1
|
Pelaez MC, Fiore F, Larochelle N, Dabbaghizadeh A, Comaduran MF, Arbour D, Minotti S, Marcadet L, Semaan M, Robitaille R, Nalbantoglu JN, Sephton CF, Durham HD. Reversal of cognitive deficits in FUS R521G amyotrophic lateral sclerosis mice by arimoclomol and a class I histone deacetylase inhibitor independent of heat shock protein induction. Neurotherapeutics 2024; 21:e00388. [PMID: 38972779 DOI: 10.1016/j.neurot.2024.e00388] [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: 04/08/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 07/09/2024] Open
Abstract
Protein misfolding and mislocalization are common to both familial and sporadic forms of amyotrophic lateral sclerosis (ALS). Maintaining proteostasis through induction of heat shock proteins (HSP) to increase chaperoning capacity is a rational therapeutic strategy in the treatment of ALS. However, the threshold for upregulating stress-inducible HSPs remains high in neurons, presenting a therapeutic obstacle. This study used mouse models expressing the ALS variants FUSR521G or SOD1G93A to follow up on previous work in cultured motor neurons showing varied effects of the HSP co-inducer, arimoclomol, and class I histone deacetylase (HDAC) inhibitors on HSP expression depending on the ALS variant being expressed. As in cultured neurons, neither expression of the transgene nor drug treatments induced expression of HSPs in cortex, spinal cord or muscle of FUSR521G mice, indicating suppression of the heat shock response. Nonetheless, arimoclomol, and RGFP963, restored performance on cognitive tests and improved cortical dendritic spine densities. In SOD1G93A mice, multiple HSPs were upregulated in hindlimb skeletal muscle, but not in lumbar spinal cord with the exception of HSPB1 associated with astrocytosis. Drug treatments improved contractile force but reduced the increase in HSPs in muscle rather than facilitating their expression. The data point to mechanisms other than amplification of the heat shock response underlying recovery of cognitive function in ALS-FUS mice by arimoclomol and class I HDAC inhibition and suggest potential benefits in counteracting cognitive impairment in ALS, frontotemporal dementia and related disorders.
Collapse
Affiliation(s)
- Mari Carmen Pelaez
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC Canada.
| | - Frédéric Fiore
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Nancy Larochelle
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Afrooz Dabbaghizadeh
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Mario Fernández Comaduran
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Danielle Arbour
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Sandra Minotti
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Laetitia Marcadet
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC Canada.
| | - Martine Semaan
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Richard Robitaille
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Josephine N Nalbantoglu
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC Canada.
| | - Heather D Durham
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| |
Collapse
|
2
|
Percio A, Cicchinelli M, Masci D, Summo M, Urbani A, Greco V. Oxidative Cysteine Post Translational Modifications Drive the Redox Code Underlying Neurodegeneration and Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2024; 13:883. [PMID: 39199129 PMCID: PMC11351139 DOI: 10.3390/antiox13080883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
Redox dysregulation, an imbalance between oxidants and antioxidants, is crucial in the pathogenesis of various neurodegenerative diseases. Within this context, the "redoxome" encompasses the network of redox molecules collaborating to maintain cellular redox balance and signaling. Among these, cysteine-sensitive proteins are fundamental for this homeostasis. Due to their reactive thiol groups, cysteine (Cys) residues are particularly susceptible to oxidative post-translational modifications (PTMs) induced by free radicals (reactive oxygen, nitrogen, and sulfur species) which profoundly affect protein functions. Cys-PTMs, forming what is referred to as "cysteinet" in the redox proteome, are essential for redox signaling in both physiological and pathological conditions, including neurodegeneration. Such modifications significantly influence protein misfolding and aggregation, key hallmarks of neurodegenerative diseases such as Alzheimer's, Parkinson's, and notably, amyotrophic lateral sclerosis (ALS). This review aims to explore the complex landscape of cysteine PTMs in the cellular redox environment, elucidating their impact on neurodegeneration at protein level. By investigating specific cysteine-sensitive proteins and the regulatory networks involved, particular emphasis is placed on the link between redox dysregulation and ALS, highlighting this pathology as a prime example of a neurodegenerative disease wherein such redox dysregulation is a distinct hallmark.
Collapse
Affiliation(s)
- Anna Percio
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| | - Michela Cicchinelli
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| | - Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
| | - Mariagrazia Summo
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| | - Viviana Greco
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| |
Collapse
|
3
|
Li Z, Cai X. Baicalein targets STMN1 to inhibit the progression of nasopharyngeal carcinoma via regulating the Wnt/β-catenin pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:3003-3013. [PMID: 38317500 DOI: 10.1002/tox.24173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 01/24/2024] [Accepted: 01/27/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUNDS Nasopharyngeal carcinoma is a common malignancy in the head and neck. Baicalein has been reported to exert the anticancer effects on various cancers. In this study, our aim was to explore the function of baicalein in the development of nasopharyngeal carcinoma and further investigate the potential underlying mechanisms. METHODS Cell Counting Kit (CCK)-8 assay, EdU assay, sphere formation assay, flow cytometry, and transwell invasion assay were conducted to determine cell proliferation, stemness, apoptosis, and invasion, respectively. Western blot was performed to examine the protein levels of PCNA, MMP9, STMN1, β-catenin, and Wnt3A. The mRNA level of STMN1 was assessed using real-time quantitative polymerase chain reaction (RT-qPCR). Xenograft tumor model was carried out to evaluate the effects of baicalein on tumor growth in vivo. Immunohistochemistry (IHC) assay was used to detect the levels of PCNA, MMP9, and STMN1 in tumor tissues from mice. RESULTS Baicalein significantly induced cell apoptosis and impeded cell proliferation, invasion, and stemness of nasopharyngeal carcinoma cells. STMN1 was highly expressed in nasopharyngeal carcinoma, and baicalein could directly downregulate STMN1 expression. STMN1 knockdown hampered the progression of nasopharyngeal carcinoma cells. Moreover, the effects of baicalein on cell proliferation, stemness, invasion, and apoptosis in nasopharyngeal carcinoma cells were harbored by STMN1 overexpression. Baicalein regulated STMN1 to inhibit the activation of the Wnt/β-catenin pathway. SKL2001, an agonist of the Wnt/β-catenin pathway, could reverse the effects of STMN1 knockdown on the progression of nasopharyngeal carcinoma. In addition, baicalein markedly impeded tumor growth in vivo. CONCLUSION Baicalein regulated the STMN1/Wnt/β-catenin pathway to restrain the development of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Zheng Li
- Department of Otolaryngology, Nanyang First People's Hospital, Nanyang, China
| | - Xiaohang Cai
- The Second Department of Cardiology, Nanyang First People's Hospital, Nanyang, China
| |
Collapse
|
4
|
Sp1-Mediated Prdx6 Upregulation Leads to Clasmatodendrosis by Increasing Its aiPLA2 Activity in the CA1 Astrocytes in Chronic Epilepsy Rats. Antioxidants (Basel) 2022; 11:antiox11101883. [PMID: 36290607 PMCID: PMC9598987 DOI: 10.3390/antiox11101883] [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: 08/02/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/29/2022] Open
Abstract
Clasmatodendrosis is an autophagic astroglial degeneration (a non-apoptotic (type II) programmed cell death) whose underlying mechanisms are fully understood. Peroxiredoxin-6 (Prdx6), the “non-selenium glutathione peroxidase (NSGPx)”, is the only member of the 1-cysteine peroxiredoxin family. Unlike the other Prdx family, Prdx6 has multiple functions as glutathione peroxidase (GPx) and acidic calcium-independent phospholipase (aiPLA2). The present study shows that Prdx6 was upregulated in CA1 astrocytes in chronic epilepsy rats. 2-Cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) and N-acetylcysteine (NAC, a precursor of glutathione) ameliorated clasmatodendrosis accompanied by reduced Prdx6 level in CA1 astrocytes. Specificity protein 1 (Sp1) expression was upregulated in CA1 astrocyte, which was inhibited by mithramycin A (MMA). MMA alleviated clasmatodendrosis and Prdx6 upregulation. Sp1 expression was also downregulated by CDDO-Me and NAC. Furthermore, 1-hexadecyl-3-(trifluoroethgl)-sn-glycerol-2 phosphomethanol (MJ33, a selective inhibitor of aiPLA2 activity of Prdx6) attenuated clasmatodendrosis without affecting Prdx6 expression. All chemicals shortened spontaneous seizure duration but not seizure frequency and behavioral seizure severity in chronic epilepsy rats. Therefore, our findings suggest that Sp1 activation may upregulate Prdx6, whose aiPLA2 activity would dominate over GPx activity in CA1 astrocytes and may lead to prolonged seizure activity due to autophagic astroglial degeneration.
Collapse
|
5
|
Kulkarni R, Thakur A, Kumar H. Microtubule Dynamics Following Central and Peripheral Nervous System Axotomy. ACS Chem Neurosci 2022; 13:1358-1369. [PMID: 35451811 DOI: 10.1021/acschemneuro.2c00189] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Disturbance in the neuronal network leads to instability in the microtubule (MT) railroad of axons, causing hindrance in the intra-axonal transport and making it difficult to re-establish the broken network. Peripheral nervous system (PNS) neurons can stabilize their MTs, leading to the formation of regeneration-promoting structures called "growth cones". However, central nervous system (CNS) neurons lack this intrinsic reparative capability and, instead, form growth-incompetent structures called "retraction bulbs", which have a disarrayed MT network. It is evident from various studies that although axonal regeneration depends on both cell-extrinsic and cell-intrinsic factors, any therapy that aims at axonal regeneration ultimately converges onto MTs. Understanding the neuronal MT dynamics will help develop effective therapeutic strategies in diseases where the MT network gets disrupted, such as spinal cord injury, traumatic brain injury, multiple sclerosis, and amyotrophic lateral sclerosis. It is also essential to know the factors that aid or inhibit MT stabilization. In this review, we have discussed the MT dynamics postaxotomy in the CNS and PNS, and factors that can directly influence MT stability in various diseases.
Collapse
Affiliation(s)
- Riya Kulkarni
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Akshata Thakur
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- National Institute of Pharmaceutical Education and Research, Ahmedabad, Opposite Air Force Station, Palaj, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
6
|
PTEN loss promotes oncogenic function of STMN1 via PI3K/AKT pathway in lung cancer. Sci Rep 2021; 11:14318. [PMID: 34253824 PMCID: PMC8275769 DOI: 10.1038/s41598-021-93815-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 06/30/2021] [Indexed: 12/25/2022] Open
Abstract
Among all cancer types, lung cancer has already become the leading cause of cancer-related death around the world. The molecular mechanism understanding this development is still needed to be improved to treat lung cancer. Stathmin (STMN1) was initially identified as a cytoplasmic protein phosphorylated responding to cell signal and controlled cell physiological processes. The dysregulation of STMN1 is found in various kinds of tumors. However, the molecular mechanism of STMN1 regulating lung cancer is still unclear. Here, we found that STMN1 was overexpressed in lung cancer tissues and associated with worse survival rates of lung cancer patients. Inhibition of STMN1 suppressed lung cancer cell growth, migration and invasion, and promoted drug sensitivity. Moreover, PTEN loss promoted STMN1 expression via PI3K/AKT pathway. PTEN loss ameliorated the inhibition of cell growth, migration and invasion, and drug sensitivity induced by STMN1 knockdown in lung cancer. The high expression of STMN1 was negatively correlated with the low expression of PTEN in lung cancer specimens. Overall, our work demonstrated that PTEN regulated the oncogenic function of STMN1 in lung cancer.
Collapse
|
7
|
Liao J, Zhang Y, Chen X, Zhang J. The Roles of Peroxiredoxin 6 in Brain Diseases. Mol Neurobiol 2021; 58:4348-4364. [PMID: 34013449 DOI: 10.1007/s12035-021-02427-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023]
Abstract
Peroxiredoxin 6 (PRDX6), the only mammalian 1-Cys member of the peroxiredoxins (PRDXs) family, has multiple functions of glutathione peroxidase (Gpx) activity, acidic calcium-independent phospholipase (aiPLA2) activity, and lysophosphatidylcholine acyl transferase (LPCAT) activity. It has been documented to be involved in redox homeostasis, phospholipid turnover, glycolipid metabolism, and cellular signaling. Here, we reviewed the characteristics of the available Prdx6 genetic mouse models and the research progresses made with regard to PRDX6 in neuropsychiatric disorders, including neurodegenerative diseases, brain aging, stroke, neurotrauma, gliomas, major depressive disorder, drug addiction, post-traumatic stress disorder, and schizophrenia. The present review highlights the important roles of PRDX6 in neuropsychiatric disorders and may provide novel insights for the development of effective pharmacological treatments and genetic therapies.
Collapse
Affiliation(s)
- Jiangfeng Liao
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350005, Fujian, China
| | - Yusi Zhang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350005, Fujian, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China. .,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350005, Fujian, China.
| | - Jing Zhang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China. .,Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350005, Fujian, China.
| |
Collapse
|
8
|
Jagaraj CJ, Parakh S, Atkin JD. Emerging Evidence Highlighting the Importance of Redox Dysregulation in the Pathogenesis of Amyotrophic Lateral Sclerosis (ALS). Front Cell Neurosci 2021; 14:581950. [PMID: 33679322 PMCID: PMC7929997 DOI: 10.3389/fncel.2020.581950] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
The cellular redox state, or balance between cellular oxidation and reduction reactions, serves as a vital antioxidant defence system that is linked to all important cellular activities. Redox regulation is therefore a fundamental cellular process for aerobic organisms. Whilst oxidative stress is well described in neurodegenerative disorders including amyotrophic lateral sclerosis (ALS), other aspects of redox dysfunction and their contributions to pathophysiology are only just emerging. ALS is a fatal neurodegenerative disease affecting motor neurons, with few useful treatments. Hence there is an urgent need to develop more effective therapeutics in the future. Here, we discuss the increasing evidence for redox dysregulation as an important and primary contributor to ALS pathogenesis, which is associated with multiple disease mechanisms. Understanding the connection between redox homeostasis, proteins that mediate redox regulation, and disease pathophysiology in ALS, may facilitate a better understanding of disease mechanisms, and lead to the design of better therapeutic strategies.
Collapse
Affiliation(s)
- Cyril Jones Jagaraj
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sonam Parakh
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Julie D Atkin
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
9
|
Szeliga M. Peroxiredoxins in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:E1203. [PMID: 33265993 PMCID: PMC7761365 DOI: 10.3390/antiox9121203] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022] Open
Abstract
Substantial evidence indicates that oxidative/nitrosative stress contributes to the neurodegenerative diseases. Peroxiredoxins (PRDXs) are one of the enzymatic antioxidant mechanisms neutralizing reactive oxygen/nitrogen species. Since mammalian PRDXs were identified 30 years ago, their significance was long overshadowed by the other well-studied ROS/RNS defense systems. An increasing number of studies suggests that these enzymes may be involved in the neurodegenerative process. This article reviews the current knowledge on the expression and putative roles of PRDXs in neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and dementia with Lewy bodies, multiple sclerosis, amyotrophic lateral sclerosis and Huntington's disease.
Collapse
Affiliation(s)
- Monika Szeliga
- Mossakowski Medical Research Centre, Department of Neurotoxicology, Polish Academy of Sciences, 5 Pawinskiego Street, 02-106 Warsaw, Poland
| |
Collapse
|
10
|
Miller JH, Das V. Potential for Treatment of Neurodegenerative Diseases with Natural Products or Synthetic Compounds that Stabilize Microtubules. Curr Pharm Des 2020; 26:4362-4372. [DOI: 10.2174/1381612826666200621171302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 05/08/2020] [Indexed: 01/04/2023]
Abstract
No effective therapeutics to treat neurodegenerative diseases exist, despite significant attempts to find
drugs that can reduce or rescue the debilitating symptoms of tauopathies such as Alzheimer’s disease, Parkinson’s
disease, frontotemporal dementia, amyotrophic lateral sclerosis, or Pick’s disease. A number of in vitro and in
vivo models exist for studying neurodegenerative diseases, including cell models employing induced-pluripotent
stem cells, cerebral organoids, and animal models of disease. Recent research has focused on microtubulestabilizing
agents, either natural products or synthetic compounds that can prevent the axonal destruction caused
by tau protein pathologies. Although promising results have come from animal model studies using brainpenetrant
natural product microtubule-stabilizing agents, such as paclitaxel analogs that can access the brain,
epothilones B and D, and other synthetic compounds such as davunetide or the triazolopyrimidines, early clinical
trials in humans have been disappointing. This review aims to summarize the research that has been carried out in
this area and discuss the potential for the future development of an effective microtubule stabilizing drug to treat
neurodegenerative disease.
Collapse
Affiliation(s)
- John H. Miller
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington 6140, New Zealand
| | - Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Hněvotínska 5, 77900 Olomouc, Czech Republic
| |
Collapse
|
11
|
Villalón E, Kline RA, Smith CE, Lorson ZC, Osman EY, O'Day S, Murray LM, Lorson CL. AAV9-Stathmin1 gene delivery improves disease phenotype in an intermediate mouse model of spinal muscular atrophy. Hum Mol Genet 2020; 28:3742-3754. [PMID: 31363739 DOI: 10.1093/hmg/ddz188] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a devastating infantile genetic disorder caused by the loss of survival motor neuron (SMN) protein that leads to premature death due to loss of motor neurons and muscle atrophy. The approval of an antisense oligonucleotide therapy for SMA was an important milestone in SMA research; however, effective next-generation therapeutics will likely require combinatorial SMN-dependent therapeutics and SMN-independent disease modifiers. A recent cross-disease transcriptomic analysis identified Stathmin-1 (STMN1), a tubulin-depolymerizing protein, as a potential disease modifier across different motor neuron diseases, including SMA. Here, we investigated whether viral-based delivery of STMN1 decreased disease severity in a well-characterized SMA mouse model. Intracerebroventricular delivery of scAAV9-STMN1 in SMA mice at P2 significantly increased survival and weight gain compared to untreated SMA mice without elevating Smn levels. scAAV9-STMN1 improved important hallmarks of disease, including motor function, NMJ pathology and motor neuron cell preservation. Furthermore, scAAV9-STMN1 treatment restored microtubule networks and tubulin expression without affecting tubulin stability. Our results show that scAAV9-STMN1 treatment improves SMA pathology possibly by increasing microtubule turnover leading to restored levels of stable microtubules. Overall, these data demonstrate that STMN1 can significantly reduce the SMA phenotype independent of restoring SMN protein and highlight the importance of developing SMN-independent therapeutics for the treatment of SMA.
Collapse
Affiliation(s)
- E Villalón
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - R A Kline
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - C E Smith
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Z C Lorson
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - E Y Osman
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - S O'Day
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| | - L M Murray
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - C L Lorson
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
12
|
Peroxiredoxin 5 Silencing Sensitizes Dopaminergic Neuronal Cells to Rotenone via DNA Damage-Triggered ATM/p53/PUMA Signaling-Mediated Apoptosis. Cells 2019; 9:cells9010022. [PMID: 31861721 PMCID: PMC7016837 DOI: 10.3390/cells9010022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Peroxiredoxins (Prxs) are a family of thioredoxin peroxidases. Accumulating evidence suggests that changes in the expression of Prxs may be involved in neurodegenerative diseases pathology. However, the expression and function of Prxs in Parkinson’s disease (PD) remains unclear. Here, we showed that Prx5 was the most downregulated of the six Prx subtypes in dopaminergic (DA) neurons in rotenone-induced cellular and rat models of PD, suggesting possible roles in regulating their survival. Depletion of Prx5 sensitized SH-SY5Y DA neuronal cells to rotenone-induced apoptosis. The extent of mitochondrial membrane potential collapse, cytochrome c release, and caspase activation was increased by Prx5 loss. Furthermore, Prx5 knockdown enhanced the induction of PUMA by rotenone through a p53-dependent mechanism. Using RNA interference approaches, we demonstrated that the p53/PUMA signaling was essential for Prx5 silencing-exacerbated mitochondria-driven apoptosis. Additionally, downregulation of Prx5 augmented rotenone-induced DNA damage manifested as induction of phosphorylated histone H2AX (γ-H2AX) and activation of ataxia telangiectasia mutated (ATM) kinase. The pharmacological inactivation of ATM revealed that ATM was integral to p53 activation by DNA damage. These findings provided a novel link between Prx5 and DNA damage-triggered ATM/p53/PUMA signaling in a rotenone-induced PD model. Thus, Prx5 might play an important role in protection against rotenone-induced DA neurodegeneration.
Collapse
|
13
|
Golgi Fragmentation in Neurodegenerative Diseases: Is There a Common Cause? Cells 2019; 8:cells8070748. [PMID: 31331075 PMCID: PMC6679019 DOI: 10.3390/cells8070748] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/17/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
In most mammalian cells, the Golgi complex forms a continuous ribbon. In neurodegenerative diseases, the Golgi ribbon of a specific group of neurons is typically broken into isolated elements, a very early event which happens before clinical and other pathological symptoms become evident. It is not known whether this phenomenon is caused by mechanisms associated with cell death or if, conversely, it triggers apoptosis. When the phenomenon was studied in diseases such as Parkinson’s and Alzheimer’s or amyotrophic lateral sclerosis, it was attributed to a variety of causes, including the presence of cytoplasmatic protein aggregates, malfunctioning of intracellular traffic and/or alterations in the cytoskeleton. In the present review, we summarize the current findings related to these and other neurodegenerative diseases and try to search for clues on putative common causes.
Collapse
|
14
|
Pacifici F, Della Morte D, Capuani B, Pastore D, Bellia A, Sbraccia P, Di Daniele N, Lauro R, Lauro D. Peroxiredoxin6, a Multitask Antioxidant Enzyme Involved in the Pathophysiology of Chronic Noncommunicable Diseases. Antioxid Redox Signal 2019; 30:399-414. [PMID: 29160110 DOI: 10.1089/ars.2017.7427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE Chronic noncommunicable diseases (NCDs) are the leading causes of disability and death worldwide. NCDs mainly comprise diabetes mellitus, cardiovascular diseases, chronic obstructive pulmonary disease, cancer, and neurological degenerative diseases, which kill more than 80% of population, especially the elderly, worldwide. Recent Advances: Several recent theories established NCDs as multifactorial diseases, where a combination of genetic, epigenetic, and environmental factors contributes to their pathogenesis. Nevertheless, recent findings suggest that the common factor linking all these pathologies is an increase in oxidative stress and the age-related loss of the antioxidant mechanisms of defense against it. Impairment in mitochondrial homeostasis with consequent deregulation in oxidative stress balance has also been suggested. CRITICAL ISSUES Therefore, antioxidant proteins deserve particular attention for their potential role against NCDs. In particular, peroxiredoxin(Prdx)6 is a unique antioxidant enzyme, belonging to the Prdx family, with double properties, peroxidase and phospholipase activities. Through these activities, Prdx6 has been shown to be a powerful antioxidant enzyme, implicated in the pathogenesis of different NCDs. Recently, we described a phenotype of diabetes mellitus in Prdx6 knockout mice, suggesting a pivotal role of Prdx6 in the pathogenesis of cardiometabolic diseases. FUTURE DIRECTIONS Increasing awareness on the role of antioxidant defenses in the pathogenesis of NCDs may open novel therapeutic approaches to reduce the burden of this pandemic phenomenon. However, knowledge of the role of Prdx6 in NCD prevention and pathogenesis is still not clarified.
Collapse
Affiliation(s)
- Francesca Pacifici
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy
| | - David Della Morte
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy .,2 Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University , Rome, Italy
| | - Barbara Capuani
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy
| | - Donatella Pastore
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy
| | - Alfonso Bellia
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy .,3 Policlinico Tor Vergata Foundation, University Hospital , Rome, Italy
| | - Paolo Sbraccia
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy .,3 Policlinico Tor Vergata Foundation, University Hospital , Rome, Italy
| | - Nicola Di Daniele
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy .,3 Policlinico Tor Vergata Foundation, University Hospital , Rome, Italy
| | - Renato Lauro
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy
| | - Davide Lauro
- 1 Department of Systems Medicine, University of Rome Tor Vergata , Rome, Italy .,3 Policlinico Tor Vergata Foundation, University Hospital , Rome, Italy
| |
Collapse
|
15
|
Premature polyadenylation-mediated loss of stathmin-2 is a hallmark of TDP-43-dependent neurodegeneration. Nat Neurosci 2019; 22:180-190. [PMID: 30643298 PMCID: PMC6348009 DOI: 10.1038/s41593-018-0293-z] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are associated with loss of nuclear TDP-43. Here we identify that TDP-43 regulates expression of the neuronal growth-associated factor stathmin-2. Lowered TDP-43 levels, which reduce its binding to sites within the first intron of stathmin-2 pre-mRNA, uncover a cryptic polyadenylation site whose utilization produces a truncated, non-functional mRNA. Reduced stathmin-2 expression is found in neurons trans-differentiated from patient fibroblasts expressing an ALS-causing TDP-43 mutation, in motor cortex and spinal motor neurons from sporadic ALS patients and familial ALS patients with expansion in C9orf72, and in induced pluripotent stem cell (iPSC)-derived motor neurons depleted of TDP-43. Remarkably, while reduction in TDP-43 is shown to inhibit axonal regeneration of iPSC-derived motor neurons, rescue of stathmin-2 expression restores axonal regenerative capacity. Thus, premature polyadenylation-mediated reduction in stathmin-2 is a hallmark of ALS/FTD that functionally links reduced nuclear TDP-43 function to enhanced neuronal vulnerability.
Collapse
|
16
|
Šoltić D, Bowerman M, Stock J, Shorrock HK, Gillingwater TH, Fuller HR. Multi-Study Proteomic and Bioinformatic Identification of Molecular Overlap between Amyotrophic Lateral Sclerosis (ALS) and Spinal Muscular Atrophy (SMA). Brain Sci 2018; 8:brainsci8120212. [PMID: 30518112 PMCID: PMC6315439 DOI: 10.3390/brainsci8120212] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022] Open
Abstract
Unravelling the complex molecular pathways responsible for motor neuron degeneration in amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) remains a persistent challenge. Interest is growing in the potential molecular similarities between these two diseases, with the hope of better understanding disease pathology for the guidance of therapeutic development. The aim of this study was to conduct a comparative analysis of published proteomic studies of ALS and SMA, seeking commonly dysregulated molecules to be prioritized as future therapeutic targets. Fifteen proteins were found to be differentially expressed in two or more proteomic studies of both ALS and SMA, and bioinformatics analysis identified over-representation of proteins known to associate in vesicles and molecular pathways, including metabolism of proteins and vesicle-mediated transport—both of which converge on endoplasmic reticulum (ER)-Golgi trafficking processes. Calreticulin, a calcium-binding chaperone found in the ER, was associated with both pathways and we independently confirm that its expression was decreased in spinal cords from SMA and increased in spinal cords from ALS mice. Together, these findings offer significant insights into potential common targets that may help to guide the development of new therapies for both diseases.
Collapse
Affiliation(s)
- Darija Šoltić
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (D.S.); (M.B.)
- Institute for Science and Technology in Medicine, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Melissa Bowerman
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (D.S.); (M.B.)
- Institute for Science and Technology in Medicine, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Joanne Stock
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (D.S.); (M.B.)
- Institute for Science and Technology in Medicine, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Hannah K. Shorrock
- Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9AG, UK; (H.K.S.); (T.H.G.)
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Thomas H. Gillingwater
- Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH8 9AG, UK; (H.K.S.); (T.H.G.)
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Heidi R. Fuller
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK; (D.S.); (M.B.)
- Institute for Science and Technology in Medicine, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
- Correspondence: ; Tel.: +44-169-140-4693; Fax: +44-169-140-4065
| |
Collapse
|
17
|
Fisher AB. The phospholipase A 2 activity of peroxiredoxin 6. J Lipid Res 2018; 59:1132-1147. [PMID: 29716959 DOI: 10.1194/jlr.r082578] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/30/2018] [Indexed: 12/21/2022] Open
Abstract
Peroxiredoxin 6 (Prdx6) is a Ca2+-independent intracellular phospholipase A2 (called aiPLA2) that is localized to cytosol, lysosomes, and lysosomal-related organelles. Activity is minimal at cytosolic pH but is increased significantly with enzyme phosphorylation, at acidic pH, and in the presence of oxidized phospholipid substrate; maximal activity with phosphorylated aiPLA2 is ∼2 µmol/min/mg protein. Prdx6 is a "moonlighting" protein that also expresses glutathione peroxidase and lysophosphatidylcholine acyl transferase activities. The catalytic site for aiPLA2 activity is an S32-H26-D140 triad; S32-H26 is also the phospholipid binding site. Activity is inhibited by a serine "protease" inhibitor (diethyl p-nitrophenyl phosphate), an analog of the PLA2 transition state [1-hexadecyl-3-(trifluoroethyl)-sn-glycero-2-phosphomethanol (MJ33)], and by two naturally occurring proteins (surfactant protein A and p67phox), but not by bromoenol lactone. aiPLA2 activity has important physiological roles in the turnover (synthesis and degradation) of lung surfactant phospholipids, in the repair of peroxidized cell membranes, and in the activation of NADPH oxidase type 2 (NOX2). The enzyme has been implicated in acute lung injury, carcinogenesis, neurodegenerative diseases, diabetes, male infertility, and sundry other conditions, although its specific roles have not been well defined. Protein mutations and animal models are now available to further investigate the roles of Prdx6-aiPLA2 activity in normal and pathological physiology.
Collapse
Affiliation(s)
- Aron B Fisher
- Institute for Environmental Medicine of the Department of Physiology, University of Pennsylvania, Philadelphia, PA 19103
| |
Collapse
|
18
|
San Gil R, Ooi L, Yerbury JJ, Ecroyd H. The heat shock response in neurons and astroglia and its role in neurodegenerative diseases. Mol Neurodegener 2017; 12:65. [PMID: 28923065 PMCID: PMC5604514 DOI: 10.1186/s13024-017-0208-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 09/01/2017] [Indexed: 12/13/2022] Open
Abstract
Protein inclusions are a predominant molecular pathology found in numerous neurodegenerative diseases, including amyotrophic lateral sclerosis and Huntington's disease. Protein inclusions form in discrete areas of the brain characteristic to the type of neurodegenerative disease, and coincide with the death of neurons in that region (e.g. spinal cord motor neurons in amyotrophic lateral sclerosis). This suggests that the process of protein misfolding leading to inclusion formation is neurotoxic, and that cell-autonomous and non-cell autonomous mechanisms that maintain protein homeostasis (proteostasis) can, at times, be insufficient to prevent protein inclusion formation in the central nervous system. The heat shock response is a pro-survival pathway induced under conditions of cellular stress that acts to maintain proteostasis through the up-regulation of heat shock proteins, a superfamily of molecular chaperones, other co-chaperones and mitotic regulators. The kinetics and magnitude of the heat shock response varies in a stress- and cell-type dependent manner. It remains to be determined if and/or how the heat shock response is activated in the different cell-types that comprise the central nervous system (e.g. neurons and astroglia) in response to protein misfolding events that precede cellular dysfunctions in neurodegenerative diseases. This is particularly relevant considering emerging evidence demonstrating the non-cell autonomous nature of amyotrophic lateral sclerosis and Huntington's disease (and other neurodegenerative diseases) and the destructive role of astroglia in disease progression. This review highlights the complexity of heat shock response activation and addresses whether neurons and glia sense and respond to protein misfolding and aggregation associated with neurodegenerative diseases, in particular Huntington's disease and amyotrophic lateral sclerosis, by inducing a pro-survival heat shock response.
Collapse
Affiliation(s)
- Rebecca San Gil
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| | - Justin J. Yerbury
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| | - Heath Ecroyd
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| |
Collapse
|
19
|
Kim JE, Hong YH, Kim JY, Jeon GS, Jung JH, Yoon BN, Son SY, Lee KW, Kim JI, Sung JJ. Altered nucleocytoplasmic proteome and transcriptome distributions in an in vitro model of amyotrophic lateral sclerosis. PLoS One 2017; 12:e0176462. [PMID: 28453527 PMCID: PMC5409181 DOI: 10.1371/journal.pone.0176462] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/11/2017] [Indexed: 11/22/2022] Open
Abstract
Aberrant nucleocytoplasmic localization of proteins has been implicated in many neurodegenerative diseases. Evidence suggests that cytoplasmic mislocalization of nuclear proteins such as transactive response DNA-binding protein 43 (TDP-43) and fused in sarcoma (FUS) may be associated with neurotoxicity in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration. This study investigated the changes in nucleocytoplasmic distributions of the proteome and transcriptome in an in vitro model of ALS. After subcellular fractionation of motor neuron-like cell lines expressing wild-type or G93A mutant hSOD1, quantitative mass spectrometry and next-generation RNA sequencing (RNA-seq) were performed for the nuclear and cytoplasmic compartments. A subset of the results was validated via immunoblotting. A total of 1,925 proteins were identified in either the nuclear or cytoplasmic fractions, and 32% of these proteins were quantified in both fractions. The nucleocytoplasmic distribution of 37 proteins was significantly changed in mutant cells with nuclear and cytoplasmic shifts in 13 and 24 proteins, respectively (p<0.05). The proteins shifted towards the nucleus were enriched regarding pathways of RNA transport and processing (Dhx9, Fmr1, Srsf3, Srsf6, Tra2b), whereas protein folding (Cct5, Cct7, Cct8), aminoacyl-tRNA biosynthesis (Farsb, Nars, Txnrd1), synaptic vesicle cycle (Cltc, Nsf), Wnt signalling (Cltc, Plcb3, Plec, Psmd3, Ruvbl1) and Hippo signalling (Camk2d, Plcb3, Ruvbl1) pathways were over-represented in the proteins shifted to the cytoplasm. A weak correlation between the changes in protein and mRNA levels was found only in the nucleus, where mRNA was relatively abundant in mutant cells. This study provides a comprehensive dataset of the nucleocytoplasmic distribution of the proteome and transcriptome in an in vitro model of ALS. An integrated analysis of the nucleocytoplasmic distribution of the proteome and transcriptome demonstrated multiple candidate pathways including RNA processing/transport and protein synthesis and folding that may be relevant to the pathomechanism of ALS.
Collapse
Affiliation(s)
- Jee-Eun Kim
- Department of Neurology, Seoul Medical Center, Seoul, Republic of Korea
| | - Yoon Ho Hong
- Department of Neurology, Seoul National University Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Jin Young Kim
- Division of Mass Spectrometry Research, Korea Basic Science Institute, Daejun, Korea
- * E-mail: (JYK); (JIK); (JJS)
| | - Gye Sun Jeon
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | | | - Byung-Nam Yoon
- Department of Neurology, Inha University Hospital, Incheon, Korea
| | - Sung-Yeon Son
- Department of Neurology, Eulji University Hospital, Daejun, Korea
| | - Kwang-Woo Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - Jong-Il Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
- * E-mail: (JYK); (JIK); (JJS)
| | - Jung-Joon Sung
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
- * E-mail: (JYK); (JIK); (JJS)
| |
Collapse
|
20
|
Kline RA, Kaifer KA, Osman EY, Carella F, Tiberi A, Ross J, Pennetta G, Lorson CL, Murray LM. Comparison of independent screens on differentially vulnerable motor neurons reveals alpha-synuclein as a common modifier in motor neuron diseases. PLoS Genet 2017; 13:e1006680. [PMID: 28362802 PMCID: PMC5391970 DOI: 10.1371/journal.pgen.1006680] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/14/2017] [Accepted: 03/09/2017] [Indexed: 02/04/2023] Open
Abstract
The term “motor neuron disease” encompasses a spectrum of disorders in which motor neurons are the primary pathological target. However, in both patients and animal models of these diseases, not all motor neurons are equally vulnerable, in that while some motor neurons are lost very early in disease, others remain comparatively intact, even at late stages. This creates a valuable system to investigate the factors that regulate motor neuron vulnerability. In this study, we aim to use this experimental paradigm to identify potential transcriptional modifiers. We have compared the transcriptome of motor neurons from healthy wild-type mice, which are differentially vulnerable in the childhood motor neuron disease Spinal Muscular Atrophy (SMA), and have identified 910 transcriptional changes. We have compared this data set with published microarray data sets on other differentially vulnerable motor neurons. These neurons were differentially vulnerable in the adult onset motor neuron disease Amyotrophic Lateral Sclerosis (ALS), but the screen was performed on the equivalent population of neurons from neurologically normal human, rat and mouse. This cross species comparison has generated a refined list of differentially expressed genes, including CELF5, Col5a2, PGEMN1, SNCA, Stmn1 and HOXa5, alongside a further enrichment for synaptic and axonal transcripts. As an in vivo validation, we demonstrate that the manipulation of a significant number of these transcripts can modify the neurodegenerative phenotype observed in a Drosophila line carrying an ALS causing mutation. Finally, we demonstrate that vector-mediated expression of alpha-synuclein (SNCA), a transcript decreased in selectively vulnerable motor neurons in all four screens, can extend life span, increase weight and decrease neuromuscular junction pathology in a mouse model of SMA. In summary, we have combined multiple data sets to identify transcripts, which are strong candidates for being phenotypic modifiers, and demonstrated SNCA is a modifier of pathology in motor neuron disease. The term “motor neuron disease” refers to a group of disorders, causing progressive paralysis of affected patients due to the degeneration of motor neurons cells which control voluntary movements. Importantly, not all motor neurons appear to be affected in the same way, with those that control the face being affected less that those that control the abdomen. The reason why some motor neurons are more vulnerable is unknown; however, understanding this may provide new targets for therapeutics to slow motor neuron degeneration either as stand-alone therapeutics or in combination with SMN-inducing compounds. In this study, we analysed gene expression in different groups of motor neurons and compared this to previously published expression data to identify commonalities. One of the common transcripts was alpha-synuclein (SNCA), which was consistently expressed at lower levels in vulnerable motor neurons. Importantly, when SNCA levels were increased in a mouse model of motor neuron disease, the disease phenotype was significantly reduced, including an extension in survival and reduction in motor neuron pathology. Collectively, these results demonstrate that this approach can identify disease modifiers that can reduce disease severity in models of motor neuron disease and potentially identify new therapeutic targets.
Collapse
Affiliation(s)
- Rachel A. Kline
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Kevin A. Kaifer
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
| | - Erkan Y. Osman
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Francesco Carella
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Ariana Tiberi
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Jolill Ross
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Giuseppa Pennetta
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Christian L. Lorson
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Lyndsay M. Murray
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
21
|
Tu WY, Simpson JE, Highley JR, Heath PR. Spinal muscular atrophy: Factors that modulate motor neurone vulnerability. Neurobiol Dis 2017; 102:11-20. [PMID: 28161391 DOI: 10.1016/j.nbd.2017.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 01/07/2023] Open
Abstract
Spinal muscular atrophy (SMA), a leading genetic cause of infant death, is a neurodegenerative disease characterised by the selective loss of particular groups of motor neurones in the anterior horn of the spinal cord with concomitant muscle weakness. To date, no effective treatment is available, however, there are ongoing clinical trials are in place which promise much for the future. However, there remains an ongoing problem in trying to link a single gene loss to motor neurone degeneration. Fortunately, given successful disease models that have been established and intensive studies on SMN functions in the past ten years, we are fast approaching the stage of identifying the underlying mechanisms of SMA pathogenesis Here we discuss potential disease modifying factors on motor neurone vulnerability, in the belief that these factors give insight into the pathological mechanisms of SMA and therefore possible therapeutic targets.
Collapse
Affiliation(s)
- Wen-Yo Tu
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK.
| |
Collapse
|
22
|
Bellouze S, Baillat G, Buttigieg D, de la Grange P, Rabouille C, Haase G. Stathmin 1/2-triggered microtubule loss mediates Golgi fragmentation in mutant SOD1 motor neurons. Mol Neurodegener 2016; 11:43. [PMID: 27277231 PMCID: PMC4899909 DOI: 10.1186/s13024-016-0111-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/01/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pathological Golgi fragmentation represents a constant pre-clinical feature of many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) but its molecular mechanisms remain hitherto unclear. RESULTS Here, we show that the severe Golgi fragmentation in transgenic mutant SOD1(G85R) and SOD1(G93A) mouse motor neurons is associated with defective polymerization of Golgi-derived microtubules, loss of the COPI coat subunit β-COP, cytoplasmic dispersion of the Golgi tether GM130, strong accumulation of the ER-Golgi v-SNAREs GS15 and GS28 as well as tubular/vesicular Golgi fragmentation. Data mining, transcriptomic and protein analyses demonstrate that both SOD1 mutants cause early presymptomatic and rapidly progressive up-regulation of the microtubule-destabilizing proteins Stathmins 1 and 2. Remarkably, mutant SOD1-triggered Golgi fragmentation and Golgi SNARE accumulation are recapitulated by Stathmin 1/2 overexpression but completely rescued by Stathmin 1/2 knockdown or the microtubule-stabilizing drug Taxol. CONCLUSIONS We conclude that Stathmin-triggered microtubule destabilization mediates Golgi fragmentation in mutant SOD1-linked ALS and potentially also in related motor neuron diseases.
Collapse
Affiliation(s)
- Sarah Bellouze
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France
| | - Gilbert Baillat
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France
| | - Dorothée Buttigieg
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France
| | - Pierre de la Grange
- GenoSplice technology, iPEPS - ICM, Hôpital Pitié Salpêtrière, 47/83, bd de l'Hôpital, 75013 Paris, France
| | - Catherine Rabouille
- Department of Cell Biology, Hubrecht Institute of the KNAW & UMC Utrecht, Uppsalalaan 8, 3584 CT Utrecht, Netherlands
| | - Georg Haase
- Institut de Neurosciences de la Timone, UMR 7289, Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université, 27 bd Jean Moulin, 13005 Marseille, France.
| |
Collapse
|
23
|
Haase G, Rabouille C. Golgi Fragmentation in ALS Motor Neurons. New Mechanisms Targeting Microtubules, Tethers, and Transport Vesicles. Front Neurosci 2015; 9:448. [PMID: 26696811 PMCID: PMC4672084 DOI: 10.3389/fnins.2015.00448] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/13/2015] [Indexed: 12/12/2022] Open
Abstract
Pathological alterations of the Golgi apparatus, such as its fragmentation represent an early pre-clinical feature of many neurodegenerative diseases and have been widely studied in the motor neuron disease amyotrophic lateral sclerosis (ALS). Yet, the underlying molecular mechanisms have remained cryptic. In principle, Golgi fragmentation may result from defects in three major classes of proteins: structural Golgi proteins, cytoskeletal proteins and molecular motors, as well as proteins mediating transport to and through the Golgi. Here, we present the different mechanisms that may underlie Golgi fragmentation in animal and cellular models of ALS linked to mutations in SOD1, TARDBP (TDP-43), VAPB, and C9Orf72 and we propose a novel one based on findings in progressive motor neuronopathy (pmn) mice. These mice are mutated in the TBCE gene encoding the cis-Golgi localized tubulin-binding cofactor E, one of five chaperones that assist in tubulin folding and microtubule polymerization. Loss of TBCE leads to alterations in Golgi microtubules, which in turn impedes on the maintenance of the Golgi architecture. This is due to down-regulation of COPI coat components, dispersion of Golgi tethers and strong accumulation of ER-Golgi SNAREs. These effects are partially rescued by the GTPase ARF1 through recruitment of TBCE to the Golgi. We hypothesize that defects in COPI vesicles, microtubules and their interaction may also underlie Golgi fragmentation in human ALS linked to other mutations, spinal muscular atrophy (SMA), and related motor neuron diseases. We also discuss the functional relevance of pathological Golgi alterations, in particular their potential causative, contributory, or compensatory role in the degeneration of motor neuron cell bodies, axons and synapses.
Collapse
Affiliation(s)
- Georg Haase
- Centre National de la Recherche Scientifique and Aix-Marseille Université UMR 7289, Institut de Neurosciences de la Timone Marseille, France
| | - Catherine Rabouille
- The Department of Cell Biology, Hubrecht Institute of the Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
24
|
Alvarez-Miranda EA, Sinnl M, Farhan H. Alteration of Golgi Structure by Stress: A Link to Neurodegeneration? Front Neurosci 2015; 9:435. [PMID: 26617486 PMCID: PMC4641911 DOI: 10.3389/fnins.2015.00435] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/29/2015] [Indexed: 12/14/2022] Open
Abstract
The Golgi apparatus is well-known for its role as a sorting station in the secretory pathway as well as for its role in regulating post-translational protein modification. Another role for the Golgi is the regulation of cellular signaling by spatially regulating kinases, phosphatases, and GTPases. All these roles make it clear that the Golgi is a central regulator of cellular homeostasis. The response to stress and the initiation of adaptive responses to cope with it are fundamental abilities of all living cells. It was shown previously that the Golgi undergoes structural rearrangements under various stress conditions such as oxidative or osmotic stress. Neurodegenerative diseases are also frequently associated with alterations of Golgi morphology and many stress factors have been described to play an etiopathological role in neurodegeneration. It is however unclear whether the stress-Golgi connection plays a role in neurodegenerative diseases. Using a combination of bioinformatics modeling and literature mining, we will investigate evidence for such a tripartite link and we ask whether stress-induced Golgi arrangements are cause or consequence in neurodegeneration.
Collapse
Affiliation(s)
| | - Markus Sinnl
- Department of Statistics and Operations Research, University of Vienna Vienna, Austria
| | - Hesso Farhan
- Biotechnology Institute Thurgau Kreuzlingen, Switzerland ; Department of Biology, University of Konstanz Konstanz, Germany
| |
Collapse
|
25
|
Chidlow G, Wood JPM, Knoops B, Casson RJ. Expression and distribution of peroxiredoxins in the retina and optic nerve. Brain Struct Funct 2015; 221:3903-3925. [PMID: 26501408 PMCID: PMC5065902 DOI: 10.1007/s00429-015-1135-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/13/2015] [Indexed: 02/08/2023]
Abstract
Oxidative stress is implicated in various pathological conditions of the retina and optic nerve. Peroxiredoxins (Prdxs) comprise a recently characterized family of antioxidant enzymes. To date, little information exists regarding the distribution of Prdxs in the eye. Herein, we employed a combination of qRT-PCR, immunohistochemistry and Western blotting to determine the level of expression and distribution of the six Prdx isoforms in the retina and optic nerve of the rat. In addition, we performed some parallel analyses on the common marmoset (Callithrix Jacchus). In the rat, all of the Prdx transcripts were expressed in relatively high amounts in both retina and optic nerve, with abundances ranging from approximately 3–50 % of the level of the housekeeping gene cyclophilin. With regard to protein expression, each isoform was detected in the retina and optic nerve by either Western blotting and/or immunohistochemistry. Excepting Prdx4, there was a good correspondence between the rodent and primate results. In the retina, Prdx1 and Prdx2 were principally localized to neurons in the inner nuclear layer and cone photoreceptors, Prdx3 and Prdx5 displayed characteristic mitochondrial immunolabeling, while Prdx6 was associated with astrocytes and Müller cells. In the optic nerve, Prdx1 was robustly expressed by oligodendrocytes, Prdx3 and Prdx5 were observed in axons, and Prdx6 was restricted to astrocytes. The present findings augment our understanding of the distribution and expression of the Prdxs in the retina and optic nerve of rodents and primates and lay the foundation for subsequent analysis of their involvement in relevant blinding diseases.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Rd, Adelaide, SA, 5000, Australia. .,Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Rd, Adelaide, SA, 5000, Australia.
| | - John P M Wood
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Rd, Adelaide, SA, 5000, Australia.,Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Rd, Adelaide, SA, 5000, Australia
| | - Bernard Knoops
- Group of Animal Molecular and Cellular Biology, Institut des Sciences de la Vie (ISV), Université catholique de Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Robert J Casson
- Ophthalmic Research Laboratories, South Australian Institute of Ophthalmology, Hanson Institute Centre for Neurological Diseases, Frome Rd, Adelaide, SA, 5000, Australia.,Department of Ophthalmology and Visual Sciences, University of Adelaide, Frome Rd, Adelaide, SA, 5000, Australia
| |
Collapse
|
26
|
Buonora JE, Mousseau M, Jacobowitz DM, Lazarus RC, Yarnell AM, Olsen CH, Pollard HB, Diaz-Arrastia R, Latour L, Mueller GP. Autoimmune Profiling Reveals Peroxiredoxin 6 as a Candidate Traumatic Brain Injury Biomarker. J Neurotrauma 2015; 32:1805-14. [PMID: 25938937 PMCID: PMC4651056 DOI: 10.1089/neu.2014.3736] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Autoimmune profiling in rats revealed the antioxidant enzyme, peroxiredoxin 6 (PRDX6), as a target for autoantibodies evoked in response to traumatic brain injury (TBI). Consistent with this proposal, immunohistochemical analysis of rat cerebral cortex demonstrated that PRDX6 is highly expressed in the perivascular space, presumably contained within astrocytic foot processes. Accordingly, an immunosorbent electrochemiluminescence assay was developed for investigating PRDX6 in human samples. PRDX6 was found to be measurable in human blood and highly expressed in human cerebral cortex and platelets. Circulating levels of PRDX6 were elevated fourfold over control values 4 to 24 h following mild-to-moderate TBI. These findings suggest that PRDX6 may serve as a biomarker for TBI and that autoimmune profiling is a viable strategy for the discovery of novel TBI biomarkers.
Collapse
Affiliation(s)
- John E Buonora
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Michael Mousseau
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - David M Jacobowitz
- 2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Rachel C Lazarus
- 2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Angela M Yarnell
- 2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Cara H Olsen
- 4 Biostatistics Consulting Center, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Harvey B Pollard
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Ramon Diaz-Arrastia
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,5 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Lawrence Latour
- 6 Section on Stroke Diagnostics and Therapeutics, National Institute of Neurological Disorders and Stroke , Bethesda, Maryland
| | - Gregory P Mueller
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| |
Collapse
|
27
|
Chauvin S, Sobel A. Neuronal stathmins: A family of phosphoproteins cooperating for neuronal development, plasticity and regeneration. Prog Neurobiol 2015; 126:1-18. [DOI: 10.1016/j.pneurobio.2014.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/23/2014] [Accepted: 09/29/2014] [Indexed: 02/06/2023]
|
28
|
Gershoni-Emek N, Chein M, Gluska S, Perlson E. Amyotrophic lateral sclerosis as a spatiotemporal mislocalization disease: location, location, location. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 315:23-71. [PMID: 25708461 DOI: 10.1016/bs.ircmb.2014.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Spatiotemporal localization of signals is a fundamental feature impacting cell survival and proper function. The cell needs to respond in an accurate manner in both space and time to both intra- and intercellular environment cues. The regulation of this comprehensive process involves the cytoskeleton and the trafficking machinery, as well as local protein synthesis and ligand-receptor mechanisms. Alterations in such mechanisms can lead to cell dysfunction and disease. Motor neurons that can extend over tens of centimeters are a classic example for the importance of such events. Changes in spatiotemporal localization mechanisms are thought to play a role in motor neuron degeneration that occurs in amyotrophic lateral sclerosis (ALS). In this review we will discuss these mechanisms and argue that possible misregulated factors can lead to motor neuron degeneration in ALS.
Collapse
Affiliation(s)
- Noga Gershoni-Emek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Michael Chein
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shani Gluska
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
29
|
|
30
|
Bellouze S, Schäfer MK, Buttigieg D, Baillat G, Rabouille C, Haase G. Golgi fragmentation in pmn mice is due to a defective ARF1/TBCE cross-talk that coordinates COPI vesicle formation and tubulin polymerization. Hum Mol Genet 2014; 23:5961-75. [DOI: 10.1093/hmg/ddu320] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
31
|
Maximino JR, de Oliveira GP, Alves CJ, Chadi G. Deregulated expression of cytoskeleton related genes in the spinal cord and sciatic nerve of presymptomatic SOD1(G93A) Amyotrophic Lateral Sclerosis mouse model. Front Cell Neurosci 2014; 8:148. [PMID: 24904291 PMCID: PMC4033281 DOI: 10.3389/fncel.2014.00148] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 05/08/2014] [Indexed: 12/11/2022] Open
Abstract
Early molecular events related to cytoskeleton are poorly described in Amyotrophic Lateral Sclerosis (ALS), especially in the Schwann cell (SC), which offers strong trophic support to motor neurons. Database for Annotation, Visualization and Integrated Discovery (DAVID) tool identified cytoskeleton-related genes by employing the Cellular Component Ontology (CCO) in a large gene profiling of lumbar spinal cord and sciatic nerve of presymptomatic SOD1(G93A) mice. One and five CCO terms related to cytoskeleton were described from the spinal cord deregulated genes of 40 days (actin cytoskeleton) and 80 days (microtubule cytoskeleton, cytoskeleton part, actin cytoskeleton, neurofilament cytoskeleton, and cytoskeleton) old transgene mice, respectively. Also, four terms were depicted from the deregulated genes of sciatic nerve of 60 days old transgenes (actin cytoskeleton, cytoskeleton part, microtubule cytoskeleton and cytoskeleton). Kif1b was the unique deregulated gene in more than one studied region or presymptomatic age. The expression of Kif1b [quantitative polymerase chain reaction (qPCR)] elevated in the lumbar spinal cord (40 days old) and decreased in the sciatic nerve (60 days old) of presymptomatic ALS mice, results that were in line to microarray findings. Upregulation (24.8 fold) of Kif1b was seen in laser microdissected enriched immunolabeled motor neurons from the spinal cord of 40 days old presymptomatic SOD1(G93A) mice. Furthermore, Kif1b was dowregulated in the sciatic nerve Schwann cells of presymptomatic ALS mice (60 days old) that were enriched by means of cell microdissection (6.35 fold), cell sorting (3.53 fold), and primary culture (2.70 fold) technologies. The gene regulation of cytoskeleton molecules is an important occurrence in motor neurons and Schwann cells in presymptomatic stages of ALS and may be relevant in the dying back mechanisms of neuronal death. Furthermore, a differential regulation of Kif1b in the spinal cord and sciatic nerve cells emerged as key event in ALS.
Collapse
Affiliation(s)
- Jessica R Maximino
- Department of Neurology, Neuroregeneration Research Center, University of São Paulo School of Medicine São Paulo, Brazil
| | - Gabriela P de Oliveira
- Department of Neurology, Neuroregeneration Research Center, University of São Paulo School of Medicine São Paulo, Brazil
| | - Chrystian J Alves
- Department of Neurology, Neuroregeneration Research Center, University of São Paulo School of Medicine São Paulo, Brazil
| | - Gerson Chadi
- Department of Neurology, Neuroregeneration Research Center, University of São Paulo School of Medicine São Paulo, Brazil
| |
Collapse
|
32
|
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common form of motor neuron disease. ALS is a fatal neurodegenerative disease and clinical diagnosis typically takes many months to complete. Early disease diagnosis through the use of biomarkers may aid in correct clinical management of patients and possibly delay time to ventilator and morbidity. This review explores the progress of biomarker discovery efforts for ALS and the many challenges that remain. Included are different technologies utilized in biomarker discovery efforts (proteomic, genomic and metabolomic) and putative biomarkers uncovered using these techniques. These studies have discovered genetic mutations leading to familial forms of ALS, and specific protein alterations that occur in biological fluids (cerebrospinal fluid and blood) and/or tissues of ALS subjects. More recent high-throughput technologies have revealed panels of proteomic or metabolic biomarkers that can discriminate between ALS and control groups. The identification of disease-specific biomarkers will provide opportunities to develop early diagnostic measures as well as surrogate markers to monitor disease progression and test drug efficacy in clinical trials.
Collapse
Affiliation(s)
- Robert Bowser
- University of Pittsburgh, Department of Pathology, School of Medicine, ST S-420, 200 Lothrop Street, Pittsburgh, PA 15261, USA.
| | | | | |
Collapse
|
33
|
Wiggins LM. Morphological changes and altered expression of antioxidant proteins in a heterozygous dynein mutant; a mouse model of spinal muscular atrophy. ACTA ACUST UNITED AC 2014; 3:161-173. [PMID: 25866698 DOI: 10.5455/oams.310714.or.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE There is increased evidence that oxidative stress is involved in exacerbations of neurodegenerative diseases and spinal muscular atrophies. METHODS We examined changes in morphology and expression of antioxidant proteins and peroxiredoxins in motor neurons of lumbar spinal cord, dorsal root ganglion sensory neurons, macroglial cells and quadriceps muscles of newborn heterozygous Loa/+ mice ("legs at odd angles"), a mouse model for early onset of the spinal muscular atrophy with lower extremity predominance (SMA-LED). RESULTS Our data indicate that newborn Loa-mice develop: neuroinflammation of the sensory and motor neurons; muscular inflammation with atrophic and denervated myofibers; increased expression of neuronal mitochondrial peroxiredoxins (Prxs) 3, 5 and cytoplasmic Prx 6 in motor and sensory neurons, myofibers, fibroblasts of perimysium and chondrocytes of cartilage; and decreased expression of Prx 6 by glial cells and in extracellular space surrounding motor neurons. CONCLUSION The decrease in expression of Prx 6 by glial cells and extracellular Prx 6 secretion in early stages of the pathological conditions is consistent with the hypothesis that chronic oxidative stress may lead to neurodegeneration of motor neurons and exacerbation of the pathology.
Collapse
Affiliation(s)
- Larisa M Wiggins
- Department of Physiology and Cell Biology, University of Nevada, Reno
| |
Collapse
|
34
|
Hanschmann EM, Godoy JR, Berndt C, Hudemann C, Lillig CH. Thioredoxins, glutaredoxins, and peroxiredoxins--molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling. Antioxid Redox Signal 2013; 19:1539-605. [PMID: 23397885 PMCID: PMC3797455 DOI: 10.1089/ars.2012.4599] [Citation(s) in RCA: 496] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 02/01/2013] [Accepted: 02/07/2013] [Indexed: 12/19/2022]
Abstract
Thioredoxins (Trxs), glutaredoxins (Grxs), and peroxiredoxins (Prxs) have been characterized as electron donors, guards of the intracellular redox state, and "antioxidants". Today, these redox catalysts are increasingly recognized for their specific role in redox signaling. The number of publications published on the functions of these proteins continues to increase exponentially. The field is experiencing an exciting transformation, from looking at a general redox homeostasis and the pathological oxidative stress model to realizing redox changes as a part of localized, rapid, specific, and reversible redox-regulated signaling events. This review summarizes the almost 50 years of research on these proteins, focusing primarily on data from vertebrates and mammals. The role of Trx fold proteins in redox signaling is discussed by looking at reaction mechanisms, reversible oxidative post-translational modifications of proteins, and characterized interaction partners. On the basis of this analysis, the specific regulatory functions are exemplified for the cellular processes of apoptosis, proliferation, and iron metabolism. The importance of Trxs, Grxs, and Prxs for human health is addressed in the second part of this review, that is, their potential impact and functions in different cell types, tissues, and various pathological conditions.
Collapse
Affiliation(s)
- Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| | - José Rodrigo Godoy
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Christoph Hudemann
- Institute of Laboratory Medicine, Molecular Diagnostics, Philipps University, Marburg, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| |
Collapse
|
35
|
Alterations in energy metabolism, neuroprotection and visual signal transduction in the retina of Parkinsonian, MPTP-treated monkeys. PLoS One 2013; 8:e74439. [PMID: 24040246 PMCID: PMC3764107 DOI: 10.1371/journal.pone.0074439] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/01/2013] [Indexed: 11/27/2022] Open
Abstract
Parkinson disease is mainly characterized by the degeneration of dopaminergic neurons in the central nervous system, including the retina. Different interrelated molecular mechanisms underlying Parkinson disease-associated neuronal death have been put forward in the brain, including oxidative stress and mitochondrial dysfunction. Systemic injection of the proneurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to monkeys elicits the appearance of a parkinsonian syndrome, including morphological and functional impairments in the retina. However, the intracellular events leading to derangement of dopaminergic and other retinal neurons in MPTP-treated animal models have not been so far investigated. Here we have used a comparative proteomics approach to identify proteins differentially expressed in the retina of MPTP-treated monkeys. Proteins were solubilized from the neural retinas of control and MPTP-treated animals, labelled separately with two different cyanine fluorophores and run pairwise on 2D DIGE gels. Out of >700 protein spots resolved and quantified, 36 were found to exhibit statistically significant differences in their expression levels, of at least ±1.4-fold, in the parkinsonian monkey retina compared with controls. Most of these spots were excised from preparative 2D gels, trypsinized and subjected to MALDI-TOF MS and LC-MS/MS analyses. Data obtained were used for protein sequence database interrogation, and 15 different proteins were successfully identified, of which 13 were underexpressed and 2 overexpressed. These proteins were involved in key cellular functional pathways such as glycolysis and mitochondrial electron transport, neuronal protection against stress and survival, and phototransduction processes. These functional categories underscore that alterations in energy metabolism, neuroprotective mechanisms and signal transduction are involved in MPTP-induced neuronal degeneration in the retina, in similarity to mechanisms thought to underlie neuronal death in the Parkinson’s diseased brain and neurodegenerative diseases of the retina proper.
Collapse
|
36
|
Dowell JA, Johnson JA. Mechanisms of Nrf2 protection in astrocytes as identified by quantitative proteomics and siRNA screening. PLoS One 2013; 8:e70163. [PMID: 23922950 PMCID: PMC3726381 DOI: 10.1371/journal.pone.0070163] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/16/2013] [Indexed: 12/30/2022] Open
Abstract
The Nrf2 (NF-E2 related factor 2)-ARE (antioxidant response element) pathway controls a powerful array of endogenous cellular antioxidant systems and is an important pathway in the detoxification of reactive oxygen species (ROS) in the brain. Using a combination of quantitative proteomics and siRNA screening, we have identified novel protective mechanisms of the Nrf2-ARE pathway against oxidative stress in astrocytes. Studies from our lab and others have shown Nrf2 overexpression protects astrocytes from oxidative stress. However, the exact mechanisms by which Nrf2 elicits these effects are unknown. In this study, we show that induction of Nrf2 reduces levels of reactive oxygen species (ROS) produced by various oxidative stressors and results in robust cytoprotection. To identify the enzymes responsible for these effects, we used stable isotope labeling by amino acids in cell culture (SILAC) and quantitative shotgun proteomics to identify 72 Nrf2-regulated proteins in astrocytes. We hypothesized a subset of these proteins might play a critical role in Nrf2 protection. In order to identify these critical proteins, we used bioinformatics to narrow our target list of proteins and then systematically screened each candidate with siRNA to assess the role of each in Nrf2 protection. We screened each target against H2O2, tert-butyl hydroperoxide, and 4-hydroxynonenal and subsequently identified three enzymes-catalase, prostaglandin reductase-1, and peroxiredoxin-6-that are critical for Nrf2-mediated protection in astrocytes.
Collapse
Affiliation(s)
- James A. Dowell
- Division of Pharmaceutical Science, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jeffrey A. Johnson
- Division of Pharmaceutical Science, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
37
|
Elkharaz J, Ugun-Klusek A, Constantin-Teodosiu D, Lawler K, Mayer RJ, Billett E, Lowe J, Bedford L. Implications for oxidative stress and astrocytes following 26S proteasomal depletion in mouse forebrain neurones. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1930-8. [PMID: 23851049 DOI: 10.1016/j.bbadis.2013.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/25/2013] [Accepted: 07/01/2013] [Indexed: 02/06/2023]
Abstract
Neurodegenerative diseases are characterized by progressive degeneration of selective neurones in the nervous system, but the underlying mechanisms involved in neuroprotection and neurodegeneration remain unclear. Dysfunction of the ubiquitin proteasome system is one of the proposed hypotheses for the cause and progression of neuronal loss. We have performed quantitative two-dimensional fluorescence difference in-gel electrophoresis combined with peptide mass fingerprinting to reveal proteome changes associated with neurodegeneration following 26S proteasomal depletion in mouse forebrain neurones. Differentially expressed proteins were validated by Western blotting, biochemical assays and immunohistochemistry. Of significance was increased expression of the antioxidant enzyme peroxiredoxin 6 (PRDX6) in astrocytes, associated with oxidative stress. Interestingly, PRDX6 is a bifunctional enzyme with antioxidant peroxidase and phospholipase A2 (PLA2) activities. The PLA2 activity of PRDX6 was also increased following 26S proteasomal depletion and may be involved in neuroprotective or neurodegenerative mechanisms. This is the first in vivo report of oxidative stress caused directly by neuronal proteasome dysfunction in the mammalian brain. The results contribute to understanding neuronal-glial interactions in disease pathogenesis, provide an in vivo link between prominent disease hypotheses and importantly, are of relevance to a heterogeneous spectrum of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jamal Elkharaz
- School of Biomedical Sciences, University of Nottingham, Nottingham, UK
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Fazeli AS, Nasrabadi D, Pouya A, Mirshavaladi S, Sanati MH, Baharvand H, Salekdeh GH. Proteome analysis of post-transplantation recovery mechanisms of an EAE model of multiple sclerosis treated with embryonic stem cell-derived neural precursors. J Proteomics 2013; 94:437-50. [PMID: 23791935 DOI: 10.1016/j.jprot.2013.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 06/02/2013] [Accepted: 06/09/2013] [Indexed: 12/21/2022]
Abstract
UNLABELLED Multiple sclerosis (MS) is a chronic inflammatory and progressive disorder of the central nervous system (CNS), which ultimately causes demyelination and subsequent axonal injury. Experimental autoimmune encephalomyelitis (EAE) is a well-characterized animal model to study the etiology and pathogenesis of MS. This model can also be used to investigate various therapeutic approaches for MS. Herein; we have treated a score 3 EAE mouse model with an embryonic stem cell-derived neural precursor. Clinical analysis showed recovery of the EAE model of MS following transplantation. We analyzed the proteome of spinal cords of healthy and EAE samples before and after transplantation. Proteome analysis revealed that expressions of 86 spinal cord protein spots changed in the EAE or transplanted mouse compared to controls. Mass spectrometry resulted in identification of 72 proteins. Of these, the amounts of 27 differentially expressed proteins in EAE samples returned to sham levels after transplantation, suggesting a possible correlation between changes at the proteome level and clinical signs of EAE in transplanted mice. The recovered proteins belonged to various functional groups that included disturbances in ionic and neurotransmitter release, apoptosis, iron hemostasis, and signal transduction. Our results provided a proteomic view of the molecular mechanisms of EAE recovery after stem cell transplantation. BIOLOGICAL SIGNIFICANCE In this study, we applied proteomics to analyze the changes in proteome pattern of EAE mouse model after embryonic stem cell-derived neural precursor transplantation. Our proteome results clearly showed that the expression levels of several differentially expressed proteins in EAE samples returned to sham levels after transplantation, which suggested a possible correlation between changes at the proteome level and decreased clinical signs of EAE in transplanted mice. These results will serve as a basis to address new questions and design new experiments to elucidate the biology of EAE and recovery after transplantation. A thorough understanding of stem cell-mediated therapeutic mechanisms might result in the development of more efficacious therapies for MS than are currently available.
Collapse
Affiliation(s)
- Abolhassan Shahzadeh Fazeli
- Department of Molecular Systems Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Genetics at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
39
|
Abrahamsen G, Fan Y, Matigian N, Wali G, Bellette B, Sutharsan R, Raju J, Wood SA, Veivers D, Sue CM, Mackay-Sim A. A patient-derived stem cell model of hereditary spastic paraplegia with SPAST mutations. Dis Model Mech 2012; 6:489-502. [PMID: 23264559 PMCID: PMC3597030 DOI: 10.1242/dmm.010884] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) leads to progressive gait disturbances with lower limb muscle weakness and spasticity. Mutations in SPAST are a major cause of adult-onset, autosomal-dominant HSP. Spastin, the protein encoded by SPAST, is a microtubule-severing protein that is enriched in the distal axon of corticospinal motor neurons, which degenerate in HSP patients. Animal and cell models have identified functions of spastin and mutated spastin but these models lack the gene dosage, mutation variability and genetic background that characterize patients with the disease. In this study, this genetic variability is encompassed by comparing neural progenitor cells derived from biopsies of the olfactory mucosa from healthy controls with similar cells from HSP patients with SPAST mutations, in order to identify cell functions altered in HSP. Patient-derived cells were similar to control-derived cells in proliferation and multiple metabolic functions but had major dysregulation of gene expression, with 57% of all mRNA transcripts affected, including many associated with microtubule dynamics. Compared to control cells, patient-derived cells had 50% spastin, 50% acetylated α-tubulin and 150% stathmin, a microtubule-destabilizing enzyme. Patient-derived cells were smaller than control cells. They had altered intracellular distributions of peroxisomes and mitochondria and they had slower moving peroxisomes. These results suggest that patient-derived cells might compensate for reduced spastin, but their increased stathmin expression reduced stabilized microtubules and altered organelle trafficking. Sub-nanomolar concentrations of the microtubule-binding drugs, paclitaxel and vinblastine, increased acetylated α-tubulin levels in patient cells to control levels, indicating the utility of this cell model for screening other candidate compounds for drug therapies.
Collapse
Affiliation(s)
- Greger Abrahamsen
- National Centre for Adult Stem Cell Research, Eskitis Institute for Cell and Molecular Therapies, Griffith University, Brisbane, Queensland 4111, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Fanara P, Wong PYA, Husted KH, Liu S, Liu VM, Kohlstaedt LA, Riiff T, Protasio JC, Boban D, Killion S, Killian M, Epling L, Sinclair E, Peterson J, Price RW, Cabin DE, Nussbaum RL, Brühmann J, Brandt R, Christine CW, Aminoff MJ, Hellerstein MK. Cerebrospinal fluid-based kinetic biomarkers of axonal transport in monitoring neurodegeneration. J Clin Invest 2012; 122:3159-69. [PMID: 22922254 DOI: 10.1172/jci64575] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 07/12/2012] [Indexed: 12/14/2022] Open
Abstract
Progress in neurodegenerative disease research is hampered by the lack of biomarkers of neuronal dysfunction. We here identified a class of cerebrospinal fluid-based (CSF-based) kinetic biomarkers that reflect altered neuronal transport of protein cargo, a common feature of neurodegeneration. After a pulse administration of heavy water (2H2O), distinct, newly synthesized 2H-labeled neuronal proteins were transported to nerve terminals and secreted, and then appeared in CSF. In 3 mouse models of neurodegeneration, distinct 2H-cargo proteins displayed delayed appearance and disappearance kinetics in the CSF, suggestive of aberrant transport kinetics. Microtubule-modulating pharmacotherapy normalized CSF-based kinetics of affected 2H-cargo proteins and ameliorated neurodegenerative symptoms in mice. After 2H2O labeling, similar neuronal transport deficits were observed in CSF of patients with Parkinson's disease (PD) compared with non-PD control subjects, which indicates that these biomarkers are translatable and relevant to human disease. Measurement of transport kinetics may provide a sensitive method to monitor progression of neurodegeneration and treatment effects.
Collapse
|
41
|
Goemaere J, Knoops B. Peroxiredoxin distribution in the mouse brain with emphasis on neuronal populations affected in neurodegenerative disorders. J Comp Neurol 2012; 520:258-80. [PMID: 21674491 DOI: 10.1002/cne.22689] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Redox changes are observed in neurodegenerative diseases, ranging from increased levels of reactive oxygen/nitrogen species and disturbance of antioxidant systems, to nitro-oxidative damage. By reducing hydrogen peroxide, peroxynitrite, and organic hydroperoxides, peroxiredoxins (Prdxs) represent a major potential protective barrier against nitro-oxidative insults in the brain. While recent works have investigated the putative role of Prdxs in neurodegenerative disorders, less is known about their expression in the healthy brain. Here we used immunohistochemistry to map basal expression of Prdxs throughout C57BL/6 mouse brain. We first confirmed the neuronal localization of Prdx2-5 and the glial expression of Prdx1, Prdx4, and Prdx6. Then we performed an in-depth analysis of neuronal Prdx distribution in the brain. Our results show that Prdx2-5 are widely detected in the different neuronal populations, and especially well expressed in the olfactory bulb, in the cerebral cortex, in pons nuclei, in the red nucleus, in all cranial nerve nuclei, in the cerebellum, and in motor neurons of the spinal cord. In contrast, Prdx expression is very low in the dopaminergic neurons of substantia nigra pars compacta and in the CA1/2 pyramidal cells of hippocampus. This low basal expression may contribute to the vulnerability of these neurons to nitro-oxidative attacks occurring in Parkinson's disease and Alzheimer's disease. In addition, we found that Prdx expression levels are unevenly distributed among neurons of a determined region and that distinct regional patterns of expression are observed between isoforms, reinforcing the hypothesis of the nonredundant function of Prdxs.
Collapse
Affiliation(s)
- Julie Goemaere
- Laboratory of Cell Biology, Institut des Sciences de la Vie (ISV), Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | | |
Collapse
|
42
|
Song IS, Kim HK, Jeong SH, Lee SR, Kim N, Rhee BD, Ko KS, Han J. Mitochondrial peroxiredoxin III is a potential target for cancer therapy. Int J Mol Sci 2011; 12:7163-85. [PMID: 22072940 PMCID: PMC3211031 DOI: 10.3390/ijms12107163] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/30/2011] [Accepted: 10/20/2011] [Indexed: 01/01/2023] Open
Abstract
Mitochondria are involved either directly or indirectly in oncogenesis and the alteration of metabolism in cancer cells. Cancer cells contain large numbers of abnormal mitochondria and produce large amounts of reactive oxygen species (ROS). Oxidative stress is caused by an imbalance between the production of ROS and the antioxidant capacity of the cell. Several cancer therapies, such as chemotherapeutic drugs and radiation, disrupt mitochondrial homeostasis and release cytochrome c, leading to apoptosome formation, which activates the intrinsic pathway. This is modulated by the extent of mitochondrial oxidative stress. The peroxiredoxin (Prx) system is a cellular defense system against oxidative stress, and mitochondria in cancer cells are known to contain high levels of Prx III. Here, we review accumulating evidence suggesting that mitochondrial oxidative stress is involved in cancer, and discuss the role of the mitochondrial Prx III antioxidant system as a potential target for cancer therapy. We hope that this review will provide the basis for new strategic approaches in the development of effective cancer treatments.
Collapse
Affiliation(s)
- In-Sung Song
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 614-7-5, Korea; E-Mails: (I.-S.S.); (H.-K.K.); (S.-H.J.); (S.-R.L.); (N.K.); (B.D.R.); (K.S.K.)
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Fisher AB. Peroxiredoxin 6: a bifunctional enzyme with glutathione peroxidase and phospholipase A₂ activities. Antioxid Redox Signal 2011; 15:831-44. [PMID: 20919932 PMCID: PMC3125547 DOI: 10.1089/ars.2010.3412] [Citation(s) in RCA: 298] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peroxiredoxin 6 (Prdx6) is the prototype and the only mammalian 1-Cys member of the Prdx family. Major differences from 2-Cys Prdxs include the use of glutathione (GSH) instead of thioredoxin as the physiological reductant, heterodimerization with πGSH S-transferase as part of the catalytic cycle, and the ability either to reduce the oxidized sn-2 fatty acyl group of phospholipids (peroxidase activity) or to hydrolyze the sn-2 ester (alkyl) bond of phospholipids (phospholipase A(2) [PLA(2)] activity). The bifunctional protein has separate active sites for peroxidase (C47, R132, H39) and PLA(2) (S32, D140, H26) activities. These activities are dependent on binding of the protein to phospholipids at acidic pH and to oxidized phospholipids at cytosolic pH. Prdx6 can be phosphorylated by MAP kinases at T177, which markedly increases its PLA(2) activity and broadens its pH-activity spectrum. Prdx6 is primarily cytosolic but also is targeted to acidic organelles (lysosomes, lamellar bodies) by a specific targeting sequence (amino acids 31-40). Oxidant stress and keratinocyte growth factor are potent regulators of Prdx6 gene expression. Prdx6 has important roles in both antioxidant defense based on its ability to reduce peroxidized membrane phospholipids and in phospholipid homeostasis based on its ability to generate lysophospholipid substrate for the remodeling pathway of phospholipid synthesis.
Collapse
Affiliation(s)
- Aron B Fisher
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
44
|
Piscopo P, Crestini A, Adduci A, Ferrante A, Massari M, Popoli P, Vanacore N, Confaloni A. Altered oxidative stress profile in the cortex of mice fed an enriched branched-chain amino acids diet: Possible link with amyotrophic lateral sclerosis? J Neurosci Res 2011; 89:1276-83. [DOI: 10.1002/jnr.22655] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 02/24/2011] [Accepted: 03/07/2011] [Indexed: 12/11/2022]
|
45
|
Wu CY, Whye D, Glazewski L, Choe L, Kerr D, Lee KH, Mason RW, Wang W. Proteomic assessment of a cell model of spinal muscular atrophy. BMC Neurosci 2011; 12:25. [PMID: 21385431 PMCID: PMC3063191 DOI: 10.1186/1471-2202-12-25] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 03/08/2011] [Indexed: 12/29/2022] Open
Abstract
Background Deletion or mutation(s) of the survival motor neuron 1 (SMN1) gene causes spinal muscular atrophy (SMA), a neuromuscular disease characterized by spinal motor neuron death and muscle paralysis. Complete loss of the SMN protein is embryonically lethal, yet reduced levels of this protein result in selective death of motor neurons. Why motor neurons are specifically targeted by SMN deficiency remains to be determined. In this study, embryonic stem (ES) cells derived from a severe SMA mouse model were differentiated into motor neurons in vitro by addition of retinoic acid and sonic hedgehog agonist. Proteomic and western blot analyses were used to probe protein expression alterations in this cell-culture model of SMA that could be relevant to the disease. Results When ES cells were primed with Noggin/fibroblast growth factors (bFGF and FGF-8) in a more robust neural differentiation medium for 2 days before differentiation induction, the efficiency of in vitro motor neuron differentiation was improved from ~25% to ~50%. The differentiated ES cells expressed a pan-neuronal marker (neurofilament) and motor neuron markers (Hb9, Islet-1, and ChAT). Even though SMN-deficient ES cells had marked reduced levels of SMN (~20% of that in control ES cells), the morphology and differentiation efficiency for these cells are comparable to those for control samples. However, proteomics in conjunction with western blot analyses revealed 6 down-regulated and 14 up-regulated proteins with most of them involved in energy metabolism, cell stress-response, protein degradation, and cytoskeleton stability. Some of these activated cellular pathways showed specificity for either undifferentiated or differentiated cells. Increased p21 protein expression indicated that SMA ES cells were responding to cellular stress. Up-regulation of p21 was confirmed in spinal cord tissues from the same SMA mouse model from which the ES cells were derived. Conclusion SMN-deficient ES cells provide a cell-culture model for SMA. SMN deficiency activates cellular stress pathways, causing a dysregulation of energy metabolism, protein degradation, and cytoskeleton stability.
Collapse
Affiliation(s)
- Chia-Yen Wu
- Department of Biological Science, University of Delaware, Newark, DE, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Yang HY, Kwon J, Cho EJ, Choi HI, Park C, Park HR, Park SH, Chung KJ, Ryoo ZY, Cho KO, Lee TH. Proteomic analysis of protein expression affected by peroxiredoxin V knock-down in hypoxic kidney. J Proteome Res 2010; 9:4003-15. [PMID: 20553050 DOI: 10.1021/pr100190b] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Peroxiredoxin V, an atypical thioredoxin peroxidase, is widely expressed in mammalian tissues. In addition, Prdx V is localized in mitochondria, peroxisome, cytosol, and the nucleus. Prdx V has been reported to protect a wide range of cellular environments as an antioxidant enzyme, and its dysfunctions may be implicated in several diseases, such as cancer, inflammation, and neurodegenerative disease. Identification and relative quantification of proteins affected by Prdx V may help identify novel signaling mechanisms that are important for oxidative stress response. However, the role of Prdx V in the modulation of hypoxia-related cellular response is not studied yet. To examine the function of endogenous Prdx V in hypoxic condition in vivo, we generated a transgenic mouse model with Prdx V siRNA expression controlled by U6 promoter. Of many tissues, the knockdown of Prdx V expression was displayed in the kidney, lung, and liver but not the spleen and skin. We conducted on the basis of nano-UPLC-MS(E) proteomic study to identify the Prdx V-affected protein networks in hypoxic kidneys. In this study, we identified protein networks associated with oxidative stress, fatty acid metabolism, and mitochondrial dysfunction. Our results indicated that Prdx V affected to regulation of kidney homeostasis under hypoxia stress.
Collapse
Affiliation(s)
- Hee-Young Yang
- Department of Oral Biochemistry, Dental Science Research Institute, The second Stage of Brain Korea 21 for Dental School, Chonnam National University, Gwangju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Fazeli AS, Nasrabadi D, Sanati MH, Pouya A, Ibrahim SM, Baharvand H, Salekdeh GH. Proteome analysis of brain in murine experimental autoimmune encephalomyelitis. Proteomics 2010; 10:2822-32. [PMID: 20540118 DOI: 10.1002/pmic.200900507] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Multiple sclerosis is considered a prototype inflammatory autoimmune disorder of the CNS. Experimental autoimmune encephalomyelitis (EAE) induced by myelin oligodendrocyte glycoprotein is one of the best-characterized animal models of multiple sclerosis. Comprehensive understanding of gene expression in EAE can help identify genes that are important in drug response and pathogenesis. We applied a 2-DE-based proteomics approach to analyze the protein expression pattern of the brain in healthy and EAE samples. Of more than 1000 protein spots we analyzed, 70 showed reproducible and significant changes in EAE compared to controls. Of these, 42 protein spots could be identified using MALDI TOF-TOF-MS. They included mitochondrial and structural proteins as well as proteins involved in ionic and neurotransmitter release, blood barriers, apoptosis, and signal transduction. The possible role of these proteins in the responses of mice to animal models of multiple sclerosis is discussed.
Collapse
Affiliation(s)
- Abolhassan Shahzadeh Fazeli
- Department of Molecular Systems Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
48
|
Rodríguez JE, Willis MS. The therapeutic potential of heat shock proteins in cardiomyopathies due to mutations in cardiac structural proteins. J Mol Cell Cardiol 2010; 49:904-7. [PMID: 20920511 DOI: 10.1016/j.yjmcc.2010.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 11/29/2022]
|
49
|
Dejda A, Chan P, Seaborn T, Coquet L, Jouenne T, Fournier A, Vaudry H, Vaudry D. Involvement of stathmin 1 in the neurotrophic effects of PACAP in PC12 cells. J Neurochem 2010; 114:1498-510. [PMID: 20569302 DOI: 10.1111/j.1471-4159.2010.06873.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rat pheochromocytoma PC12 cells have been widely used to investigate the neurotrophic activities of pituitary adenylate cyclase-activating polypeptide (PACAP). In particular, PACAP has been shown to promote differentiation and to inhibit apoptosis of PC12 cells. In order to identify the mechanisms mediating these effects, we sought for proteins that are phosphorylated upon PACAP treatment. High-performance liquid chromatography and 2D gel electrophoresis analysis, coupled with mass spectrometry, revealed that stathmin 1 is strongly phosphorylated within only 5 min of exposure to PACAP. Western blot experiments confirmed that PACAP induced a robust phosphorylation of stathmin 1 in a time-dependent manner. On the other hand, PACAP decreased stathmin 1 gene expression. Investigations of the signaling mechanisms known to be activated by PACAP revealed that phosphorylation of stathmin 1 was mainly mediated through the protein kinase A and mitogen-activated protein kinase pathways. Blockage of stathmin 1 expression with small interfering RNA did not affect PC12 cell differentiation induced by PACAP but reduced the ability of the peptide to inhibit caspase 3 activity and significantly decreased its neuroprotective action. Taken together, these data demonstrate that stathmin 1 is involved in the neurotrophic effect of PACAP in PC12 cells.
Collapse
Affiliation(s)
- Agnieszka Dejda
- INSERM U982, Institut Fédératif de Recherches Multidisciplinaires sur les Peptides (IFRMP 23), Université de Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kassa RM, Kasensa NL, Monterroso VH, Kayton RJ, Klimek JE, David LL, Lunganza KR, Kayembe KT, Bentivoglio M, Juliano SL, Tshala-Katumbay DD. On the biomarkers and mechanisms of konzo, a distinct upper motor neuron disease associated with food (cassava) cyanogenic exposure. Food Chem Toxicol 2010; 49:571-8. [PMID: 20538033 DOI: 10.1016/j.fct.2010.05.080] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 05/18/2010] [Accepted: 05/19/2010] [Indexed: 12/29/2022]
Abstract
Konzo is a self-limiting central motor-system disease associated with food dependency on cassava and low dietary intake of sulfur amino acids (SAA). Under conditions of SAA-deficiency, ingested cassava cyanogens yield metabolites that include thiocyanate and cyanate, a protein-carbamoylating agent. We studied the physical and biochemical modifications of rat serum and spinal cord proteins arising from intoxication of young adult rats with 50-200mg/kg linamarin, or 200mg/kg sodium cyanate (NaOCN), or vehicle (saline) and fed either a normal amino acid- or SAA-deficient diet for up to 2 weeks. Animals under SAA-deficient diet and treatment with linamarin or NaOCN developed hind limb tremors or motor weakness, respectively. LC/MS-MS analysis revealed differential albumin carbamoylation in animals treated with NaOCN, vs. linamarin/SAA-deficient diet, or vehicle. 2D-DIGE and MALDI-TOF/MS-MS analysis of the spinal cord proteome showed differential expression of proteins involved in oxidative mechanisms (e.g. peroxiredoxin 6), endocytic vesicular trafficking (e.g. dynamin 1), protein folding (e.g. protein disulfide isomerase), and maintenance of the cytoskeleton integrity (e.g. α-spectrin). Studies are needed to elucidate the role of the aformentioned modifications in the pathogenesis of cassava-associated motor-system disease.
Collapse
Affiliation(s)
- Roman M Kassa
- Center for Research on Occupational and Environmental Toxicology, Oregon Health and Science University, Portland, OR, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|