hospital of Pediatrics, Hanoi, Vietnam
| | - Ha Thu Nguyen
- Department of Endocrinology, Metabolism and Genetics, National Hospital of Pediatrics, Hanoi, Vietnam
| | | | - The-Hung Bui
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
- Center for Molecular Medicine, Clinical Genetics Unit, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Thanh Binh Nguyen
- Center for Gene and Protein Research, Hanoi Medical University, Hanoi, Vietnam
| | - Thanh Van Ta
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
- Clinical Laboratory, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| | - Thinh Huy Tran
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
- Clinical Laboratory, Hanoi Medical University Hospital, Hanoi Medical University, Hanoi, Vietnam
| |
Collapse
|
6
|
Della Marina A, Hentschel A, Czech A, Schara-Schmidt U, Preusse C, Laner A, Abicht A, Ruck T, Weis J, Choueiri C, Lochmüller H, Kölbel H, Roos A. Novel Genetic and Biochemical Insights into the Spectrum of NEFL-Associated Phenotypes. J Neuromuscul Dis 2024; 11:625-645. [PMID: 38578900 PMCID: PMC11091643 DOI: 10.3233/jnd-230230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
Background NEFL encodes for the neurofilament light chain protein. Pathogenic variants in NEFL cause demyelinating, axonal and intermediate forms of Charcot-Marie-Tooth disease (CMT) which present with a varying degree of severity and somatic mutations have not been described yet. Currently, 34 different CMT-causing pathogenic variants in NEFL in 174 patients have been reported. Muscular involvement was also described in CMT2E patients mostly as a secondary effect. Also, there are a few descriptions of a primary muscle vulnerability upon pathogenic NEFL variants. Objectives To expand the current knowledge on the genetic landscape, clinical presentation and muscle involvement in NEFL-related neurological diseases by retrospective case study and literature review. Methods We applied in-depth phenotyping of new and already reported cases, molecular genetic testing, light-, electron- and Coherent Anti-Stokes Raman Scattering-microscopic studies and proteomic profiling in addition to in silico modelling of NEFL-variants. Results We report on a boy with a muscular phenotype (weakness, myalgia and cramps, Z-band alterations and mini-cores in some myofibers) associated with the heterozygous p.(Phe104Val) NEFL-variant, which was previously described in a neuropathy case. Skeletal muscle proteomics findings indicated affection of cytoskeletal proteins. Moreover, we report on two further neuropathic patients (16 years old girl and her father) both carrying the heterozygous p.(Pro8Ser) variant, which has been identified as 15% somatic mosaic in the father. While the daughter presented with altered neurophysiology,neurogenic clump feet and gait disturbances, the father showed clinically only feet deformities. As missense variants affecting proline at amino acid position 8 are leading to neuropathic manifestations of different severities, in silico modelling of these different amino acid substitutions indicated variable pathogenic impact correlating with disease onset. Conclusions Our findings provide new morphological and biochemical insights into the vulnerability of denervated muscle (upon NEFL-associated neuropathy) as well as novel genetic findings expanding the current knowledge on NEFL-related neuromuscular phenotypes and their clinical manifestations. Along this line, our data show that even subtle expression of somatic NEFL variants can lead to neuromuscular symptoms.
Collapse
Affiliation(s)
- Adela Della Marina
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Artur Czech
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Corinna Preusse
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | | | - Angela Abicht
- Medical Genetics Center, Munich, Germany
- Friedrich-Baur Institute, Ludwig Maximilian University, Munich, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Catherine Choueiri
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Hanns Lochmüller
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Heike Kölbel
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Andreas Roos
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| |
Collapse
|
7
|
Kotaich F, Caillol D, Bomont P. Neurofilaments in health and Charcot-Marie-Tooth disease. Front Cell Dev Biol 2023; 11:1275155. [PMID: 38164457 PMCID: PMC10758125 DOI: 10.3389/fcell.2023.1275155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024] Open
Abstract
Neurofilaments (NFs) are the most abundant component of mature neurons, that interconnect with actin and microtubules to form the cytoskeleton. Specifically expressed in the nervous system, NFs present the particularity within the Intermediate Filament family of being formed by four subunits, the neurofilament light (NF-L), medium (NF-M), heavy (NF-H) proteins and α-internexin or peripherin. Here, we review the current knowledge on NF proteins and neurofilaments, from their domain structures and their model of assembly to the dynamics of their transport and degradation along the axon. The formation of the filament and its behaviour are regulated by various determinants, including post-transcriptional (miRNA and RBP proteins) and post-translational (phosphorylation and ubiquitination) modifiers. Altogether, the complex set of modifications enable the neuron to establish a stable but elastic NF array constituting the structural scaffold of the axon, while permitting the local expression of NF proteins and providing the dynamics necessary to fulfil local demands and respond to stimuli and injury. Thus, in addition to their roles in mechano-resistance, radial axonal outgrowth and nerve conduction, NFs control microtubule dynamics, organelle distribution and neurotransmission at the synapse. We discuss how the studies of neurodegenerative diseases with NF aggregation shed light on the biology of NFs. In particular, the NEFL and NEFH genes are mutated in Charcot-Marie-Tooth (CMT) disease, the most common inherited neurological disorder of the peripheral nervous system. The clinical features of the CMT forms (axonal CMT2E, CMT2CC; demyelinating CMT1F; intermediate I-CMT) with symptoms affecting the central nervous system (CNS) will allow us to further investigate the physiological roles of NFs in the brain. Thus, NF-CMT mouse models exhibit various degrees of sensory-motor deficits associated with CNS symptoms. Cellular systems brought findings regarding the dominant effect of NF-L mutants on NF aggregation and transport, although these have been recently challenged. Neurofilament detection without NF-L in recessive CMT is puzzling, calling for a re-examination of the current model in which NF-L is indispensable for NF assembly. Overall, we discuss how the fundamental and translational fields are feeding each-other to increase but also challenge our knowledge of NF biology, and to develop therapeutic avenues for CMT and neurodegenerative diseases with NF aggregation.
Collapse
Affiliation(s)
| | | | - Pascale Bomont
- ERC team, NeuroMyoGene Institute-Pathophysiology and Genetics of Neuron and Muscle, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| |
Collapse
|
8
|
Espinoza KS, Hermanson KN, Beard CA, Schwartz NU, Snider JM, Low BE, Wiles MV, Hannun YA, Obeid LM, Snider AJ. A novel HSPB1 S139F mouse model of Charcot-Marie-Tooth Disease. Prostaglandins Other Lipid Mediat 2023; 169:106769. [PMID: 37625781 PMCID: PMC10843462 DOI: 10.1016/j.prostaglandins.2023.106769] [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: 05/31/2023] [Revised: 08/01/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Charcot-Marie-Tooth Disease (CMT) is a commonly inherited peripheral polyneuropathy. Clinical manifestations for this disease include symmetrical distal polyneuropathy, altered deep tendon reflexes, distal sensory loss, foot deformities, and gait abnormalities. Genetic mutations in heat shock proteins have been linked to CMT2. Specifically, mutations in the heat shock protein B1 (HSPB1) gene encoding for heat shock protein 27 (Hsp27) have been linked to CMT2F and distal hereditary motor and sensory neuropathy type 2B (dHMSN2B) subtype. The goal of the study was to examine the role of an endogenous mutation in HSPB1 in vivo and to define the effects of this mutation on motor function and pathology in a novel animal model. As sphingolipids have been implicated in hereditary and sensory neuropathies, we examined sphingolipid metabolism in central and peripheral nervous tissues in 3-month-old HspS139F mice. Though sphingolipid levels were not altered in sciatic nerves from HspS139F mice, ceramides and deoxyceramides, as well as sphingomyelins (SMs) were elevated in brain tissues from HspS139F mice. Histology was utilized to further characterize HspS139F mice. HspS139F mice exhibited no alterations to the expression and phosphorylation of neurofilaments, or in the expression of acetylated α-tubulin in the brain or sciatic nerve. Interestingly, HspS139F mice demonstrated cerebellar demyelination. Locomotor function, grip strength and gait were examined to define the role of HspS139F in the clinical phenotypes associated with CMT2F. Gait analysis revealed no differences between HspWT and HspS139F mice. However, both coordination and grip strength were decreased in 3-month-old HspS139F mice. Together these data suggest that the endogenous S139F mutation in HSPB1 may serve as a mouse model for hereditary and sensory neuropathies such as CMT2F.
Collapse
Affiliation(s)
- Keila S Espinoza
- Department of Physiology, University of Arizona, Tucson, AZ 85721, USA
| | - Kyra N Hermanson
- Department of Physiology, University of Arizona, Tucson, AZ 85721, USA
| | - Cameron A Beard
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ 85721, USA
| | - Nicholas U Schwartz
- Department of Neurology, Stanford University Medical Center, Stanford, CA 94304, USA
| | - Justin M Snider
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ 85721, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| | - Benjamin E Low
- Technology Evaluation and Development, The Jackson Laboratory, Bar Harbor, ME, USA; Genetic Resource Science, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Michael V Wiles
- Technology Evaluation and Development, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Yusuf A Hannun
- Department of Medicine and Stony Brook Cancer Center, Stony Brook, NY 11794, USA; Northport Veterans Affairs Medical Center, Northport, NY 11768, USA
| | - Lina M Obeid
- Department of Medicine and Stony Brook Cancer Center, Stony Brook, NY 11794, USA; Northport Veterans Affairs Medical Center, Northport, NY 11768, USA
| | - Ashley J Snider
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ 85721, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
9
|
Han JE, Kang KH, Kim H, Hong YB, Choi BO, Koh H. PINK1 and Parkin rescue motor defects and mitochondria dysfunction induced by a patient-derived HSPB3 mutant in Drosophila models. Biochem Biophys Res Commun 2023; 682:71-76. [PMID: 37804589 DOI: 10.1016/j.bbrc.2023.09.092] [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: 09/11/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/09/2023]
Abstract
Small heat shock proteins (sHSPs) are ATP-independent molecular chaperones with the α-crystalline domain that is critical to their chaperone activity. Within the sHSP family, three (HSPB1, HSPB3, and HSPB8) proteins are linked with inherited peripheral neuropathies, including distal hereditary motor neuropathy (dHMN) and Charco-Marie-Tooth disease (CMT). In this study, we introduced the HSPB3 Y118H (HSPB3Y118H) mutant gene identified from the CMT2 family in Drosophila. With a missense mutation on its α-crystalline domain, this human HSPB3 mutant gene induced a loss of motor activity accompanied by reduced mitochondrial membrane potential in fly neuronal tissues. Moreover, mitophagy, a critical mechanism of mitochondrial quality control, is downregulated in fly motor neurons expressing HSPB3Y118H. Surprisingly, PINK1 and Parkin, the core regulators of mitophagy, successfully rescued these motor and mitochondrial abnormalities in HSPB3 mutant flies. Results from the first animal model of HSPB3 mutations suggest that mitochondrial dysfunction plays a critical role in HSPB3-associated human pathology.
Collapse
Affiliation(s)
- Ji Eun Han
- Department of Pharmacology, Dong-A University College of Medicine, Busan, 49201, South Korea; Department of Translational Biomedical Sciences, Dong-A University College of Medicine, Busan, 49201, South Korea
| | - Kyong-Hwa Kang
- Department of Pharmacology, Dong-A University College of Medicine, Busan, 49201, South Korea; Neuroscience Translational Research Solution Center, Dong-A University College of Medicine, Busan, 49201, South Korea
| | - Hyunjin Kim
- Department of Pharmacology, Dong-A University College of Medicine, Busan, 49201, South Korea; Neuroscience Translational Research Solution Center, Dong-A University College of Medicine, Busan, 49201, South Korea
| | - Young Bin Hong
- Department of Biochemistry, Dong-A University College of Medicine, Busan, 49201, South Korea
| | - Byung-Ok Choi
- Department of Neurology, SAIHST, Sungkyunkwan University School of Medicine, Seoul, 06351, South Korea
| | - Hyongjong Koh
- Department of Pharmacology, Dong-A University College of Medicine, Busan, 49201, South Korea; Department of Translational Biomedical Sciences, Dong-A University College of Medicine, Busan, 49201, South Korea; Neuroscience Translational Research Solution Center, Dong-A University College of Medicine, Busan, 49201, South Korea.
| |
Collapse
|
10
|
Ecroyd H, Bartelt-Kirbach B, Ben-Zvi A, Bonavita R, Bushman Y, Casarotto E, Cecconi C, Lau WCY, Hibshman JD, Joosten J, Kimonis V, Klevit R, Liberek K, McMenimen KA, Miwa T, Mogk A, Montepietra D, Peters C, Rocchetti MT, Saman D, Sisto A, Secco V, Strauch A, Taguchi H, Tanguay M, Tedesco B, Toth ME, Wang Z, Benesch JLP, Carra S. The beauty and complexity of the small heat shock proteins: a report on the proceedings of the fourth workshop on small heat shock proteins. Cell Stress Chaperones 2023; 28:621-629. [PMID: 37462824 PMCID: PMC10746627 DOI: 10.1007/s12192-023-01360-x] [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] [Accepted: 05/29/2023] [Indexed: 12/23/2023] Open
Abstract
The Fourth Cell Stress Society International workshop on small heat shock proteins (sHSPs), a follow-up to successful workshops held in 2014, 2016 and 2018, took place as a virtual meeting on the 17-18 November 2022. The meeting was designed to provide an opportunity for those working on sHSPs to reconnect and discuss their latest work. The diversity of research in the sHSP field is reflected in the breadth of topics covered in the talks presented at this meeting. Here we summarise the presentations at this meeting and provide some perspectives on exciting future topics to be addressed in the field.
Collapse
Affiliation(s)
- Heath Ecroyd
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia.
| | | | - Anat Ben-Zvi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Raffaella Bonavita
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", 80131, Naples, Italy
| | - Yevheniia Bushman
- Center for Protein Assemblies and Department Chemie, Technische Universität München, München, Germany
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti" (DiSFeB), Dipartimento di Eccellenza, Università degli Studi di Milano, Milan, Italy
| | - Ciro Cecconi
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Modena, Italy
- Istituto Nanoscienze-CNR-NANO, Center S3, Modena, Italy
| | - Wilson Chun Yu Lau
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Jonathan D Hibshman
- Biology Department, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joep Joosten
- Department of Synthetic Organic Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Virginia Kimonis
- Division of Genetics and Genomic Medicine, Department of Pediatrics, University of California - Irvine, Orange, CA, 92868, USA
- Department of Neurology and Department of Pathology, University of California, Irvine, CA, 92697, USA
| | - Rachel Klevit
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Krzysztof Liberek
- Intercollegiate Faculty of Biotechnology, University of Gdansk, Gdansk, Poland
| | - Kathryn A McMenimen
- Program in Biochemistry and Department of Chemistry, Mount Holyoke College, South Hadley, MA, 01075, USA
| | - Tsukumi Miwa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8503, Japan
| | - Axel Mogk
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld, 282, Heidelberg, Germany
| | - Daniele Montepietra
- Istituto Nanoscienze-CNR-NANO, Center S3, Modena, Italy
- Department of Department of Chemical, Life and Environmental sustainability sciences, University of Parma, Parma, Italy
| | - Carsten Peters
- Center for Protein Assemblies and Department Chemie, Technische Universität München, München, Germany
| | - Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122, Foggio, Italy
| | - Dominik Saman
- Department of Chemistry, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Angela Sisto
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences and Institute Born Bunge, University of Antwerp, Antwerpen, Belgium
| | - Valentina Secco
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Annika Strauch
- Center for Protein Assemblies and Department Chemie, Technische Universität München, München, Germany
| | - Hideki Taguchi
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8503, Japan
| | - Morgan Tanguay
- Program in Biochemistry and Department of Chemistry, Mount Holyoke College, South Hadley, MA, 01075, USA
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti" (DiSFeB), Dipartimento di Eccellenza, Università degli Studi di Milano, Milan, Italy
| | - Melinda E Toth
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, H-6726, Hungary
| | - Zihao Wang
- Department of Chemistry, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Justin L P Benesch
- Department of Chemistry, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| | - Serena Carra
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
11
|
Naruse H, Okubo S, Sudo A, Mitsui J, Mikata T, Ishiura H, Morishita S, Tsuji S, Toda T. Clinical features of a family with late-onset distal hereditary motor neuropathy harboring p.Pro39Leu variant of HSPB1. J Peripher Nerv Syst 2023; 28:518-521. [PMID: 37249095 DOI: 10.1111/jns.12567] [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: 04/12/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND AND AIMS Pathogenic variants of HSPB1, the gene encoding the small heat shock protein 27, have been reported to cause autosomal dominant distal hereditary motor neuropathy (dHMN) type II and autosomal dominant Charcot-Marie-Tooth (CMT) disease with minimal sensory involvement (CMT2F). This study aimed to describe the clinical features of patients in a family with late-onset dHMN carrying the Pro39Leu variant of HSPB1. METHODS Whole-exome sequence analysis identified a heterozygous pathogenic variant (Pro39Leu) of HSPB1 in the proband. The presence of the HSPB1 Pro39Leu variant in two affected individuals was confirmed using direct nucleotide sequence analysis. RESULTS Both patients exhibited distal muscle weakness with lower extremity predominance and no obvious sensory deficits, leading to a clinical diagnosis of late-onset dHMN. Nerve conduction studies (NCSs) revealed a subclinical complication of sensory disturbance in one of the patients. The clinical and electrophysiological findings of patients with the HSPB1 Pro39Leu variant in this study and previous reports are summarized. INTERPRETATION This study suggests that the clinical spectrum of patients carrying HSPB1 Pro39Leu variants, especially the disease onset, might be broader than expected, and HSPB1 variants should be considered in patients diagnosed with late-onset dHMN. Furthermore, patients with dHMN may have concomitant sensory deficits that should be evaluated using NCSs.
Collapse
Affiliation(s)
- Hiroya Naruse
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Precision Medicine Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - So Okubo
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Sudo
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jun Mitsui
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Precision Medicine Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Mikata
- Department of Neurology, National Hospital Organization Shimoshizu National Hospital, Yotsukaido, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Morishita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Shoji Tsuji
- Institute of Medical Genomics, International University of Health and Welfare, Chiba, Japan
| | - Tatsushi Toda
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Moon S, Wang B, Ahn BS, Ryu AH, Hard ER, Javed A, Pratt MR. O-GlcNAc Modification Alters the Chaperone Activity of HSP27 Charcot-Marie-Tooth Type 2 (CMT2) Variants in a Mutation-Selective Fashion. ACS Chem Biol 2023; 18:1705-1712. [PMID: 37540114 PMCID: PMC10442854 DOI: 10.1021/acschembio.3c00292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Increased O-GlcNAc is a common feature of cellular stress, and the upregulation of this dynamic modification is associated with improved survival under these conditions. Likewise, the heat shock proteins are also increased under stress and prevent protein misfolding and aggregation. We previously linked these two phenomena by demonstrating that O-GlcNAc directly increases the chaperone of certain small heat shock proteins, including HSP27. Here, we examine this linkage further by exploring the potential function of O-GlcNAc on mutants of HSP27 that cause a heritable neuropathy called Charcot-Marie-Tooth type 2 (CMT2) disease. Using synthetic protein chemistry, we prepared five of these mutants bearing an O-GlcNAc at the major site of modification. Upon subsequent biochemical analysis of these proteins, we found that O-GlcNAc has different effects, depending on the location of the individual mutants. We believe that this has important implications for O-GlcNAc and other PTMs in the context of polymorphisms or diseases with high levels of protein mutation.
Collapse
Affiliation(s)
- Stuart
P. Moon
- Departments
of Chemistry and Biological Sciences, University of Southern
California, Los Angeles, California 90089, United States
| | - Binyou Wang
- Departments
of Chemistry and Biological Sciences, University of Southern
California, Los Angeles, California 90089, United States
| | - Benjamin S. Ahn
- Departments
of Chemistry and Biological Sciences, University of Southern
California, Los Angeles, California 90089, United States
| | - Andrew H. Ryu
- Departments
of Chemistry and Biological Sciences, University of Southern
California, Los Angeles, California 90089, United States
| | - Eldon R. Hard
- Departments
of Chemistry and Biological Sciences, University of Southern
California, Los Angeles, California 90089, United States
| | - Afraah Javed
- Departments
of Chemistry and Biological Sciences, University of Southern
California, Los Angeles, California 90089, United States
| | - Matthew R. Pratt
- Departments
of Chemistry and Biological Sciences, University of Southern
California, Los Angeles, California 90089, United States
| |
Collapse
|
13
|
Abstract
Neurons are markedly compartmentalized, which makes them reliant on axonal transport to maintain their health. Axonal transport is important for anterograde delivery of newly synthesized macromolecules and organelles from the cell body to the synapse and for the retrograde delivery of signaling endosomes and autophagosomes for degradation. Dysregulation of axonal transport occurs early in neurodegenerative diseases and plays a key role in axonal degeneration. Here, we provide an overview of mechanisms for regulation of axonal transport; discuss how these mechanisms are disrupted in neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, hereditary spastic paraplegia, amyotrophic lateral sclerosis, and Charcot-Marie-Tooth disease; and discuss therapeutic approaches targeting axonal transport.
Collapse
|
14
|
Szebesczyk A, Słowik J. Heat shock proteins and metal ions - Reaction or interaction? Comput Struct Biotechnol J 2023; 21:3103-3108. [PMID: 37273852 PMCID: PMC10236365 DOI: 10.1016/j.csbj.2023.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/06/2023] Open
Abstract
Heat shock proteins (HSPs) are part of the cell's molecular chaperone system responsible for the proper folding (or refolding) of proteins. They are expressed in cells of a wide variety of organisms, from bacteria and fungi to humans. While some HSPs require metal ions for proper functioning, others are expressed as a response of the organism to either essential or toxic metal ions. Their presence can influence the occurrence of cellular processes, even those as significant as programmed cell death. The development of research methods and structural modeling has enabled increasingly accurate recognition of new HSP functions, including their role in maintaining metal ion homeostasis. Current investigations on the expression of HSPs in response to heavy metal ions include not only the direct effect of these ions on the cell but also analysis of reactive oxygen species (ROS) and the increased production of HSPs with increasing ROS concentration. This minireview contains information about the direct and indirect interactions of heat shock proteins with metal ions, both those of biological importance and heavy metals.
Collapse
|
15
|
Small heat shock proteins operate as molecular chaperones in the mitochondrial intermembrane space. Nat Cell Biol 2023; 25:467-480. [PMID: 36690850 PMCID: PMC10014586 DOI: 10.1038/s41556-022-01074-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/12/2022] [Indexed: 01/24/2023]
Abstract
Mitochondria are complex organelles with different compartments, each harbouring their own protein quality control factors. While chaperones of the mitochondrial matrix are well characterized, it is poorly understood which chaperones protect the mitochondrial intermembrane space. Here we show that cytosolic small heat shock proteins are imported under basal conditions into the mitochondrial intermembrane space, where they operate as molecular chaperones. Protein misfolding in the mitochondrial intermembrane space leads to increased recruitment of small heat shock proteins. Depletion of small heat shock proteins leads to mitochondrial swelling and reduced respiration, while aggregation of aggregation-prone substrates is countered in their presence. Charcot-Marie-Tooth disease-causing mutations disturb the mitochondrial function of HSPB1, potentially linking previously observed mitochondrial dysfunction in Charcot-Marie-Tooth type 2F to its role in the mitochondrial intermembrane space. Our results reveal that small heat shock proteins form a chaperone system that operates in the mitochondrial intermembrane space.
Collapse
|
16
|
Gallagher ER, Holzbaur ELF. The selective autophagy adaptor p62/SQSTM1 forms phase condensates regulated by HSP27 that facilitate the clearance of damaged lysosomes via lysophagy. Cell Rep 2023; 42:112037. [PMID: 36701233 PMCID: PMC10366342 DOI: 10.1016/j.celrep.2023.112037] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/16/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
In response to lysosomal damage, cells engage several quality-control mechanisms, including the selective isolation and degradation of damaged lysosomes by lysophagy. Here, we report that the selective autophagy adaptor SQSTM1/p62 is recruited to damaged lysosomes in both HeLa cells and neurons and is required for lysophagic flux. The Phox and Bem1p (PB1) domain of p62 mediates oligomerization and is specifically required for lysophagy. Consistent with this observation, we find that p62 forms condensates on damaged lysosomes. These condensates are precisely tuned by the small heat shock protein HSP27, which is phosphorylated in response to lysosomal injury and maintains the liquidity of p62 condensates, facilitating autophagosome formation. Mutations in p62 have been identified in patients with amyotrophic lateral sclerosis (ALS); ALS-associated mutations in p62 impair lysophagy, suggesting that deficits in this pathway may contribute to neurodegeneration. Thus, p62 condensates regulated by HSP27 promote lysophagy by forming platforms for autophagosome biogenesis at damaged lysosomes.
Collapse
Affiliation(s)
- Elizabeth R Gallagher
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
17
|
Quan G, Duan J, Fick W, Candau JN. Expression of the gonad-specific small heat shock protein, CfHSP20.2, in the spruce budworm, Choristoneura fumiferana (Clem.). J Therm Biol 2023; 112:103463. [PMID: 36796908 DOI: 10.1016/j.jtherbio.2023.103463] [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: 02/16/2022] [Revised: 12/30/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
Small heat shock proteins (sHSPs) play important roles in insect development and stress resistance. However, the in vivo functions and mechanisms of action remain largely unknown or unclear for most members of the sHSPs in insects. This study investigated the expression of CfHSP20.2 in the spruce budworm, Choristoneura fumiferana (Clem.) under normal and heat-stress conditions. Under normal conditions, CfHSP20.2 transcript and protein were highly and constantly expressed in the testes of male larvae, pupae and young adults and in the ovaries of female late-stage pupae and adults. After adult eclosion, CfHSP20.2 remained highly and almost constantly expressed in the ovaries, but in contrast, was downregulated in the testes. Upon heat stress, CfHSP20.2 was upregulated in the gonads and non-gonadal tissues in both sexes. These results indicate that CfHSP20.2 expression is gonad-specific and heat-inducible. This provides evidence that the CfHSP20.2 protein plays important roles during reproductive development under normal environmental conditions, while under heat-stress conditions, it may also enhance the thermal tolerance of the gonads and non-gonadal tissues.
Collapse
Affiliation(s)
- Guoxing Quan
- Natural Resources Canada, Canadian Forest Service, Great Lakes Forestry Centre, 1219 Queen Street East, Sault Ste. Marie, Ontario, P6A 2E5, Canada.
| | - Jun Duan
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - William Fick
- Natural Resources Canada, Canadian Forest Service, Great Lakes Forestry Centre, 1219 Queen Street East, Sault Ste. Marie, Ontario, P6A 2E5, Canada
| | - Jean-Noël Candau
- Natural Resources Canada, Canadian Forest Service, Great Lakes Forestry Centre, 1219 Queen Street East, Sault Ste. Marie, Ontario, P6A 2E5, Canada
| |
Collapse
|
18
|
Braun JEA. Extracellular chaperone networks and the export of J-domain proteins. J Biol Chem 2023; 299:102840. [PMID: 36581212 PMCID: PMC9867986 DOI: 10.1016/j.jbc.2022.102840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022] Open
Abstract
An extracellular network of molecular chaperones protects a diverse array of proteins that reside in or pass through extracellular spaces. Proteins in the extracellular milieu face numerous challenges that can lead to protein misfolding and aggregation. As a checkpoint for proteins that move between cells, extracellular chaperone networks are of growing clinical relevance. J-domain proteins (JDPs) are ubiquitous molecular chaperones that are known for their essential roles in a wide array of fundamental cellular processes through their regulation of heat shock protein 70s. As the largest molecular chaperone family, JDPs have long been recognized for their diverse functions within cells. Some JDPs are elegantly selective for their "client proteins," some do not discriminate among substrates and others act cooperatively on the same target. The realization that JDPs are exported through both classical and unconventional secretory pathways has fueled investigation into the roles that JDPs play in protein quality control and intercellular communication. The proposed functions of exported JDPs are diverse. Studies suggest that export of DnaJB11 enhances extracellular proteostasis, that intercellular movement of DnaJB1 or DnaJB6 enhances the proteostasis capacity in recipient cells, whereas the import of DnaJB8 increases resistance to chemotherapy in recipient cancer cells. In addition, the export of DnaJC5 and concurrent DnaJC5-dependent ejection of dysfunctional and aggregation-prone proteins are implicated in the prevention of neurodegeneration. This review provides a brief overview of the current understanding of the extracellular chaperone networks and outlines the first wave of studies describing the cellular export of JDPs.
Collapse
Affiliation(s)
- Janice E A Braun
- Department of Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
19
|
Weterman MAJ, Bronk M, Jongejan A, Hoogendijk JE, Krudde J, Karjosukarso D, Goebel HH, Aronica E, Jöbsis GJ, van Ruissen F, van Spaendonck-Zwarts KY, de Visser M, Baas F. Pathogenic variants in three families with distal muscle involvement. Neuromuscul Disord 2023; 33:58-64. [PMID: 36539320 DOI: 10.1016/j.nmd.2022.11.007] [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: 07/05/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
Three families suspected of distal hereditary motor neuropathy underwent genetic screening with the aim to identify the molecular defect underlying the disease. The description of the identification reflects the shift in molecular diagnostics that was made during the last decades. Our candidate gene approach yielded a known pathogenic variant in BSCL2 (p.Asn88Ser) in one family, and via a CMT-capture, in HSPB1 (p.Arg127Trp), in addition to five other variations in Charcot-Marie-Tooth-related genes in the proband of the second family. In the third family, using whole exome sequencing, followed by linkage-by-location, a three base pair deletion in exon 33 of MYH7 (p.Glu1508del) was found, a reported pathogenic allele albeit for a myopathy. After identification of the causative molecular defect, cardiac examination was performed for patients of the third family and this demonstrated abnormalities in three out of five affected family members. Heterogeneity and expansion of clinical phenotypes beyond known characteristics requires a wider set of genes to be screened. Whole exome/genome analysis with limited prior clinical information may therefore be used to precede a detailed clinical evaluation in cases of large families, preventing screening of a too narrow set of genes, and enabling the identification of novel disease-associated genes. In our cases, the variants had been reported, and co-segregation analysis confirmed the molecular diagnosis.
Collapse
Affiliation(s)
- Marian A J Weterman
- Department of Genome Analysis/Clinical Genetics, Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, the Netherlands; Dept Clinical Genetics, LUMC, Leiden, the Netherlands.
| | - Marieke Bronk
- Department of Neurology, University Medical Center Amsterdam, location Academic Medical Center, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Department of Bio-informatics, University Medical Center Amsterdam, location Academic Medical Center, Amsterdam, the Netherlands
| | - Jessica E Hoogendijk
- Department of Neurology, UMC Brain Center, University Medical Center, Utrecht, the Netherlands
| | - Judith Krudde
- Department of Neurology, University Medical Center Amsterdam, location Academic Medical Center, Amsterdam, the Netherlands
| | - Dyah Karjosukarso
- Department of Genome Analysis/Clinical Genetics, Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, the Netherlands
| | - Hans H Goebel
- Department of Neurology, University Medical Center Amsterdam, location Academic Medical Center, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Department of Pathology, Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, the Netherlands
| | - G Joost Jöbsis
- Department of Neurology, University Medical Center Amsterdam, location Academic Medical Center, Amsterdam, the Netherlands
| | - Fred van Ruissen
- Department of Genome Analysis/Clinical Genetics, Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, the Netherlands; Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Karin Y van Spaendonck-Zwarts
- Department of Neurology, University Medical Center Amsterdam, location Academic Medical Center, Amsterdam, the Netherlands
| | - Marianne de Visser
- Department of Neurology, University Medical Center Amsterdam, location Academic Medical Center, Amsterdam, the Netherlands
| | - Frank Baas
- Department of Genome Analysis/Clinical Genetics, Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, the Netherlands; Dept Clinical Genetics, LUMC, Leiden, the Netherlands
| |
Collapse
|
20
|
Intracellular Citrate/acetyl-CoA flux and endoplasmic reticulum acetylation: Connectivity is the answer. Mol Metab 2022; 67:101653. [PMID: 36513219 PMCID: PMC9792894 DOI: 10.1016/j.molmet.2022.101653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Key cellular metabolites reflecting the immediate activity of metabolic enzymes as well as the functional metabolic state of intracellular organelles can act as powerful signal regulators to ensure the activation of homeostatic responses. The citrate/acetyl-CoA pathway, initially recognized for its role in intermediate metabolism, has emerged as a fundamental branch of this nutrient-sensing homeostatic response. Emerging studies indicate that fluctuations in acetyl-CoA availability within different cellular organelles and compartments provides substrate-level regulation of many biological functions. A fundamental aspect of these regulatory functions involves Nε-lysine acetylation. SCOPE OF REVIEW Here, we will examine the emerging regulatory functions of the citrate/acetyl-CoA pathway and the specific role of the endoplasmic reticulum (ER) acetylation machinery in the maintenance of intracellular crosstalk and homeostasis. These functions will be analyzed in the context of associated human diseases and specific mouse models of dysfunctional ER acetylation and citrate/acetyl-CoA flux. A primary objective of this review is to highlight the complex yet integrated response of compartment- and organelle-specific Nε-lysine acetylation to the intracellular availability and flux of acetyl-CoA, linking this important post-translational modification to cellular metabolism. MAJOR CONCLUSIONS The ER acetylation machinery regulates the proteostatic functions of the organelle as well as the metabolic crosstalk between different intracellular organelles and compartments. This crosstalk enables the cell to impart adaptive responses within the ER and the secretory pathway. However, it also enables the ER to impart adaptive responses within different cellular organelles and compartments. Defects in the homeostatic balance of acetyl-CoA flux and ER acetylation reflect different but converging disease states in humans as well as converging phenotypes in relevant mouse models. In conclusion, citrate and acetyl-CoA should not only be seen as metabolic substrates of intermediate metabolism but also as signaling molecules that direct functional adaptation of the cell to both intracellular and extracellular messages. Future discoveries in CoA biology and acetylation are likely to yield novel therapeutic approaches.
Collapse
|
21
|
刘 小, 段 晓, 张 朔, 孙 阿, 张 英, 樊 东. [Genetic distribution in Chinese patients with hereditary peripheral neuropathy]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2022; 54:874-883. [PMID: 36241230 PMCID: PMC9568373 DOI: 10.19723/j.issn.1671-167x.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To analyze the distribution characteristics of hereditary peripheral neuropathy (HPN) pathogenic genes in Chinese Han population, and to explore the potential pathogenesis and treatment prospects of HPN and related diseases. METHODS Six hundred and fifty-six index patients with HPN were enrolled in Peking University Third Hospital and China-Japan Friendship Hospital from January 2007 to May 2022. The PMP22 duplication and deletion mutations were screened and validated by multiplex ligation probe amplification technique. The next-generation sequencing gene panel or whole exome sequencing was used, and the suspected genes were validated by Sanger sequencing. RESULTS Charcot-Marie-Tooth (CMT) accounted for 74.3% (495/666) of the patients with HPN, of whom 69.1% (342/495) were genetically confirmed. The most common genes of CMT were PMP22 duplication, MFN2 and GJB1 mutations, which accounted for 71.3% (244/342) of the patients with genetically confirmed CMT. Hereditary motor neuropathy (HMN) accounted for 16.1% (107/666) of HPN, and 43% (46/107) of HPN was genetically confirmed. The most common genes of HMN were HSPB1, aminoacyl tRNA synthetases and SORD mutations, which accounted for 56.5% (26/46) of the patients with genetically confirmed HMN. Most genes associated with HMN could cause different phenotypes. HMN and CMT shared many genes (e.g. HSPB1, GARS, IGHMBP2). Some genes associated with dHMN-plus shared genes associated with amyotrophic lateral sclerosis (KIF5A, FIG4, DCTN1, SETX, VRK1), hereditary spastic paraplegia (KIF5A, ZFYVE26, BSCL2) and spinal muscular atrophy (MORC2, IGHMBP, DNAJB2), suggesting that HMN was a continuum rather than a distinct entity. Hereditary sensor and autosomal neuropathy (HSAN) accounted for a small proportion of 2.6% (17/666) in HPN. The most common pathogenic gene was SPTLC1 mutation. TTR was the main gene causing hereditary amyloid peripheral neuropathy. The most common types of gene mutations were p.A117S and p.V50M. The symptoms were characterized by late-onset and prominent autonomic nerve involvement. CONCLUSION CMT and HMN are the most common diseases of HPN. There is a large overlap between HMN and motor-CMT2 pathogenic genes, and some HMN pathogenic genes overlap with amyotrophic lateral sclerosis, hereditary spastic hemiplegia and spinal muscular atrophy, suggesting that there may be a potential common pathogenic pathway between different diseases.
Collapse
Affiliation(s)
- 小璇 刘
- 北京大学第三医院神经内科,北京 100191Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - 晓慧 段
- 中日友好医院神经内科,北京 100029Department of Neurology, China-Japan Friendship Hospital, Beijing 100029, China
| | - 朔 张
- 北京大学第三医院神经内科,北京 100191Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - 阿萍 孙
- 北京大学第三医院神经内科,北京 100191Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - 英爽 张
- 北京大学第三医院神经内科,北京 100191Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - 东升 樊
- 北京大学第三医院神经内科,北京 100191Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
22
|
Heterogeneous Clinical Phenotypes of dHMN Caused by Mutation in HSPB1 Gene: A Case Series. Biomolecules 2022; 12:biom12101382. [PMID: 36291591 PMCID: PMC9599773 DOI: 10.3390/biom12101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022] Open
Abstract
Mutations in HSPB1 are known to cause Charcot-Marie-Tooth disease type 2F (CMT2F) and distal hereditary motor neuropathy (dHMN). In this study, we presented three patients with mutation in HSPB1 who were diagnosed with dHMN. Proband 1 was a 14-year-old male with progressive bilateral lower limb weakness and walking difficulty for four years. Proband 2 was a 65-year-old male with chronic lower limb weakness and restless legs syndrome from the age of 51. Proband 3 was a 50-year-old female with progressive weakness, lower limbs atrophy from the age of 44. The nerve conduction studies (NCS) suggested axonal degeneration of the peripheral motor nerves and needle electromyography (EMG) revealed chronic neurogenic changes in probands. Open sural nerve biopsy for proband 2 and the mother of proband 1 showed mild to moderate loss of myelinated nerve fibers with some nerve fiber regeneration. A novel p.V97L in HSPB1 was identified in proband 3, the other two variants (p.P182A and p.R127W) in HSPB1 have been reported previously. The functional studies showed that expressing mutant p.V97L HSPB1 in SH-SY5Y cells displayed a decreased cell activity and increased apoptosis under stress condition. Our study expands the clinical phenotypic spectrum and etiological spectrum of HSPB1 mutation.
Collapse
|
23
|
Zhang X, Qiao Y, Han R, Gao Y, Yang X, Zhang Y, Wan Y, Yu W, Pan X, Xing J. A Charcot-Marie-Tooth-Causing Mutation in HSPB1 Decreases Cell Adaptation to Repeated Stress by Disrupting Autophagic Clearance of Misfolded Proteins. Cells 2022; 11:cells11182886. [PMID: 36139461 PMCID: PMC9496658 DOI: 10.3390/cells11182886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is the most common inherited neurodegenerative disorder with selective degeneration of peripheral nerves. Despite advances in identifying CMT-causing genes, the underlying molecular mechanism, particularly of selective degeneration of peripheral neurons remains to be elucidated. Since peripheral neurons are sensitive to multiple stresses, we hypothesized that daily repeated stress might be an essential contributor to the selective degeneration of peripheral neurons induced by CMT-causing mutations. Here, we mainly focused on the biological effects of the dominant missense mutation (S135F) in the 27-kDa small heat-shock protein HSPB1 under repeated heat shock. HSPB1S135F presented hyperactive binding to both α-tubulin and acetylated α-tubulin during repeated heat shock when compared with the wild type. The aberrant interactions with tubulin prevented microtubule-based transport of heat shock-induced misfolded proteins for the formation of perinuclear aggresomes. Furthermore, the transport of autophagosomes along microtubules was also blocked. These results indicate that the autophagy pathway was disrupted, leading to an accumulation of ubiquitinated protein aggregates and a significant decrease in cell adaptation to repeated stress. Our findings provide novel insights into the molecular mechanisms of HSPB1S135F-induced selective degeneration of peripheral neurons and perspectives for targeting autophagy as a promising therapeutic strategy for CMT neuropathy.
Collapse
Affiliation(s)
- Xuelian Zhang
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Yaru Qiao
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ronglin Han
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Yingjie Gao
- Department of Medicine Chemistry, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xun Yang
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ying Zhang
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ying Wan
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Wei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
- Correspondence: (J.X.); (X.P.); (W.Y.)
| | - Xianchao Pan
- Department of Medicine Chemistry, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Correspondence: (J.X.); (X.P.); (W.Y.)
| | - Juan Xing
- Department of Pathophysiology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
- Correspondence: (J.X.); (X.P.); (W.Y.)
| |
Collapse
|
24
|
Lu S, Hu J, Arogundade OA, Goginashvili A, Vazquez-Sanchez S, Diedrich JK, Gu J, Blum J, Oung S, Ye Q, Yu H, Ravits J, Liu C, Yates JR, Cleveland DW. Heat-shock chaperone HSPB1 regulates cytoplasmic TDP-43 phase separation and liquid-to-gel transition. Nat Cell Biol 2022; 24:1378-1393. [PMID: 36075972 PMCID: PMC9872726 DOI: 10.1038/s41556-022-00988-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 07/28/2022] [Indexed: 01/27/2023]
Abstract
While acetylated, RNA-binding-deficient TDP-43 reversibly phase separates within nuclei into complex droplets (anisosomes) comprised of TDP-43-containing liquid outer shells and liquid centres of HSP70-family chaperones, cytoplasmic aggregates of TDP-43 are hallmarks of multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Here we show that transient oxidative stress, proteasome inhibition or inhibition of the ATP-dependent chaperone activity of HSP70 provokes reversible cytoplasmic TDP-43 de-mixing and transition from liquid to gel/solid, independently of RNA binding or stress granules. Isotope labelling mass spectrometry was used to identify that phase-separated cytoplasmic TDP-43 is bound by the small heat-shock protein HSPB1. Binding is direct, mediated through TDP-43's RNA binding and low-complexity domains. HSPB1 partitions into TDP-43 droplets, inhibits TDP-43 assembly into fibrils, and is essential for disassembly of stress-induced TDP-43 droplets. A decrease in HSPB1 promotes cytoplasmic TDP-43 de-mixing and mislocalization. HSPB1 depletion was identified in spinal motor neurons of patients with ALS containing aggregated TDP-43. These findings identify HSPB1 to be a regulator of cytoplasmic TDP-43 phase separation and aggregation.
Collapse
Affiliation(s)
- Shan Lu
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA
- Ludwig Institute for Cancer Research, San Diego, CA, USA
| | - Jiaojiao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Alexander Goginashvili
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA
- Ludwig Institute for Cancer Research, San Diego, CA, USA
| | - Sonia Vazquez-Sanchez
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA
- Ludwig Institute for Cancer Research, San Diego, CA, USA
| | | | - Jinge Gu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jacob Blum
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Spencer Oung
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA
- Ludwig Institute for Cancer Research, San Diego, CA, USA
| | - Qiaozhen Ye
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA
| | - Haiyang Yu
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John Ravits
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - John R Yates
- The Scripps Research Institute, La Jolla, CA, USA
| | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, USA.
- Ludwig Institute for Cancer Research, San Diego, CA, USA.
- Department of Neurosciences, University of California, San Diego, CA, USA.
| |
Collapse
|
25
|
Holguin BA, Hildenbrand ZL, Bernal RA. Insights Into the Role of Heat Shock Protein 27 in the Development of Neurodegeneration. Front Mol Neurosci 2022; 15:868089. [PMID: 35431800 PMCID: PMC9005852 DOI: 10.3389/fnmol.2022.868089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/09/2022] [Indexed: 12/11/2022] Open
Abstract
Small heat shock protein 27 is a critically important chaperone, that plays a key role in several essential and varied physiological processes. These include thermotolerance, apoptosis, cytoskeletal dynamics, cell differentiation, protein folding, among others. Despite its relatively small size and intrinsically disordered termini, it forms large and polydisperse oligomers that are in equilibrium with dimers. This equilibrium is driven by transient interactions between the N-terminal region, the α-crystallin domain, and the C-terminal region. The continuous redistribution of binding partners results in a conformationally dynamic protein that allows it to adapt to different functions where substrate capture is required. However, the intrinsic disorder of the amino and carboxy terminal regions and subsequent conformational variability has made structural investigations challenging. Because heat shock protein 27 is critical for so many key cellular functions, it is not surprising that it also has been linked to human disease. Charcot-Marie-Tooth and distal hereditary motor neuropathy are examples of neurodegenerative disorders that arise from single point mutations in heat shock protein 27. The development of possible treatments, however, depends on our understanding of its normal function at the molecular level so we might be able to understand how mutations manifest as disease. This review will summarize recent reports describing investigations into the structurally elusive regions of Hsp27. Recent insights begin to provide the required context to explain the relationship between a mutation and the resulting loss or gain of function that leads to Charcot-Marie Tooth disease and distal hereditary motor neuropathy.
Collapse
|
26
|
Wu C, Xiang H, Chen R, Zheng Y, Zhu M, Chen S, Yu Y, Peng Y, Yu Y, Deng J, Zhou M, Hong D. Genetic spectrum in a cohort of patients with distal hereditary motor neuropathy. Ann Clin Transl Neurol 2022; 9:633-643. [PMID: 35297556 PMCID: PMC9082376 DOI: 10.1002/acn3.51543] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/08/2022] [Accepted: 02/26/2022] [Indexed: 12/26/2022] Open
Abstract
Background Distal hereditary motor neuropathy (dHMN) is a heterogeneous group of diseases characterized by exclusive degeneration of peripheral motor nerves, while only 20.0–47.8% of dHMN patients are genetically identified. Recently, GGC expansion in the 5’UTR of NOTCH2NLC has been associated with dHMN. Accordingly, short tandem repeat (STR) should be further explored in genetically unsolved patients with dHMN. Methods A total of 128 patients from 90 unrelated families were clinically diagnosed as dHMN, and underwent a comprehensively genetic screening. Skin biopsies were conducted with routine protocols. Results Most patients showed chronic distal weakness of lower limbs (121/128), while 20 patients initially had asymmetrical involvements, 14 had subclinical sensory abnormalities, 11 had pyramidal impairments, five had cerebellar disturbance, and four had hyperCKmia. The rate of genetic detection was achieved in 36.7% (33/90), and the rate increased to 46.7% (42/90) if patients with variants uncertain significance were included. The most common causative genes included chaperone‐related genes (8/33, 24.2%), tRNA synthetase genes (4/33, 12.1%), and cytoskeleton‐related genes (4/33, 12.1%). Additionally, two dominant inherited families were attributed to abnormal expansion of GGC repeats in the 5‘UTR of NOTCH2NLC; and a patient with dHMN and cerebellar symptoms had CAG repeat expansion in the ATXN2 gene. Skin biopsy from patients with GGC expansion in NOTCH2NLC revealed typical intranuclear inclusions on histological and ultrastructural examinations. Interpretations This study further extends the genetic heterogeneity of dHMN. Given some dHMN patients may be associated with nucleotides repeat expansion, STR screening is necessary to perform in genetically unsolved patients.
Collapse
Affiliation(s)
- Chengsi Wu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Haijie Xiang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Ran Chen
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yilei Zheng
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Min Zhu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.,Department of Medical Genetics, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Shuyun Chen
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yanyan Yu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.,Department of Medical Genetics, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yun Peng
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.,Department of Medical Genetics, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yaqing Yu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Meihong Zhou
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.,Department of Medical Genetics, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.,Department of Neurology, Peking University People Hospital, Beijing, 100044, China
| |
Collapse
|
27
|
Genetic and Clinical Studies of Peripheral Neuropathies with Three Small Heat Shock Protein Gene Variants in Korea. Genes (Basel) 2022; 13:genes13030462. [PMID: 35328016 PMCID: PMC8949397 DOI: 10.3390/genes13030462] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 01/25/2023] Open
Abstract
Small heat shock proteins (sHSPs) are ATP-independent chaperones that help correct the folding of denatured proteins and protect cells from stress. Mutations in HSPB1, HSPB8, and HSPB3 are implicated in inherited peripheral neuropathies (IPNs), such as Charcot-Marie-Tooth disease type 2 (CMT2) and distal hereditary motor neuropathies (dHMN). This study, using whole exome sequencing or targeted gene sequencing, identified 9 pathogenic or likely pathogenic variants in these three sHSP genes from 11 Korean IPN families. Most variants were located in the evolutionally well conserved α-crystallin domain, except for p.P182S and p.S187L in HSPB1. As an atypical case, a patient with dHMN2 showed two compound heterozygous variants of p.R127Q and p.Y142H in HSPB1, suggesting a putative case of recessive inheritance, which requires additional research to confirm. Three HSPB8 variants were located in the p.K141 residue, which seemed to be a mutational hot spot. There were no significant differences between patient groups, which divided by sHSP genes for clinical symptoms such as onset age, severity, and nerve conduction. Early-onset patients showed a tendency of slightly decreased sensory nerve conduction values compared with late-onset patients. As a first Korean IPN cohort study examining sHSP genes, these results will, we believe, be helpful for molecular diagnosis and care of patients with CMT2 and dHMN.
Collapse
|
28
|
Tedesco B, Cristofani R, Ferrari V, Cozzi M, Rusmini P, Casarotto E, Chierichetti M, Mina F, Galbiati M, Piccolella M, Crippa V, Poletti A. Insights on Human Small Heat Shock Proteins and Their Alterations in Diseases. Front Mol Biosci 2022; 9:842149. [PMID: 35281256 PMCID: PMC8913478 DOI: 10.3389/fmolb.2022.842149] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
The family of the human small Heat Shock Proteins (HSPBs) consists of ten members of chaperones (HSPB1-HSPB10), characterized by a low molecular weight and capable of dimerization and oligomerization forming large homo- or hetero-complexes. All HSPBs possess a highly conserved centrally located α-crystallin domain and poorly conserved N- and C-terminal domains. The main feature of HSPBs is to exert cytoprotective functions by preserving proteostasis, assuring the structural maintenance of the cytoskeleton and acting in response to cellular stresses and apoptosis. HSPBs take part in cell homeostasis by acting as holdases, which is the ability to interact with a substrate preventing its aggregation. In addition, HSPBs cooperate in substrates refolding driven by other chaperones or, alternatively, promote substrate routing to degradation. Notably, while some HSPBs are ubiquitously expressed, others show peculiar tissue-specific expression. Cardiac muscle, skeletal muscle and neurons show high expression levels for a wide variety of HSPBs. Indeed, most of the mutations identified in HSPBs are associated to cardiomyopathies, myopathies, and motor neuropathies. Instead, mutations in HSPB4 and HSPB5, which are also expressed in lens, have been associated with cataract. Mutations of HSPBs family members encompass base substitutions, insertions, and deletions, resulting in single amino acid substitutions or in the generation of truncated or elongated proteins. This review will provide an updated overview of disease-related mutations in HSPBs focusing on the structural and biochemical effects of mutations and their functional consequences.
Collapse
Affiliation(s)
- B. Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - R. Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - P. Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - E. Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - F. Mina
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - M. Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - V. Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - A. Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
- *Correspondence: A. Poletti,
| |
Collapse
|
29
|
Sainio MT, Rasila T, Molchanova SM, Järvilehto J, Torregrosa-Muñumer R, Harjuhaahto S, Pennonen J, Huber N, Herukka SK, Haapasalo A, Zetterberg H, Taira T, Palmio J, Ylikallio E, Tyynismaa H. Neurofilament Light Regulates Axon Caliber, Synaptic Activity, and Organelle Trafficking in Cultured Human Motor Neurons. Front Cell Dev Biol 2022; 9:820105. [PMID: 35237613 PMCID: PMC8883324 DOI: 10.3389/fcell.2021.820105] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/28/2021] [Indexed: 11/27/2022] Open
Abstract
Neurofilament light (NFL) is one of the proteins forming multimeric neuron-specific intermediate filaments, neurofilaments, which fill the axonal cytoplasm, establish caliber growth, and provide structural support. Dominant missense mutations and recessive nonsense mutations in the neurofilament light gene (NEFL) are among the causes of Charcot-Marie-Tooth (CMT) neuropathy, which affects the peripheral nerves with the longest axons. We previously demonstrated that a neuropathy-causing homozygous nonsense mutation in NEFL led to the absence of NFL in patient-specific neurons. To understand the disease-causing mechanisms, we investigate here the functional effects of NFL loss in human motor neurons differentiated from induced pluripotent stem cells (iPSC). We used genome editing to generate NEFL knockouts and compared them to patient-specific nonsense mutants and isogenic controls. iPSC lacking NFL differentiated efficiently into motor neurons with normal axon growth and regrowth after mechanical axotomy and contained neurofilaments. Electrophysiological analysis revealed that motor neurons without NFL fired spontaneous and evoked action potentials with similar characteristics as controls. However, we found that, in the absence of NFL, human motor neurons 1) had reduced axonal caliber, 2) the amplitude of miniature excitatory postsynaptic currents (mEPSC) was decreased, 3) neurofilament heavy (NFH) levels were reduced and no compensatory increases in other filament subunits were observed, and 4) the movement of mitochondria and to a lesser extent lysosomes was increased. Our findings elaborate the functional roles of NFL in human motor neurons. NFL is not only a structural protein forming neurofilaments and filling the axonal cytoplasm, but our study supports the role of NFL in the regulation of synaptic transmission and organelle trafficking. To rescue the NFL deficiency in the patient-specific nonsense mutant motor neurons, we used three drugs, amlexanox, ataluren (PTC-124), and gentamicin to induce translational read-through or inhibit nonsense-mediated decay. However, the drugs failed to increase the amount of NFL protein to detectable levels and were toxic to iPSC-derived motor neurons.
Collapse
Affiliation(s)
- Markus T. Sainio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tiina Rasila
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Svetlana M. Molchanova
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Julius Järvilehto
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rubén Torregrosa-Muñumer
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sandra Harjuhaahto
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jana Pennonen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nadine Huber
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanna-Kaisa Herukka
- Department of Neurology, Kuopio University Hospital, Kuopio, Finland
- Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, Hong Kong SAR, China
| | - Tomi Taira
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, Department of Veterinary Biosciences for Electrophysiology, University of Helsinki, Helsinki, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Johanna Palmio
- Neuromuscular Research Center, Tampere University Hospital and Tampere University, Tampere, Finland
| | - Emil Ylikallio
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Henna Tyynismaa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
30
|
Karamanos TK, Clore GM. Large Chaperone Complexes Through the Lens of Nuclear Magnetic Resonance Spectroscopy. Annu Rev Biophys 2022; 51:223-246. [PMID: 35044800 DOI: 10.1146/annurev-biophys-090921-120150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Molecular chaperones are the guardians of the proteome inside the cell. Chaperones recognize and bind unfolded or misfolded substrates, thereby preventing further aggregation; promoting correct protein folding; and, in some instances, even disaggregating already formed aggregates. Chaperones perform their function by means of an array of weak protein-protein interactions that take place over a wide range of timescales and are therefore invisible to structural techniques dependent upon the availability of highly homogeneous samples. Nuclear magnetic resonance (NMR) spectroscopy, however, is ideally suited to study dynamic, rapidly interconverting conformational states and protein-protein interactions in solution, even if these involve a high-molecular-weight component. In this review, we give a brief overview of the principles used by chaperones to bind their client proteins and describe NMR methods that have emerged as valuable tools to probe chaperone-substrate and chaperone-chaperone interactions. We then focus on a few systems for which the application of these methods has greatly increased our understanding of the mechanisms underlying chaperone functions. Expected final online publication date for the Annual Review of Biophysics, Volume 51 is May 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Theodoros K Karamanos
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom;
| | - G Marius Clore
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
31
|
Park NY, Kwak G, Doo HM, Kim HJ, Jang SY, Lee YI, Choi BO, Hong YB. Farnesol Ameliorates Demyelinating Phenotype in a Cellular and Animal Model of Charcot-Marie-Tooth Disease Type 1A. Curr Issues Mol Biol 2021; 43:2011-2021. [PMID: 34889893 PMCID: PMC8928981 DOI: 10.3390/cimb43030138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/29/2021] [Accepted: 11/10/2021] [Indexed: 01/05/2023] Open
Abstract
Charcot-Marie-Tooth disease (CMT) is a genetically heterogeneous disease affecting the peripheral nervous system that is caused by either the demyelination of Schwann cells or degeneration of the peripheral axon. Currently, there are no treatment options to improve the degeneration of peripheral nerves in CMT patients. In this research, we assessed the potency of farnesol for improving the demyelinating phenotype using an animal model of CMT type 1A. In vitro treatment with farnesol facilitated myelin gene expression and ameliorated the myelination defect caused by PMP22 overexpression, the major causative gene in CMT. In vivo administration of farnesol enhanced the peripheral neuropathic phenotype, as shown by rotarod performance in a mouse model of CMT1A. Electrophysiologically, farnesol-administered CMT1A mice exhibited increased motor nerve conduction velocity and compound muscle action potential compared with control mice. The number and diameter of myelinated axons were also increased by farnesol treatment. The expression level of myelin protein zero (MPZ) was increased, while that of the demyelination marker, neural cell adhesion molecule (NCAM), was reduced by farnesol administration. These data imply that farnesol is efficacious in ameliorating the demyelinating phenotype of CMT, and further elucidation of the underlying mechanisms of farnesol’s effect on myelination might provide a potent therapeutic strategy for the demyelinating type of CMT.
Collapse
Affiliation(s)
- Na-Young Park
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan 49201, Korea;
| | - Geon Kwak
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea; (G.K.); (H.-M.D.); (H.-J.K.)
| | - Hyun-Myung Doo
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea; (G.K.); (H.-M.D.); (H.-J.K.)
| | - Hye-Jin Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea; (G.K.); (H.-M.D.); (H.-J.K.)
| | - So-Young Jang
- Departments of Biochemistry, College of Medicine, Dong-A University, Busan 49201, Korea;
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
| | - Byung-Ok Choi
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea; (G.K.); (H.-M.D.); (H.-J.K.)
- Samsung Medical Center, Department of Neurology, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Correspondence: (B.-O.C.); (Y.-B.H.); Tel.: +82-2-3410-1296 (B.-O.C.); +82-51-240-2762 (Y.-B.H.); Fax: +82-3410-0052 (B.-O.C.); +82-51-240-2971 (Y.-B.H.)
| | - Young-Bin Hong
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan 49201, Korea;
- Departments of Biochemistry, College of Medicine, Dong-A University, Busan 49201, Korea;
- Correspondence: (B.-O.C.); (Y.-B.H.); Tel.: +82-2-3410-1296 (B.-O.C.); +82-51-240-2762 (Y.-B.H.); Fax: +82-3410-0052 (B.-O.C.); +82-51-240-2971 (Y.-B.H.)
| |
Collapse
|
32
|
Bosco L, Falzone YM, Previtali SC. Animal Models as a Tool to Design Therapeutical Strategies for CMT-like Hereditary Neuropathies. Brain Sci 2021; 11:1237. [PMID: 34573256 PMCID: PMC8465478 DOI: 10.3390/brainsci11091237] [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: 07/25/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
Since ancient times, animal models have provided fundamental information in medical knowledge. This also applies for discoveries in the field of inherited peripheral neuropathies (IPNs), where they have been instrumental for our understanding of nerve development, pathogenesis of neuropathy, molecules and pathways involved and to design potential therapies. In this review, we briefly describe how animal models have been used in ancient medicine until the use of rodents as the prevalent model in present times. We then travel along different examples of how rodents have been used to improve our understanding of IPNs. We do not intend to describe all discoveries and animal models developed for IPNs, but just to touch on a few arbitrary and paradigmatic examples, taken from our direct experience or from literature. The idea is to show how strategies have been developed to finally arrive to possible treatments for IPNs.
Collapse
Affiliation(s)
| | | | - Stefano Carlo Previtali
- Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.B.); (Y.M.F.)
| |
Collapse
|
33
|
Gonçalves CC, Sharon I, Schmeing TM, Ramos CHI, Young JC. The chaperone HSPB1 prepares protein aggregates for resolubilization by HSP70. Sci Rep 2021; 11:17139. [PMID: 34429462 PMCID: PMC8384840 DOI: 10.1038/s41598-021-96518-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/11/2021] [Indexed: 01/22/2023] Open
Abstract
In human cells under stress conditions, misfolded polypeptides can form potentially cytotoxic insoluble aggregates. To eliminate aggregates, the HSP70 chaperone machinery extracts and resolubilizes polypeptides for triage to refolding or degradation. Yeast and bacterial chaperones of the small heat-shock protein (sHSP) family can bind substrates at early stages of misfolding, during the aggregation process. The co-aggregated sHSPs then facilitate downstream disaggregation by HSP70. Because it is unknown whether a human sHSP has this activity, we investigated the disaggregation role of human HSPB1. HSPB1 co-aggregated with unfolded protein substrates, firefly luciferase and mammalian lactate dehydrogenase. The co-aggregates formed with HSPB1 were smaller and more regularly shaped than those formed in its absence. Importantly, co-aggregation promoted the efficient disaggregation and refolding of the substrates, led by HSP70. HSPB1 itself was also extracted during disaggregation, and its homo-oligomerization ability was not required. Therefore, we propose that a human sHSP is an integral part of the chaperone network for protein disaggregation.
Collapse
Affiliation(s)
- Conrado C Gonçalves
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Room 900, Montreal, QC, H3G 1Y6, Canada
| | - Itai Sharon
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Room 457, Montreal, QC, H3G 0B1, Canada
| | - T Martin Schmeing
- Department of Biochemistry, McGill University, 3649 Promenade Sir William Osler, Room 457, Montreal, QC, H3G 0B1, Canada
| | - Carlos H I Ramos
- Institute of Chemistry, University of Campinas (UNICAMP), Campinas, SP, 13083-970, Brazil
| | - Jason C Young
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Room 900, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
34
|
Maung MT, Carlson A, Olea-Flores M, Elkhadragy L, Schachtschneider KM, Navarro-Tito N, Padilla-Benavides T. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J 2021; 35:e21810. [PMID: 34390520 DOI: 10.1096/fj.202100273rr] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022]
Abstract
Copper (Cu) is an essential micronutrient required for the activity of redox-active enzymes involved in critical metabolic reactions, signaling pathways, and biological functions. Transporters and chaperones control Cu ion levels and bioavailability to ensure proper subcellular and systemic Cu distribution. Intensive research has focused on understanding how mammalian cells maintain Cu homeostasis, and how molecular signals coordinate Cu acquisition and storage within organs. In humans, mutations of genes that regulate Cu homeostasis or facilitate interactions with Cu ions lead to numerous pathologic conditions. Malfunctions of the Cu+ -transporting ATPases ATP7A and ATP7B cause Menkes disease and Wilson disease, respectively. Additionally, defects in the mitochondrial and cellular distributions and homeostasis of Cu lead to severe neurodegenerative conditions, mitochondrial myopathies, and metabolic diseases. Cu has a dual nature in carcinogenesis as a promotor of tumor growth and an inducer of redox stress in cancer cells. Cu also plays role in cancer treatment as a component of drugs and a regulator of drug sensitivity and uptake. In this review, we provide an overview of the current knowledge of Cu metabolism and transport and its relation to various human pathologies.
Collapse
Affiliation(s)
- May T Maung
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Alyssa Carlson
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Monserrat Olea-Flores
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | - Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Napoleon Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | | |
Collapse
|
35
|
Fu X, Ezemaduka AN, Lu X, Chang Z. The Caenorhabditis elegans 12-kDa small heat shock proteins with little in vitro chaperone activity play crucial roles for its dauer formation, longevity, and reproduction. Protein Sci 2021; 30:2170-2182. [PMID: 34272907 DOI: 10.1002/pro.4160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/31/2023]
Abstract
Small heat shock proteins (sHSPs) are known to exhibit in vitro chaperone activity by suppressing the aggregation of misfolded proteins. The 12-kDa sHSPs (Hsp12s) subfamily members from Caenorhabditis elegans, including Hsp12.2, Hsp12.3, and Hsp12.6, however, are devoid of such chaperone activity, and their in vivo functions are poorly understood. Here we verified that Hsp12.1, similar to its homologs Hsp12.2, Hsp12.3, and Hsp12.6, hardly exhibited any chaperone activity. Strikingly, we demonstrated that these Hsp12s seem to play crucial physiological roles in C. elegans, for suppressing dauer formation and promoting both longevity and reproduction. A unique sHSP gene from Filarial nematode worm Brugia malayi was identified such that it encodes two products, one as a full-length Hsp12.6 protein and the other one having an N-terminal arm of normal length but lacks the C-terminal extension. This gene may represent an intermediate form in evolution from a common sHSP to a Hsp12. Together, our study offers insights on what biological functions the chaperone-defective sHSPs may exhibit and also implicates an evolutionary scenario for the unique Hsp12s subfamily.
Collapse
Affiliation(s)
- Xinmiao Fu
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou City, Fujian Province, China.,School of Life Sciences, Peking University, Beijing, China
| | - Anastasia N Ezemaduka
- School of Life Sciences, Peking University, Beijing, China.,Key Laboratory of Wetland Ecology and Environment, Northeast institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun, China
| | - Xinping Lu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Zengyi Chang
- School of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
36
|
Van Lent J, Verstraelen P, Asselbergh B, Adriaenssens E, Mateiu L, Verbist C, De Winter V, Eggermont K, Van Den Bosch L, De Vos WH, Timmerman V. Induced pluripotent stem cell-derived motor neurons of CMT type 2 patients reveal progressive mitochondrial dysfunction. Brain 2021; 144:2471-2485. [PMID: 34128983 PMCID: PMC8418338 DOI: 10.1093/brain/awab226] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022] Open
Abstract
Axonal Charcot-Marie-Tooth neuropathies (CMT type 2) are caused by inherited mutations in various genes functioning in different pathways. The type of genes and multiplicity of mutations reflect the clinical and genetic heterogeneity in CMT2 disease, which complicates the diagnosis and has halted therapy development. Here, we used CMT2 patient-derived pluripotent stem cells (iPSCs) to identify common hallmarks of axonal degeneration shared by different CMT2 subtypes. We compared the cellular phenotypes of neurons differentiated from CMT2 patient iPSCs with those from healthy controls and a CRISPR/Cas9-corrected isogenic line. Our results demonstrate neurite network alterations along with extracellular electrophysiological abnormalities in the differentiated motor neurons. Progressive deficits in mitochondrial and lysosomal trafficking, as well as in mitochondrial morphology, were observed in all CMT2 patient lines. Differentiation of the same CMT2 iPSC-lines into peripheral sensory neurons, only gave rise to cellular phenotypes in subtypes with sensory involvement, supporting the notion that some gene mutations predominantly affect motor neurons. We revealed a common mitochondrial dysfunction in CMT2-derived motor neurons, supported by alterations in the expression pattern and oxidative phosphorylation, which could be recapitulated in the sciatic nerve tissue of a symptomatic mouse model. Inhibition of a dual leucine zipper kinase (DLK) could partially ameliorate the mitochondrial disease phenotypes in CMT2 subtypes. Altogether, our data reveals shared cellular phenotypes across different CMT2 subtypes and suggests that targeting such common pathomechanisms could allow the development of a uniform treatment for CMT2.
Collapse
Affiliation(s)
- Jonas Van Lent
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, 2610, Belgium.,Neurogenetics Laboratory, Institute Born Bunge, Antwerp, 2610, Belgium
| | - Peter Verstraelen
- Laboratory of Cell Biology & Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, 2610, Belgium
| | - Bob Asselbergh
- Neuromics Support Facility, VIB Center for Molecular Neurology, VIB, Antwerp, 2610, Belgium.,Neuromics Support Facility, Department of Biomedical Sciences, University of Antwerp, Antwerp, 2610, Belgium
| | - Elias Adriaenssens
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, 2610, Belgium.,Neurogenetics Laboratory, Institute Born Bunge, Antwerp, 2610, Belgium
| | - Ligia Mateiu
- Neuromics Support Facility, VIB Center for Molecular Neurology, VIB, Antwerp, 2610, Belgium
| | - Christophe Verbist
- Laboratory of Molecular Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp, 2610, Belgium
| | - Vicky De Winter
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, 2610, Belgium.,Neurogenetics Laboratory, Institute Born Bunge, Antwerp, 2610, Belgium
| | - Kristel Eggermont
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, KU Leuven - University of Leuven, Leuven, 3000, Belgium.,VIB-Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, 3000, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, KU Leuven - University of Leuven, Leuven, 3000, Belgium.,VIB-Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, 3000, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology & Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, 2610, Belgium
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, 2610, Belgium.,Neurogenetics Laboratory, Institute Born Bunge, Antwerp, 2610, Belgium
| |
Collapse
|
37
|
Greenbaum L, Ben‐David M, Nikitin V, Gera O, Barel O, Hersalis‐Eldar A, Shamash J, Shimshoviz N, Reznik‐Wolf H, Shohat M, Dominissini D, Pras E, Dori A. Early and late manifestations of neuropathy due to HSPB1 mutation in the Jewish Iranian population. Ann Clin Transl Neurol 2021; 8:1260-1268. [PMID: 33973728 PMCID: PMC8164855 DOI: 10.1002/acn3.51362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Mutations in the HSPB1 gene are associated with a distal hereditary motor neuropathy type 2 (dHMN2) or Charcot-Marie-Tooth disease type 2F (CMT2F), usually with autosomal dominant inheritance. This study aimed to describe the phenotype of the HSPB1 c.407G>T (p.Arg136Leu) mutation at early and late stages of the disease course. METHODS We identified this mutation (previously reported in patients from Italy) in a heterozygous state, among 14 individuals from eight families of Jewish Iranian descent. The clinical, electrophysiological and ultrasonographic features were evaluated during early (less than 5 years, N = 9) or late disease course (N = 5). RESULTS The majority of subjects were males with a mean age at onset of 43.4 years (range 21-67). Common initial symptoms were gait imbalance, distal (often asymmetric) lower limb weakness and feet numbness. Neurological examination in early disease course showed distal lower extremity weakness in nearly all cases, and absent Achilles tendon reflex in about half. A minority had distal loss of pain, vibration or position sensation. These findings were more prevalent in late disease stage. Electrodiagnostic studies demonstrated a length-dependent axonal motor neuropathy, with typical preferential involvement of the tibial nerve. Muscle ultrasound showed a corresponding length-dependent increase of homogeneous echo-intensity, most noticeably in the gastrocnemius. One patient had a dual diagnosis of CMT2F and CMT2W. INTERPRETATION The HSPB1 c.407G>G (p.Arg136Leu) mutation causes an adult-onset, predominantly motor, axonal neuropathy in individuals of Jewish Iranian descent. Variable manifestations are noticed, and sensory involvement is more prominent in prolonged disease duration.
Collapse
Affiliation(s)
- Lior Greenbaum
- The Danek Gertner Institute of Human GeneticsSheba Medical CenterTel HashomerIsrael
- The Joseph Sagol Neuroscience CenterSheba Medical CenterTel HashomerIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Merav Ben‐David
- Department of NeurologySheba Medical CenterTel HashomerIsrael
| | - Vera Nikitin
- Department of NeurologySheba Medical CenterTel HashomerIsrael
| | - Orna Gera
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Department of NeurologySheba Medical CenterTel HashomerIsrael
| | - Ortal Barel
- The Genomic UnitSheba Cancer Research Center, Sheba Medical CenterTel HashomerIsrael
- Wohl Institute of Translational MedicineSheba Medical CenterTel HashomerIsrael
| | | | - Jana Shamash
- The Danek Gertner Institute of Human GeneticsSheba Medical CenterTel HashomerIsrael
| | - Noam Shimshoviz
- The Genomic UnitSheba Cancer Research Center, Sheba Medical CenterTel HashomerIsrael
- Wohl Institute of Translational MedicineSheba Medical CenterTel HashomerIsrael
| | - Haike Reznik‐Wolf
- The Danek Gertner Institute of Human GeneticsSheba Medical CenterTel HashomerIsrael
| | - Mordechai Shohat
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- The Genomic UnitSheba Cancer Research Center, Sheba Medical CenterTel HashomerIsrael
- Wohl Institute of Translational MedicineSheba Medical CenterTel HashomerIsrael
| | - Dan Dominissini
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- The Genomic UnitSheba Cancer Research Center, Sheba Medical CenterTel HashomerIsrael
- Wohl Institute of Translational MedicineSheba Medical CenterTel HashomerIsrael
| | - Elon Pras
- The Danek Gertner Institute of Human GeneticsSheba Medical CenterTel HashomerIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Amir Dori
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Department of NeurologySheba Medical CenterTel HashomerIsrael
| |
Collapse
|
38
|
Shan Q, Ma F, Wei J, Li H, Ma H, Sun P. Physiological Functions of Heat Shock Proteins. Curr Protein Pept Sci 2021; 21:751-760. [PMID: 31713482 DOI: 10.2174/1389203720666191111113726] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/24/2019] [Accepted: 09/21/2019] [Indexed: 01/03/2023]
Abstract
Heat shock proteins (HSPs) are molecular chaperones involved in a variety of life activities. HSPs function in the refolding of misfolded proteins, thereby contributing to the maintenance of cellular homeostasis. Heat shock factor (HSF) is activated in response to environmental stresses and binds to heat shock elements (HSEs), promoting HSP translation and thus the production of high levels of HSPs to prevent damage to the organism. Here, we summarize the role of molecular chaperones as anti-heat stress molecules and their involvement in immune responses and the modulation of apoptosis. In addition, we review the potential application of HSPs to cancer therapy, general medicine, and the treatment of heart disease.
Collapse
Affiliation(s)
- Qiang Shan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Fengtao Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Jingya Wei
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Hongyang Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Hui Ma
- Beijing Sunlon Livestock Development Co., Ltd, Beijing, China
| | - Peng Sun
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| |
Collapse
|
39
|
Kho J, Pham PC, Kwon S, Huang AY, Rivers JP, Wang H, Ecroyd H, Donald WA, McAlpine SR. De Novo Design, Synthesis, and Mechanistic Evaluation of Short Peptides That Mimic Heat Shock Protein 27 Activity. ACS Med Chem Lett 2021; 12:713-719. [PMID: 34055216 DOI: 10.1021/acsmedchemlett.0c00609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/01/2021] [Indexed: 01/17/2023] Open
Abstract
We report the first small molecule peptides based on the N-terminal sequence of heat shock protein 27 (Hsp27, gene HSPB1) that demonstrates chaperone-like activity. The peptide, comprising the SWDPF sequence located at Hsp27's amino (N)-terminal domain, directly regulates protein aggregation events, maintaining the disaggregated state of the model protein, citrate synthase. While traditional inhibitors of protein aggregation act via regulation of a protein that facilitates aggregation or disaggregation, our molecules are the first small peptides between 5 and 8 amino acids in length that are based on the N-terminus of Hsp27 and directly control protein aggregation. The presented strategy showcases a new approach for developing small peptides that control protein aggregation in proteins with high aggregate levels, making them a useful approach in developing new drugs.
Collapse
Affiliation(s)
- Jessica Kho
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - P. Chi Pham
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Suhyeon Kwon
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Alana Y. Huang
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Joel P. Rivers
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Huixin Wang
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Heath Ecroyd
- Department of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
| | - W. Alexander Donald
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Shelli R. McAlpine
- School of Chemistry, University of California Irvine, Irvine, California 92697, United States
| |
Collapse
|
40
|
Pathomechanisms of ALS8: altered autophagy and defective RNA binding protein (RBP) homeostasis due to the VAPB P56S mutation. Cell Death Dis 2021; 12:466. [PMID: 33972508 PMCID: PMC8110809 DOI: 10.1038/s41419-021-03710-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 02/03/2023]
Abstract
Mutations in RNA binding proteins (RBPs) and in genes regulating autophagy are frequent causes of familial amyotrophic lateral sclerosis (fALS). The P56S mutation in vesicle-associated membrane protein-associated protein B (VAPB) leads to fALS (ALS8) and spinal muscular atrophy (SMA). While VAPB is primarily involved in the unfolded protein response (UPR), vesicular trafficking and in initial steps of the autophagy pathway, the effect of mutant P56S-VAPB on autophagy regulation in connection with RBP homeostasis has not been explored yet. Examining the muscle biopsy of our index ALS8 patient of European origin revealed globular accumulations of VAPB aggregates co-localised with autophagy markers LC3 and p62 in partially atrophic and atrophic muscle fibres. In line with this skin fibroblasts obtained from the same patient showed accumulation of P56S-VAPB aggregates together with LC3 and p62. Detailed investigations of autophagic flux in cell culture models revealed that P56S-VAPB alters both initial and late steps of the autophagy pathway. Accordingly, electron microscopy complemented with live cell imaging highlighted the impaired fusion of accumulated autophagosomes with lysosomes in cells expressing P56S-VAPB. Consistent with these observations, neuropathological studies of brain and spinal cord of P56S-VAPB transgenic mice revealed signs of neurodegeneration associated with altered protein quality control and defective autophagy. Autophagy and RBP homeostasis are interdependent, as demonstrated by the cytoplasmic mis-localisation of several RBPs including pTDP-43, FUS, Matrin 3 which often sequestered with P56S-VAPB aggregates both in cell culture and in the muscle biopsy of the ALS8 patient. Further confirming the notion that aggregation of the RBPs proceeds through the stress granule (SG) pathway, we found persistent G3BP- and TIAR1-positive SGs in P56S-VAPB expressing cells as well as in the ALS8 patient muscle biopsy. We conclude that P56S-VAPB-ALS8 involves a cohesive pathomechanism of aberrant RBP homeostasis together with dysfunctional autophagy.
Collapse
|
41
|
Abati E, Magri S, Meneri M, Manenti G, Velardo D, Balistreri F, Pisciotta C, Saveri P, Bresolin N, Comi GP, Ronchi D, Pareyson D, Taroni F, Corti S. Charcot-Marie-Tooth disease type 2F associated with biallelic HSPB1 mutations. Ann Clin Transl Neurol 2021; 8:1158-1164. [PMID: 33943041 PMCID: PMC8108422 DOI: 10.1002/acn3.51364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 12/21/2022] Open
Abstract
Objective This work aims to expand knowledge regarding the genetic spectrum of HSPB1‐related diseases. HSPB1 is a gene encoding heat shock protein 27, and mutations in HSPB1 have been identified as the cause of axonal Charcot–Marie–Tooth (CMT) disease type 2F and distal hereditary motor neuropathy (dHMN). Methods Two patients with axonal sensorimotor neuropathy underwent detailed clinical examinations, neurophysiological studies, and next‐generation sequencing with subsequent bioinformatic prioritization of genetic variants and in silico analysis of the likely causal mutation. Results The HSPB1 p.S135F and p.R136L mutations were identified in homozygosis in the two affected individuals. Both mutations affect the highly conserved alpha‐crystallin domain and have been previously described as the cause of severe CMT2F/dHMN, showing a strictly dominant inheritance pattern. Interpretation Thus, we report for the first time two cases of biallelic HSPB1 p.S135F and p.R136L mutations in two families.
Collapse
Affiliation(s)
- Elena Abati
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, University of Milan, Milan, Italy
| | - Stefania Magri
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Megi Meneri
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Manenti
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, University of Milan, Milan, Italy
| | - Daniele Velardo
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Balistreri
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Pisciotta
- Rare Neurodegenerative and Neurometabolic Diseases Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Saveri
- Rare Neurodegenerative and Neurometabolic Diseases Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Nereo Bresolin
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, University of Milan, Milan, Italy.,Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo Pietro Comi
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, University of Milan, Milan, Italy.,Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario Ronchi
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, University of Milan, Milan, Italy
| | - Davide Pareyson
- Rare Neurodegenerative and Neurometabolic Diseases Unit, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Franco Taroni
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, University of Milan, Milan, Italy.,Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
42
|
Reid Alderson T, Adriaenssens E, Asselbergh B, Pritišanac I, Van Lent J, Gastall HY, Wälti MA, Louis JM, Timmerman V, Baldwin AJ, LP Benesch J. A weakened interface in the P182L variant of HSP27 associated with severe Charcot-Marie-Tooth neuropathy causes aberrant binding to interacting proteins. EMBO J 2021; 40:e103811. [PMID: 33644875 PMCID: PMC8047445 DOI: 10.15252/embj.2019103811] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/10/2021] [Accepted: 01/14/2021] [Indexed: 01/18/2023] Open
Abstract
HSP27 is a human molecular chaperone that forms large, dynamic oligomers and functions in many aspects of cellular homeostasis. Mutations in HSP27 cause Charcot-Marie-Tooth (CMT) disease, the most common inherited disorder of the peripheral nervous system. A particularly severe form of CMT disease is triggered by the P182L mutation in the highly conserved IxI/V motif of the disordered C-terminal region, which interacts weakly with the structured core domain of HSP27. Here, we observed that the P182L mutation disrupts the chaperone activity and significantly increases the size of HSP27 oligomers formed in vivo, including in motor neurons differentiated from CMT patient-derived stem cells. Using NMR spectroscopy, we determined that the P182L mutation decreases the affinity of the HSP27 IxI/V motif for its own core domain, leaving this binding site more accessible for other IxI/V-containing proteins. We identified multiple IxI/V-bearing proteins that bind with higher affinity to the P182L variant due to the increased availability of the IxI/V-binding site. Our results provide a mechanistic basis for the impact of the P182L mutation on HSP27 and suggest that the IxI/V motif plays an important, regulatory role in modulating protein-protein interactions.
Collapse
Affiliation(s)
- T Reid Alderson
- Chemistry Research LaboratoryUniversity of OxfordOxfordUK
- Laboratory of Chemical PhysicsNational Institutes of HealthBethesdaMDUSA
- Present address:
Department of BiochemistryTorontoONCanada
| | - Elias Adriaenssens
- Peripheral Neuropathy Research GroupDepartment of Biomedical SciencesInstitute Born BungeUniversity of AntwerpAntwerpenBelgium
| | - Bob Asselbergh
- Neuromics Support FacilityVIB Center for Molecular NeurologyVIBAntwerpenBelgium
- Neuromics Support Facility, Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | - Iva Pritišanac
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoONCanada
| | - Jonas Van Lent
- Peripheral Neuropathy Research GroupDepartment of Biomedical SciencesInstitute Born BungeUniversity of AntwerpAntwerpenBelgium
| | | | - Marielle A Wälti
- Laboratory of Chemical PhysicsNational Institutes of HealthBethesdaMDUSA
| | - John M Louis
- Laboratory of Chemical PhysicsNational Institutes of HealthBethesdaMDUSA
| | - Vincent Timmerman
- Peripheral Neuropathy Research GroupDepartment of Biomedical SciencesInstitute Born BungeUniversity of AntwerpAntwerpenBelgium
| | | | | |
Collapse
|
43
|
Shemesh N, Jubran J, Dror S, Simonovsky E, Basha O, Argov C, Hekselman I, Abu-Qarn M, Vinogradov E, Mauer O, Tiago T, Carra S, Ben-Zvi A, Yeger-Lotem E. The landscape of molecular chaperones across human tissues reveals a layered architecture of core and variable chaperones. Nat Commun 2021; 12:2180. [PMID: 33846299 PMCID: PMC8042005 DOI: 10.1038/s41467-021-22369-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The sensitivity of the protein-folding environment to chaperone disruption can be highly tissue-specific. Yet, the organization of the chaperone system across physiological human tissues has received little attention. Through computational analyses of large-scale tissue transcriptomes, we unveil that the chaperone system is composed of core elements that are uniformly expressed across tissues, and variable elements that are differentially expressed to fit with tissue-specific requirements. We demonstrate via a proteomic analysis that the muscle-specific signature is functional and conserved. Core chaperones are significantly more abundant across tissues and more important for cell survival than variable chaperones. Together with variable chaperones, they form tissue-specific functional networks. Analysis of human organ development and aging brain transcriptomes reveals that these functional networks are established in development and decline with age. In this work, we expand the known functional organization of de novo versus stress-inducible eukaryotic chaperones into a layered core-variable architecture in multi-cellular organisms.
Collapse
Affiliation(s)
- Netta Shemesh
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Juman Jubran
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Shiran Dror
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Eyal Simonovsky
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omer Basha
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Chanan Argov
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Idan Hekselman
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Mehtap Abu-Qarn
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ekaterina Vinogradov
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omry Mauer
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tatiana Tiago
- Centre for Neuroscience and Nanotechnology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Serena Carra
- Centre for Neuroscience and Nanotechnology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anat Ben-Zvi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
44
|
Sheehan BK, Orefice NS, Peng Y, Shapiro SL, Puglielli L. ATG9A regulates proteostasis through reticulophagy receptors FAM134B and SEC62 and folding chaperones CALR and HSPB1. iScience 2021; 24:102315. [PMID: 33870132 PMCID: PMC8042170 DOI: 10.1016/j.isci.2021.102315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/18/2021] [Accepted: 03/12/2021] [Indexed: 12/01/2022] Open
Abstract
The acetylation of ATG9A within the endoplasmic reticulum (ER) lumen regulates the induction of reticulophagy. ER acetylation is ensured by AT-1/SLC33A1, a membrane transporter that maintains the cytosol-to-ER flux of acetyl-CoA. Defective AT-1 activity, as caused by heterozygous/homozygous mutations and gene duplication events, results in severe disease phenotypes. Here, we show that although the acetylation of ATG9A occurs in the ER lumen, the induction of reticulophagy requires ATG9A to engage FAM134B and SEC62 on the cytosolic side of the ER. To address this conundrum, we resolved the ATG9A interactome in two mouse models of AT-1 dysregulation: AT-1 sTg, a model of systemic AT-1 overexpression with hyperacetylation of ATG9A, and AT-1S113R/+, a model of AT-1 haploinsufficiency with hypoacetylation of ATG9A. We identified CALR and HSPB1 as two ATG9A partners that regulate the induction of reticulophagy as a function of ATG9A acetylation and discovered that ATG9A associates with several proteins that maintain ER proteostasis. The ATG9A-FAM134B and ATG9A-SEC62 interaction requires specific structural features Opposite Ca++-binding EF hands regulate ATG9A-FAM134B interaction HSBP1 and CALR regulate ATG9A-mediated induction of reticulophagy Many of the proteins that ensure ER proteostasis display spatial vicinity/cross talk
Collapse
Affiliation(s)
- Brendan K Sheehan
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.,Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nicola S Orefice
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.,Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yajing Peng
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.,Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Samantha L Shapiro
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.,Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.,Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI 53705, USA.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
45
|
Keller N, Paketci C, Altmueller J, Fuhrmann N, Wunderlich G, Schrank B, Unver O, Yilmaz S, Boostani R, Karimiani EG, Motameny S, Thiele H, Nürnberg P, Maroofian R, Yis U, Wirth B, Karakaya M. Genomic variants causing mitochondrial dysfunction are common in hereditary lower motor neuron disease. Hum Mutat 2021; 42:460-472. [PMID: 33600046 DOI: 10.1002/humu.24181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/29/2020] [Accepted: 02/10/2021] [Indexed: 11/08/2022]
Abstract
Hereditary lower motor neuron diseases (LMND) other than 5q-spinal muscular atrophy (5q-SMA) can be classified according to affected muscle groups. Proximal and distal forms of non-5q-SMA represent a clinically and genetically heterogeneous spectrum characterized by significant overlaps with axonal forms of Charcot-Marie-Tooth (CMT) disease. A consensus for the best approach to molecular diagnosis needs to be reached, especially in light of continuous novel gene discovery and falling costs of next-generation sequencing (NGS). We performed exome sequencing (ES) in 41 families presenting with non-5q-SMA or axonal CMT, 25 of which had undergone a previous negative neuromuscular disease (NMD) gene panel analysis. The total diagnostic yield of ES was 41%. Diagnostic success in the cohort with a previous NMD-panel analysis was significantly extended by ES, primarily due to novel gene associated-phenotypes and uncharacteristic phenotypic presentations. We recommend early ES for individuals with hereditary LMND presenting uncharacteristic or significantly overlapping features. As mitochondrial dysfunction was the underlying pathomechanism in 47% of the solved individuals, we highlight the sensitivity of the anterior horn cell and peripheral nerve to mitochondrial imbalance as well as the necessity to screen for mitochondrial disorders in individuals presenting predominant lower motor neuron symptoms.
Collapse
Affiliation(s)
- Natalie Keller
- Institute of Human Genetics and Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Center for Rare Diseases Cologne, University Hospital Cologne, Cologne, Germany
| | - Cem Paketci
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Janine Altmueller
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Nico Fuhrmann
- Institute of Human Genetics and Institute of Genetics, University of Cologne, Cologne, Germany
| | - Gilbert Wunderlich
- Center for Rare Diseases Cologne, University Hospital Cologne, Cologne, Germany
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Bertold Schrank
- Department of Neurology, DKD HELIOS Kliniken, Wiesbaden, Germany
| | - Olcay Unver
- Department of Pediatric Neurology, Marmara University, Istanbul, Turkey
| | - Sanem Yilmaz
- Department of Pediatric Neurology, Ege University, Izmir, Turkey
| | - Reza Boostani
- Department of Neurology, Ghaem Hospital, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Ghayoor Karimiani
- Molecular and Clinical Sciences Institute, St. George's University of London, Cranmer Terrace, London, UK
| | - Susanne Motameny
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Holger Thiele
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Reza Maroofian
- Molecular and Clinical Sciences Institute, St. George's University of London, Cranmer Terrace, London, UK
| | - Uluc Yis
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Brunhilde Wirth
- Institute of Human Genetics and Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Center for Rare Diseases Cologne, University Hospital Cologne, Cologne, Germany
| | - Mert Karakaya
- Institute of Human Genetics and Institute of Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Center for Rare Diseases Cologne, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
46
|
Beijer D, Baets J. The expanding genetic landscape of hereditary motor neuropathies. Brain 2021; 143:3540-3563. [PMID: 33210134 DOI: 10.1093/brain/awaa311] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Hereditary motor neuropathies are clinically and genetically diverse disorders characterized by length-dependent axonal degeneration of lower motor neurons. Although currently as many as 26 causal genes are known, there is considerable missing heritability compared to other inherited neuropathies such as Charcot-Marie-Tooth disease. Intriguingly, this genetic landscape spans a discrete number of key biological processes within the peripheral nerve. Also, in terms of underlying pathophysiology, hereditary motor neuropathies show striking overlap with several other neuromuscular and neurological disorders. In this review, we provide a current overview of the genetic spectrum of hereditary motor neuropathies highlighting recent reports of novel genes and mutations or recent discoveries in the underlying disease mechanisms. In addition, we link hereditary motor neuropathies with various related disorders by addressing the main affected pathways of disease divided into five major processes: axonal transport, tRNA aminoacylation, RNA metabolism and DNA integrity, ion channels and transporters and endoplasmic reticulum.
Collapse
Affiliation(s)
- Danique Beijer
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium
| | - Jonathan Baets
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium.,Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Belgium
| |
Collapse
|
47
|
Chen J, Liu X, Xu Y, Fan D. [Rare variants of HSPB1 are probably associated with amyotrophic lateral sclerosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:75-78. [PMID: 33509756 DOI: 10.12122/j.issn.1673-4254.2021.01.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To explore the association between rare HSPB1 variants and amyotrophic lateral sclerosis (ALS). METHODS We performed next-generation sequencing for 166 Chinese ALS patients to screen for possible pathogenic rare variants of HSPB1. The control individuals were obtained from 1000 Genome Project and an in-house whole-exome sequencing database. The Sequence Kernel Association Test (SKAT) and the SKAT-optimal test (SKAT-O) were used to identify the association between rare HSPB1 variants and ALS. RESULTS We identified 3 possible pathogenic rare variants of HSPB1 (all were missenses), including c.379C>T (p.R127W), c.446A>C (p.D149A) and c.451A>C (p.T151P). Compared with 1000 Genome Project, SKAT p=3.61×10-7 and SKAT-O p=1.62×10-6; while compared with the in-house database, SKAT p=9.99×10-4, SKAT-O p= 1.80×10-3. We analyzed the phenotypes of rare HSPB1 variant carriers and found no specific clinical characteristics associated with these variants. CONCLUSIONS Rare variants of HSPB1 are probably associated with the pathogenesis of ALS.
Collapse
Affiliation(s)
- Junyi Chen
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Xiangyi Liu
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
48
|
The Neurochaperonopathies: Anomalies of the Chaperone System with Pathogenic Effects in Neurodegenerative and Neuromuscular Disorders. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11030898] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The chaperone (or chaperoning) system (CS) constitutes molecular chaperones, co-chaperones, and chaperone co-factors, interactors and receptors, and its canonical role is protein quality control. A malfunction of the CS may cause diseases, known as the chaperonopathies. These are caused by qualitatively and/or quantitatively abnormal molecular chaperones. Since the CS is ubiquitous, chaperonopathies are systemic, affecting various tissues and organs, playing an etiologic-pathogenic role in diverse conditions. In this review, we focus on chaperonopathies involved in the pathogenic mechanisms of diseases of the central and peripheral nervous systems: the neurochaperonopathies (NCPs). Genetic NCPs are linked to pathogenic variants of chaperone genes encoding, for example, the small Hsp, Hsp10, Hsp40, Hsp60, and CCT-BBS (chaperonin-containing TCP-1- Bardet–Biedl syndrome) chaperones. Instead, the acquired NCPs are associated with malfunctional chaperones, such as Hsp70, Hsp90, and VCP/p97 with aberrant post-translational modifications. Awareness of the chaperonopathies as the underlying primary or secondary causes of disease will improve diagnosis and patient management and open the possibility of investigating and developing chaperonotherapy, namely treatment with the abnormal chaperone as the main target. Positive chaperonotherapy would apply in chaperonopathies by defect, i.e., chaperone insufficiency, and consist of chaperone replacement or boosting, whereas negative chaperonotherapy would be pertinent when a chaperone actively participates in the initiation and progression of the disease and must be blocked and eliminated.
Collapse
|
49
|
Bomont P. The dazzling rise of neurofilaments: Physiological functions and roles as biomarkers. Curr Opin Cell Biol 2021; 68:181-191. [PMID: 33454158 DOI: 10.1016/j.ceb.2020.10.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022]
Abstract
In the last two years, neurofilaments (NFs) have become one of the most blazing topics in clinical neuroscience. NFs are major cytoskeletal constituents of neurons, can be detected in body fluids, and have recently emerged as universal biomarkers of neuronal injury and neurological diseases. This review will examine the evolving landscape of NFs, from their specific cellular functions within neurons to their broad clinical value as biomarkers. Particular attention will be given to the dynamic nature of the NF network and its novel roles in microtubule regulation, neurotransmission, and nanomedicine. Building from the initial evidence of causative mutations in NF genes in Charcot-Marie-Tooth diseases, the latest advances at the frontiers of basic and clinical sciences have expanded the scope and relevance of NFs for human health remarkably and have poised to fuel innovation in cell biology and neuroscience.
Collapse
Affiliation(s)
- Pascale Bomont
- ERC team, INMG, INSERM U1217, CNRS UMR5310, University of Lyon 1, University of Lyon, Lyon, France.
| |
Collapse
|
50
|
Moutaoufik MT, Tanguay RM. Analysis of insect nuclear small heat shock proteins and interacting proteins. Cell Stress Chaperones 2021; 26:265-274. [PMID: 32888179 PMCID: PMC7736433 DOI: 10.1007/s12192-020-01156-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022] Open
Abstract
The small heat shock proteins (sHsps) are a ubiquitous family of ATP-independent stress proteins found in all domains of life. Drosophila melanogaster Hsp27 (DmHsp27) is the only known nuclear sHsp in insect. Here analyzing sequences from HMMER, we identified 56 additional insect sHsps with conserved arginine-rich nuclear localization signal (NLS) in the N-terminal region. At this time, the exact role of nuclear sHsps remains unknown. DmHsp27 protein-protein interaction analysis from iRefIndex database suggests that this protein, in addition to a putative role of molecular chaperone, is likely involved in other nuclear processes (i.e., chromatin remodeling and transcription). Identification of DmHsp27 interactors should provide key insights on the cellular and molecular functions of this nuclear chaperone.
Collapse
Affiliation(s)
- Mohamed Taha Moutaoufik
- Lab of Cell & Developmental Genetics, Department of Cellular and Molecular Biology, Medical Biochemistry & Pathology, Medical School, Université Laval, Quebec, G1K 7P4, Canada
- Department of Biochemistry, University of Regina, Regina, SK, S4S 0A2, Canada
| | - Robert M Tanguay
- Lab of Cell & Developmental Genetics, Department of Cellular and Molecular Biology, Medical Biochemistry & Pathology, Medical School, Université Laval, Quebec, G1K 7P4, Canada.
| |
Collapse
|