1
|
Muleiro Alvarez M, Cano-Herrera G, Osorio Martínez MF, Vega Gonzales-Portillo J, Monroy GR, Murguiondo Pérez R, Torres-Ríos JA, van Tienhoven XA, Garibaldi Bernot EM, Esparza Salazar F, Ibarra A. A Comprehensive Approach to Parkinson's Disease: Addressing Its Molecular, Clinical, and Therapeutic Aspects. Int J Mol Sci 2024; 25:7183. [PMID: 39000288 PMCID: PMC11241043 DOI: 10.3390/ijms25137183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Parkinson's disease (PD) is a gradually worsening neurodegenerative disorder affecting the nervous system, marked by a slow progression and varied symptoms. It is the second most common neurodegenerative disease, affecting over six million people in the world. Its multifactorial etiology includes environmental, genomic, and epigenetic factors. Clinical symptoms consist of non-motor and motor symptoms, with motor symptoms being the classic presentation. Therapeutic approaches encompass pharmacological, non-pharmacological, and surgical interventions. Traditional pharmacological treatment consists of administering drugs (MAOIs, DA, and levodopa), while emerging evidence explores the potential of antidiabetic agents for neuroprotection and gene therapy for attenuating parkinsonian symptoms. Non-pharmacological treatments, such as exercise, a calcium-rich diet, and adequate vitamin D supplementation, aim to slow disease progression and prevent complications. For those patients who have medically induced side effects and/or refractory symptoms, surgery is a therapeutic option. Deep brain stimulation is the primary surgical option, associated with motor symptom improvement. Levodopa/carbidopa intestinal gel infusion through percutaneous endoscopic gastrojejunostomy and a portable infusion pump succeeded in reducing "off" time, where non-motor and motor symptoms occur, and increasing "on" time. This article aims to address the general aspects of PD and to provide a comparative comprehensive review of the conventional and the latest therapeutic advancements and emerging treatments for PD. Nevertheless, further studies are required to optimize treatment and provide suitable alternatives.
Collapse
Affiliation(s)
- Mauricio Muleiro Alvarez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Gabriela Cano-Herrera
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - María Fernanda Osorio Martínez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | | | - Germán Rivera Monroy
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Renata Murguiondo Pérez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Jorge Alejandro Torres-Ríos
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Ximena A. van Tienhoven
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Ernesto Marcelo Garibaldi Bernot
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Felipe Esparza Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
- Secretaria de la Defensa Nacional, Escuela Militar de Graduados en Sanidad, Ciudad de México 11200, Mexico
| |
Collapse
|
2
|
Soto I, McManus R, Navarrete W, Kasanga EA, Doshier K, Nejtek VA, Salvatore MF. Aging accelerates locomotor decline in PINK1 knockout rats in association with decreased nigral, but not striatal, dopamine and tyrosine hydroxylase expression. Exp Neurol 2024; 376:114771. [PMID: 38580154 DOI: 10.1016/j.expneurol.2024.114771] [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: 02/01/2024] [Revised: 03/15/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Parkinson's disease (PD) rodent models provide insight into the relationship between nigrostriatal dopamine (DA) signaling and locomotor function. Although toxin-based rat models produce frank nigrostriatal neuron loss and eventual motor decline characteristic of PD, the rapid nature of neuronal loss may not adequately translate premotor traits, such as cognitive decline. Unfortunately, rodent genetic PD models, like the Pink1 knockout (KO) rat, often fail to replicate the differential severity of striatal DA and tyrosine hydroxylase (TH) loss, and a bradykinetic phenotype, reminiscent of human PD. To elucidate this inconsistency, we evaluated aging as a progression factor in the timing of motor and non-motor cognitive impairments. Male PINK1 KO and age-matched wild type (WT) rats were evaluated in a longitudinal study from 3 to 16 months old in one cohort, and in a cross-sectional study of young adult (6-7 months) and aged (18-19 months) in another cohort. Young adult PINK1 KO rats exhibited hyperkinetic behavior associated with elevated DA and TH in the substantia nigra (SN), which decreased therein, but not striatum, in the aged KO rats. Additionally, norepinephrine levels decreased in aged KO rats in the prefrontal cortex (PFC), paired with a higher DA levels in young and aged KO. Although a younger age of onset characterizes familial forms of PD, our results underscore the critical need to consider age-related factors. Moreover, the results indicate that compensatory mechanisms may exist to preserve locomotor function, evidenced by increased DA in the SN early in the lifespan, in response to deficient PINK1 function, which declines with aging and the onset of motor decline.
Collapse
Affiliation(s)
- Isabel Soto
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Robert McManus
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Walter Navarrete
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Ella A Kasanga
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Kirby Doshier
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Vicki A Nejtek
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America
| | - Michael F Salvatore
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, United States of America.
| |
Collapse
|
3
|
Soto I, McManus R, Navarrete-Barahona W, Kasanga EA, Doshier K, Nejtek VA, Salvatore MF. Aging hastens locomotor decline in PINK1 knockout rats in association with decreased nigral, but not striatal, dopamine and tyrosine hydroxylase expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578317. [PMID: 38352365 PMCID: PMC10862808 DOI: 10.1101/2024.02.01.578317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Parkinson's disease (PD) rodent models provide insight into the relationship between nigrostriatal dopamine (DA) signaling and locomotor function. Although toxin-based rat models produce frank nigrostriatal neuron loss and eventual motor decline characteristic of PD, the rapid nature of neuronal loss may not adequately translate premotor traits, such as cognitive decline. Unfortunately, rodent genetic PD models, like the Pink1 knockout (KO) rat, often fail to replicate the differential severity of striatal DA and tyrosine hydroxylase (TH) loss, and a bradykinetic phenotype, reminiscent of human PD. To elucidate this inconsistency, we evaluated aging as a progression factor in the timing of motor and non-motor cognitive impairments. Male PINK1 KO and age-matched wild type (WT) rats were evaluated in a longitudinal study from 3 to 16 months old in one cohort, and in a cross-sectional study of young adult (6-7 months) and aged (18-19 months) in another cohort. Young adult PINK1 KO rats exhibited hyperkinetic behavior associated with elevated DA and TH in the substantia nigra (SN), which decreased therein, but not striatum, in the aged KO rats. Additionally, norepinephrine levels decreased in aged KO rats in the prefrontal cortex (PFC), paired with a higher DA content in young and aged KO. Although a younger age of onset characterizes familial forms of PD, our results underscore the critical need to consider age-related factors. Moreover, the results indicate that compensatory mechanisms may exist to preserve locomotor function, evidenced by increased DA in the SN early in the lifespan, in response to deficient PINK1 function, which declines with aging and the onset of motor impairment.
Collapse
|
4
|
Hebbar M, Al-Taweel N, Gill I, Boelman C, Dean RA, Goodchild SJ, Mezeyova J, Shuart NG, Johnson JP, Lee J, Michoulas A, Huh LL, Armstrong L, Connolly MB, Demos MK. Expanding the genotype-phenotype spectrum in SCN8A-related disorders. BMC Neurol 2024; 24:31. [PMID: 38233770 PMCID: PMC10792783 DOI: 10.1186/s12883-023-03478-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND SCN8A-related disorders are a group of variable conditions caused by pathogenic variations in SCN8A. Online Mendelian Inheritance in Man (OMIM) terms them as developmental and epileptic encephalopathy 13, benign familial infantile seizures 5 or cognitive impairment with or without cerebellar ataxia. METHODS In this study, we describe clinical and genetic results on eight individuals from six families with SCN8A pathogenic variants identified via exome sequencing. RESULTS Clinical findings ranged from normal development with well-controlled epilepsy to significant developmental delay with treatment-resistant epilepsy. Three novel and three reported variants were observed in SCN8A. Electrophysiological analysis in transfected cells revealed a loss-of-function variant in Patient 4. CONCLUSIONS This work expands the clinical and genotypic spectrum of SCN8A-related disorders and provides electrophysiological results on a novel loss-of-function SCN8A variant.
Collapse
Affiliation(s)
- Malavika Hebbar
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Nawaf Al-Taweel
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Inderpal Gill
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Cyrus Boelman
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Richard A Dean
- Xenon Pharmaceuticals, 200-3650 Gilmore Way, Burnaby, BC, V5G 4W8, Canada
| | - Samuel J Goodchild
- Xenon Pharmaceuticals, 200-3650 Gilmore Way, Burnaby, BC, V5G 4W8, Canada
| | - Janette Mezeyova
- Xenon Pharmaceuticals, 200-3650 Gilmore Way, Burnaby, BC, V5G 4W8, Canada
| | | | - J P Johnson
- Xenon Pharmaceuticals, 200-3650 Gilmore Way, Burnaby, BC, V5G 4W8, Canada
| | - James Lee
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Aspasia Michoulas
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Linda L Huh
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Linlea Armstrong
- Department of Medical Genetics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mary B Connolly
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Michelle K Demos
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Henrich MT, Oertel WH, Surmeier DJ, Geibl FF. Mitochondrial dysfunction in Parkinson's disease - a key disease hallmark with therapeutic potential. Mol Neurodegener 2023; 18:83. [PMID: 37951933 PMCID: PMC10640762 DOI: 10.1186/s13024-023-00676-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023] Open
Abstract
Mitochondrial dysfunction is strongly implicated in the etiology of idiopathic and genetic Parkinson's disease (PD). However, strategies aimed at ameliorating mitochondrial dysfunction, including antioxidants, antidiabetic drugs, and iron chelators, have failed in disease-modification clinical trials. In this review, we summarize the cellular determinants of mitochondrial dysfunction, including impairment of electron transport chain complex 1, increased oxidative stress, disturbed mitochondrial quality control mechanisms, and cellular bioenergetic deficiency. In addition, we outline mitochondrial pathways to neurodegeneration in the current context of PD pathogenesis, and review past and current treatment strategies in an attempt to better understand why translational efforts thus far have been unsuccessful.
Collapse
Affiliation(s)
- Martin T Henrich
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Wolfgang H Oertel
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Fanni F Geibl
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany.
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany.
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
Menšíková K, Steele JC, Rosales R, Colosimo C, Spencer P, Lannuzel A, Ugawa Y, Sasaki R, Giménez-Roldán S, Matej R, Tuckova L, Hrabos D, Kolarikova K, Vodicka R, Vrtel R, Strnad M, Hlustik P, Otruba P, Prochazka M, Bares M, Boluda S, Buee L, Ransmayr G, Kaňovský P. Endemic parkinsonism: clusters, biology and clinical features. Nat Rev Neurol 2023; 19:599-616. [PMID: 37684518 DOI: 10.1038/s41582-023-00866-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 09/10/2023]
Abstract
The term 'endemic parkinsonism' refers to diseases that manifest with a dominant parkinsonian syndrome, which can be typical or atypical, and are present only in a particular geographically defined location or population. Ten phenotypes of endemic parkinsonism are currently known: three in the Western Pacific region; two in the Asian-Oceanic region; one in the Caribbean islands of Guadeloupe and Martinique; and four in Europe. Some of these disease entities seem to be disappearing over time and therefore are probably triggered by unique environmental factors. By contrast, other types persist because they are exclusively genetically determined. Given the geographical clustering and potential overlap in biological and clinical features of these exceptionally interesting diseases, this Review provides a historical reference text and offers current perspectives on each of the 10 phenotypes of endemic parkinsonism. Knowledge obtained from the study of these disease entities supports the hypothesis that both genetic and environmental factors contribute to the development of neurodegenerative diseases, not only in endemic parkinsonism but also in general. At the same time, this understanding suggests useful directions for further research in this area.
Collapse
Affiliation(s)
- Katerina Menšíková
- Department of Neurology and Clinical Neuroscience Center, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
- University Hospital, Olomouc, Czech Republic
| | | | - Raymond Rosales
- Research Center for Health Sciences, Faculty of Medicine and Surgery, University of Santo Tomás, Manila, The Philippines
- St Luke's Institute of Neuroscience, Metro, Manila, The Philippines
| | - Carlo Colosimo
- Department of Neurology, Santa Maria University Hospital, Terni, Italy
| | - Peter Spencer
- Department of Neurology, School of Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Annie Lannuzel
- Départment de Neurologie, Centre Hospitalier Universitaire de la Guadeloupe, Pointe-á-Pitre, France
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, Fukushima Medical University, Fukushima, Japan
| | - Ryogen Sasaki
- Department of Neurology, Kuwana City Medical Center, Kuwana, Japan
| | | | - Radoslav Matej
- Department of Pathology, 3rd Medical Faculty, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
- Department of Pathology and Molecular Medicine, 3rd Medical Faculty, Charles University and Thomayer University Hospital, Prague, Czech Republic
| | - Lucie Tuckova
- University Hospital, Olomouc, Czech Republic
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Dominik Hrabos
- University Hospital, Olomouc, Czech Republic
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Kristyna Kolarikova
- University Hospital, Olomouc, Czech Republic
- Department of Clinical and Molecular Genetics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Radek Vodicka
- University Hospital, Olomouc, Czech Republic
- Department of Clinical and Molecular Genetics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Radek Vrtel
- University Hospital, Olomouc, Czech Republic
- Department of Clinical and Molecular Genetics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Miroslav Strnad
- Department of Neurology and Clinical Neuroscience Center, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
- University Hospital, Olomouc, Czech Republic
- Laboratory of Growth Regulators, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Petr Hlustik
- Department of Neurology and Clinical Neuroscience Center, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
- University Hospital, Olomouc, Czech Republic
| | - Pavel Otruba
- Department of Neurology and Clinical Neuroscience Center, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
- University Hospital, Olomouc, Czech Republic
| | - Martin Prochazka
- University Hospital, Olomouc, Czech Republic
- Department of Clinical and Molecular Genetics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Martin Bares
- First Department of Neurology, Masaryk University Medical School, Brno, Czech Republic
- St Anne University Hospital, Brno, Czech Republic
| | - Susana Boluda
- Département de Neuropathologie, Hôpital La Pitié - Salpêtrière, Paris, France
| | - Luc Buee
- Lille Neuroscience & Cognition Research Centre, INSERM U1172, Lille, France
| | - Gerhard Ransmayr
- Department of Neurology, Faculty of Medicine, Johannes Kepler University, Linz, Austria
| | - Petr Kaňovský
- Department of Neurology and Clinical Neuroscience Center, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic.
- University Hospital, Olomouc, Czech Republic.
| |
Collapse
|
7
|
Ibrahim NM, Jagota P, Pal PK, Bhidayasiri R, Lim SY, Ugawa Y, Aldaajani Z, Jeon B, Fujioka S, Lee JY, Kukkle PL, Shang H, Phokaewvarangkul O, Diesta C, Shambetova C, Lin CH. Historical and More Common Nongenetic Movement Disorders From Asia. J Mov Disord 2023; 16:248-260. [PMID: 37291830 PMCID: PMC10548075 DOI: 10.14802/jmd.22224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/28/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Nongenetic movement disorders are common throughout the world. The movement disorders encountered may vary depending on the prevalence of certain disorders across various geographical regions. In this paper, we review historical and more common nongenetic movement disorders in Asia. The underlying causes of these movement disorders are diverse and include, among others, nutritional deficiencies, toxic and metabolic causes, and cultural Latah syndrome, contributed by geographical, economic, and cultural differences across Asia. The industrial revolution in Japan and Korea has led to diseases related to environmental toxin poisoning, such as Minamata disease and β-fluoroethyl acetate-associated cerebellar degeneration, respectively, while religious dietary restriction in the Indian subcontinent has led to infantile tremor syndrome related to vitamin B12 deficiency. In this review, we identify the salient features and key contributing factors in the development of these disorders.
Collapse
Affiliation(s)
- Norlinah Mohamed Ibrahim
- Neurology Unit, Department of Medicine, Faculty of Medicine, National University of Malaysia, Kuala Lumpur, Malaysia
| | - Priya Jagota
- Chulalongkorn Centre of Excellence for Parkinson’s Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bengaluru, Karnataka, India
| | - Roongroj Bhidayasiri
- Chulalongkorn Centre of Excellence for Parkinson’s Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Shen-Yang Lim
- The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson’s & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, Faculty of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Zakiyah Aldaajani
- Neurology Unit, King Fahad Military Medical Complex, Dhahran, Saudi Arabia
| | - Beomseok Jeon
- Department of Neurology, Seoul National University, Seoul, Korea
- Movement Disorder Center, Seoul National University Hospital, Seoul, Korea
| | - Shinsuke Fujioka
- Department of Neurology, Fukuoka University, Faculty of Medicine, Fukuoka, Japan
| | - Jee-Young Lee
- Department of Neurology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University Medical College, Seoul, Korea
| | - Prashanth Lingappa Kukkle
- Center for Parkinson’s Disease and Movement Disorders, Manipal Hospital, Bangalore, India
- Parkinson’s Disease and Movement Disorders Clinic, Bangalore, India
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Onanong Phokaewvarangkul
- Chulalongkorn Centre of Excellence for Parkinson’s Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Cid Diesta
- Section of Neurology, Department of Neuroscience, Makati Medical Center, NCR, Makati, Metro Manila, Philippines
| | | | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
Hebbar M, Al-Taweel N, Gill I, Boelman C, Dean RA, Goodchild SJ, Mezeyova J, Shuart NG, Johnson JP, Lee J, Michoulas A, Huh LL, Armstrong L, Connolly MB, Demos MK. Expanding the genotype-phenotype spectrum in SCN8A-related disorders. RESEARCH SQUARE 2023:rs.3.rs-3221902. [PMID: 37609289 PMCID: PMC10441468 DOI: 10.21203/rs.3.rs-3221902/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Background SCN8A-related disorders are a group of variable conditions caused by pathogenic variations in SCN8A. Online Mendelian Inheritance in Man (OMIM) terms them as developmental and epileptic encephalopathy 13, benign familial infantile seizures 5 or cognitive impairment with or without cerebellar ataxia. Methods In this study, we describe clinical and genetic results on eight individuals from six families with SCN8A pathogenic variants identified via exome sequencing. Results Clinical findings ranged from normal development with well-controlled epilepsy to significant developmental delay with treatment-resistant epilepsy. Three novel and three reported variants were observed in SCN8A. Electrophysiological analysis in transfected cells revealed a loss-of-function variant in Patient 4. Conclusions This work expands the clinical and genotypic spectrum of SCN8A-related disorders and provides electrophysiological results on a novel loss-of-function SCN8A variant.
Collapse
Affiliation(s)
- Malavika Hebbar
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Nawaf Al-Taweel
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Inderpal Gill
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Cyrus Boelman
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Richard A Dean
- Xenon Pharmaceuticals, 200-3650 Gilmore Way, Burnaby, BC V5G 4W8
| | | | - Janette Mezeyova
- Xenon Pharmaceuticals, 200-3650 Gilmore Way, Burnaby, BC V5G 4W8
| | | | - J P Johnson
- Xenon Pharmaceuticals, 200-3650 Gilmore Way, Burnaby, BC V5G 4W8
| | - James Lee
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Aspasia Michoulas
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Linda L Huh
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Linlea Armstrong
- Department of Medical Genetics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Mary B Connolly
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| | - Michelle K Demos
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, Faculty of Medicine, University of British Columbia, Vancouver BC
| |
Collapse
|
9
|
Lamberty BG, Estrella LD, Mattingly JE, Emanuel K, Trease A, Totusek S, Sheldon L, George JW, Almikhlafi MA, Farmer T, Stauch KL. Parkinson's disease relevant pathological features are manifested in male Pink1/Parkin deficient rats. Brain Behav Immun Health 2023; 31:100656. [PMID: 37484197 PMCID: PMC10362548 DOI: 10.1016/j.bbih.2023.100656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/25/2023] Open
Abstract
Animal disease models are important for neuroscience experimentation and in the study of neurodegenerative disorders. The major neurodegenerative disorder leading to motor impairments is Parkinson's disease (PD). The identification of hereditary forms of PD uncovered gene mutations and variants, such as loss-of-function mutations in PTEN-induced putative kinase 1 (Pink1) and the E3 ubiquitin ligase Parkin, two proteins involved in mitochondrial quality control, that could be harnessed to create animal models. However, to date, such models have not reproducibly recapitulated major aspects of the disease. Here, we describe the generation and phenotypic characterization of a combined Pink1/Parkin double knockout (dKO) rat, which reproducibly exhibits PD-relevant abnormalities, particularly in male animals. Motor dysfunction in Pink1/Parkin dKO rats was characterized by gait abnormalities and decreased rearing frequency, the latter of which was responsive to levodopa treatment. Pink1/Parkin dKO rats exhibited elevated plasma levels of neurofilament light chain and significant loss of tyrosine hydroxylase expression in the substantia nigra pars compacta (SNpc). Glial cell activation was also observed in the SNpc. Pink1/Parkin dKO rats showed elevated plasma and reduced cerebrospinal levels of alpha-synuclein as well as the presence of alpha-synuclein aggregates in the striatum. Further, the profile of circulating lymphocytes was altered, as elevated CD3+CD4+ T cells and reduced CD3+CD8+ T cells in Pink1/Parkin dKO rats were found. This coincided with mitochondrial dysfunction and infiltration of CD3+ T cells in the striatum. Altogether, the Pink1/Parkin dKO rats exhibited phenotypes similar to what is seen with PD patients, thus highlighting the suitability of this model for mechanistic studies of the role of Pink1 and Parkin in PD pathogenesis and as therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Kelly L. Stauch
- Corresponding author. Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
10
|
Kolicheski A, Turcano P, Tamvaka N, McLean PJ, Springer W, Savica R, Ross OA. Early-Onset Parkinson's Disease: Creating the Right Environment for a Genetic Disorder. JOURNAL OF PARKINSON'S DISEASE 2022; 12:2353-2367. [PMID: 36502340 PMCID: PMC9837689 DOI: 10.3233/jpd-223380] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) by its common understanding is a late-onset sporadic movement disorder. However, there is a need to recognize not only the fact that PD pathogenesis expands beyond (or perhaps to) the brain but also that many early-onset patients develop motor signs before the age of 50 years. Indeed, studies have shown that it is likely the protein aggregation observed in the brains of patients with PD precedes the motor symptoms by perhaps a decade. Studies on early-onset forms of PD have shown it to be a heterogeneous disease with multiple genetic and environmental factors determining risk of different forms of disease. Genetic and neuropathological evidence suggests that there are α-synuclein centric forms (e.g., SNCA genomic triplication), and forms that are driven by a breakdown in mitochondrial function and specifically in the process of mitophagy and clearance of damaged mitochondria (e.g., PARKIN and PINK1 recessive loss-of-function mutations). Aligning genetic forms with recognized environmental influences will help better define patients, aid prognosis, and hopefully lead to more accurately targeted clinical trial design. Work is now needed to understand the cross-talk between these two pathomechanisms and determine a sense of independence, it is noted that autopsies studies for both have shown the presence or absence of α-synuclein aggregation. The integration of genetic and environmental data is critical to understand the etiology of early-onset forms of PD and determine how the different pathomechanisms crosstalk.
Collapse
Affiliation(s)
- Ana Kolicheski
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Pierpaolo Turcano
- Department of Neurology, Mayo Clinic, Jacksonville, FL, USA,
Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Nicole Tamvaka
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA
| | - Pamela J. McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA
| | - Wolfdieter Springer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA
| | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA,
Mayo Graduate School, Neuroscience Track, Mayo Clinic, Jacksonville, FL, USA,
Department of Medicine, University College Dublin, Dublin, Ireland,
Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, USA,Department of Biology, University of NorthFlorida, Jacksonville, FL, USA,Correspondence to: Owen A. Ross, PhD, Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Road, Jacksonville, FL 32224, USA. Tel.: +1 904 953 6280; Fax: +1 904 953 7370; E-mail:
| |
Collapse
|
11
|
Nguyen TT, Kim YJ, Lai TT, Nguyen PT, Koh YH, Nguyen LTN, Ma HI, Kim YE. PTEN-Induced Putative Kinase 1 Dysfunction Accelerates Synucleinopathy. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1201-1217. [PMID: 35253778 PMCID: PMC9198758 DOI: 10.3233/jpd-213065] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Mutations in PTEN-induced putative kinase 1 (PINK1) cause autosomal recessive Parkinson’s disease (PD) and contribute to the risk of sporadic PD. However, the relationship between PD-related PINK1 mutations and alpha-synuclein (α-syn) aggregation—a main pathological component of PD—remains unexplored. Objective: To investigate whether α-syn pathology is exacerbated in the absence of PINK1 after α-syn preformed fibril (PFF) injection in a PD mouse model and its effects on neurodegeneration. Methods: In this study, 10-week-old Pink1 knockout (KO) and wildtype (WT) mice received stereotaxic unilateral striatal injection of recombinant mouse α-syn PFF. Then, α-syn pathology progression, inflammatory responses, and neurodegeneration were analyzed via immunohistochemistry, western blot analysis, and behavioral testing. Results: After PFF injection, the total α-syn levels significantly increased, and pathological α-syn was markedly aggregated in Pink1 KO mice compared with Pink1 WT mice. Then, earlier and more severe neuronal loss and motor deficits occurred. Moreover, compared with WT mice, Pink1 KO mice had evident microglial/astrocytic immunoreactivity and prolonged astrocytic activation, and a higher rate of protein phosphatase 2A phosphorylation, which might explain the greater α-syn aggravation and neuronal death. Conclusion: The loss of Pink1 function accelerated α-syn aggregation, accumulation and glial activation, thereby leading to early and significant neurodegeneration and behavioral impairment in the PD mouse model. Therefore, our findings support the notion that PINK1 dysfunction increases the risk of synucleinopathy.
Collapse
Affiliation(s)
- Tinh Thi Nguyen
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, South Korea
| | - Yun Joong Kim
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, South Korea
| | - Thuy Thi Lai
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, South Korea
| | - Phuong Thi Nguyen
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Seoul, South Korea
| | - Young Ho Koh
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, South Korea.,Ilsong Institute of Life Science, Hallym University, Seoul, South Korea
| | - Linh Thi Nhat Nguyen
- Department of Medical Sciences, Graduate School of Hallym University, Chuncheon, South Korea
| | - Hyeo-Il Ma
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, South Korea
| | - Young Eun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University, Anyang, South Korea.,Hallym Neurological Institute, Hallym University, South Korea
| |
Collapse
|
12
|
Morales‐Briceno H, Ong TL, Duma SR, Murray N, Pepper EM, Ha A, Tchan MC, Fung VS. Recurrent biallelic p.
L347P
PINK1
variant in Polynesians with Parkinsonism and isolated dopa‐responsive dystonia. Mov Disord Clin Pract 2022; 9:696-697. [PMID: 35844286 PMCID: PMC9274338 DOI: 10.1002/mdc3.13467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 03/28/2022] [Accepted: 04/09/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Hugo Morales‐Briceno
- Movement Disorders Unit, Department of Neurology Westmead Hospital Westmead NSW Australia
- Sydney Medical School The University of Sydney Sydney NSW Australia
| | - Tien Lee Ong
- Movement Disorders Unit, Department of Neurology Westmead Hospital Westmead NSW Australia
| | - Stephen R. Duma
- Movement Disorders Unit, Department of Neurology Westmead Hospital Westmead NSW Australia
- Sydney Medical School The University of Sydney Sydney NSW Australia
| | - Natalia Murray
- Movement Disorders Unit, Department of Neurology Westmead Hospital Westmead NSW Australia
- Sydney Medical School The University of Sydney Sydney NSW Australia
| | - Elizabeth M. Pepper
- Department of Neurology John Hunter Hospital, Lookout Road New Lambton NSW 2305
| | - Ainhi Ha
- Movement Disorders Unit, Department of Neurology Westmead Hospital Westmead NSW Australia
| | - Michel C. Tchan
- Department of Genetic Medicine Westmead Hospital Westmead NSW Australia
| | - Victor S.C. Fung
- Movement Disorders Unit, Department of Neurology Westmead Hospital Westmead NSW Australia
- Sydney Medical School The University of Sydney Sydney NSW Australia
| |
Collapse
|
13
|
Guo L, Jing Y. Construction and Identification of a Novel 5-Gene Signature for Predicting the Prognosis in Breast Cancer. Front Med (Lausanne) 2021; 8:669931. [PMID: 34722557 PMCID: PMC8551811 DOI: 10.3389/fmed.2021.669931] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Breast cancer is one of the most common malignancies in women worldwide. The purpose of this study was to identify the hub genes and construct prognostic signature that could predict the survival of patients with breast cancer (BC). Methods: We identified differentially expressed genes between the responder group and non-responder group based on the GEO cohort. Drug-resistance hub genes were identified by weighted gene co-expression network analysis, and a multigene risk model was constructed by univariate and multivariate Cox regression analysis based on the TCGA cohort. Immune cell infiltration and mutation characteristics were analyzed. Results: A 5-gene signature (GP6, MAK, DCTN2, TMEM156, and FKBP14) was constructed as a prognostic risk model. The 5-gene signature demonstrated favorable prediction performance in different cohorts, and it has been confirmed that the signature was an independent risk indicater. The nomogram comprising 5-gene signature showed better performance compared with other clinical features, Further, in the high-risk group, high M2 macrophage scores were related with bad prognosis, and the frequency of TP53 mutations was greater in the high-risk group than in the low-risk group. In the low-risk group, high CD8+ T cell scores were associated with a good prognosis, and the frequency of CDH1 mutations was greater in the low-risk group than that in the high-risk group. At the same time, patients in the low risk group have a good response to immunotherapy in terms of immunotherapy. The results of immunohistochemistry showed that MAK, GP6, and TEMEM156 were significantly highly expressed in tumor tissues, and DCTN2 was highly expressed in normal tissues. Conclusions: Our study may find potential new targets against breast cancer, and provide new insight into the underlying mechanisms.
Collapse
Affiliation(s)
- Lingling Guo
- Department of Ultrasound, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yu Jing
- Clinical Trial Ward of the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
14
|
Spencer PS. Parkinsonism and motor neuron disorders: Lessons from Western Pacific ALS/PDC. J Neurol Sci 2021; 433:120021. [PMID: 34635325 DOI: 10.1016/j.jns.2021.120021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/06/2021] [Accepted: 09/01/2021] [Indexed: 01/16/2023]
Abstract
Recognized worldwide as an unusual "overlap" syndrome, Parkinsonism and motor neuron disease, with or without dementia, is best exemplified by the former high-incidence clusters of Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia Complex (ALS/PDC) in Guam, USA, in the Kii Peninsula of Honshu Island, Japan, and in Papua, Indonesia, on the western side of New Guinea. Western Pacific ALS/PDC is a disappearing neurodegenerative disorder with multiple and sometime overlapping phenotypes (ALS, atypical parkinsonism, dementia) that appear to constitute a single disease of environmental origin, in particular from exposure to genotoxins/neurotoxins in seed of cycad plants (Cycas spp.) formerly used as a traditional source of food (Guam) and/or medicine (Guam, Kii-Japan, Papua-Indonesia). Seed compounds include the principal cycad toxin cycasin, its active metabolite methylazoxymethanol (MAM) and a non-protein amino acid β-N-methylamino-L-alanine (L-BMAA); each reproduces components of ALS/PDC neuropathology when individually administered to laboratory species in single doses perinatally (MAM, L-BMAA) or repeatedly for prolonged periods to young adult animals (L-BMAA). Human exposure to MAM, a potent DNA-alkylating mutagen, also has potential relevance to the high incidence of diverse mutations found among Guamanians with/without ALS/PDC. In sum, seven decades of intensive study of ALS/PDC has revealed field and laboratory approaches leading to discovery of disease etiology that are now being applied to sporadic neurodegenerative disorders such as ALS beyond the Western Pacific region. This article is part of the Special Issue "Parkinsonism across the spectrum of movement disorders and beyond" edited by Joseph Jankovic, Daniel D. Truong and Matteo Bologna.
Collapse
Affiliation(s)
- Peter S Spencer
- Department of Neurology, School of Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
15
|
Yao L, Wu J, Koc S, Lu G. Genetic Imaging of Neuroinflammation in Parkinson's Disease: Recent Advancements. Front Cell Dev Biol 2021; 9:655819. [PMID: 34336822 PMCID: PMC8320775 DOI: 10.3389/fcell.2021.655819] [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: 01/19/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative aging disorders characterized by motor and non-motor symptoms due to the selective loss of midbrain dopaminergic (DA) neurons. The decreased viability of DA neurons slowly results in the appearance of motor symptoms such as rigidity, bradykinesia, resting tremor, and postural instability. These symptoms largely depend on DA nigrostriatal denervation. Pharmacological and surgical interventions are the main treatment for improving clinical symptoms, but it has not been possible to cure PD. Furthermore, the cause of neurodegeneration remains unclear. One of the possible neurodegeneration mechanisms is a chronic inflammation of the central nervous system, which is mediated by microglial cells. Impaired or dead DA neurons can directly lead to microglia activation, producing a large number of reactive oxygen species and pro-inflammatory cytokines. These cytotoxic factors contribute to the apoptosis and death of DA neurons, and the pathological process of neuroinflammation aggravates the primary morbid process and exacerbates ongoing neurodegeneration. Therefore, anti-inflammatory treatment exerts a robust neuroprotective effect in a mouse model of PD. Since discovering the first mutation in the α-synuclein gene (SNCA), which can cause disease-causing, PD has involved many genes and loci such as LRRK2, Parkin, SNCA, and PINK1. In this article, we summarize the critical descriptions of the genetic factors involved in PD's occurrence and development (such as LRRK2, SNCA, Parkin, PINK1, and inflammasome), and these factors play a crucial role in neuroinflammation. Regulation of these signaling pathways and molecular factors related to these genetic factors can vastly improve the neuroinflammation of PD.
Collapse
Affiliation(s)
- Longping Yao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiayu Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sumeyye Koc
- Department of Neuroscience, Institute of Health Sciences, Ondokuz Mayıs University, Samsun, Turkey
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
16
|
Patel SG, Buchanan CM, Mulroy E, Simpson M, Reid HA, Drake KM, Merriman ME, Phipps-Green A, Cadzow M, Merriman TR, Anderson NE, Child N, Barber PA, Roxburgh RH. Potential PINK1 Founder Effect in Polynesia Causing Early-Onset Parkinson's Disease. Mov Disord 2021; 36:2199-2200. [PMID: 34159639 DOI: 10.1002/mds.28665] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
| | | | - Eoin Mulroy
- Auckland Hospital, Auckland, New Zealand.,Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom
| | | | - Hannah A Reid
- Canterbury Health Laboratories, Christchurch, New Zealand
| | - Kylie M Drake
- Canterbury Health Laboratories, Christchurch, New Zealand
| | | | | | - Murray Cadzow
- Biochemistry Department, University of Otago, Dunedin, New Zealand
| | - Tony R Merriman
- Biochemistry Department, University of Otago, Dunedin, New Zealand.,Division of Clinical Immunology and Rheumatology, University of Alabama, Birmingham, Alabama, USA
| | | | | | | | - Richard H Roxburgh
- Auckland Hospital, Auckland, New Zealand.,Centre for Brain Research Neurogenetics Research Clinic, University of Auckland, Auckland, New Zealand
| |
Collapse
|
17
|
Su X, Li H, Chen S, Qin C. Study on the Prognostic Values of Dynactin Genes in Low-Grade Glioma. Technol Cancer Res Treat 2021; 20:15330338211010143. [PMID: 33896271 PMCID: PMC8085377 DOI: 10.1177/15330338211010143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE This present study aims to investigate the potential prognostic values of dynactin genes (DCTN) for predicting the overall survival (OS) in low-grade glioma (LGG) patients. METHODS The DCTN mRNA expression data were downloaded from The Cancer Genome Atlas database containing 518 patients with LGG. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses for DCTN genes were performed by using Database for Annotation, Visualization, and Integrated Discovery platform, and their enrichment results were verified by using the Biological Networks Gene Ontology tool. Next, the correlations between DCTN genes and LGG were identified by Pearson correlation coefficient analysis. The OS was estimated by Kaplan-Meier survival analysis. The cBio Cancer Genomics Portal was used to analyze the mutations of DCTN genes and their effects on the prognosis of LGG. The correlation between the abundance of immune infiltration and tumor purity of DCTN genes were predicted by The Tumor Immune Estimation Resource. RESULTS Our research showed that the mRNA expression of DCTN4 in tumor tissues was much higher (P < 0.01) than that in normal tissues. Meanwhile, there was a certain correlation between the DCTN genes. Survival analysis showed that the high expression of DCTN1, DCTN3, DCTN4, DCTN6, and their co-expression were significantly correlated with favorable OS in LGG patients (P < 0.05). In DCTN2, a high mutation rate was observed. Further research showed that the genetic alteration in DCTN genes was related to a poor OS and progression-free survival of LGG patients. The expression of DCTN genes had a certain correlation with immune infiltrating cells. CONCLUSION Our study showed that the high expressions of DCTN1, DCTN3, DCTN4, and DCTN6 were associated with a favorable OS of LGG patients, indicating that these DCTN genes are potential biomarkers for evaluating the prognosis of LGG patients.
Collapse
Affiliation(s)
- Xiaotao Su
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Haoyu Li
- Department of Ophthalmology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Shaohua Chen
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Chao Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
18
|
Nicoletti V, Palermo G, Del Prete E, Mancuso M, Ceravolo R. Understanding the Multiple Role of Mitochondria in Parkinson's Disease and Related Disorders: Lesson From Genetics and Protein-Interaction Network. Front Cell Dev Biol 2021; 9:636506. [PMID: 33869180 PMCID: PMC8047151 DOI: 10.3389/fcell.2021.636506] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
As neurons are highly energy-demanding cell, increasing evidence suggests that mitochondria play a large role in several age-related neurodegenerative diseases. Synaptic damage and mitochondrial dysfunction have been associated with early events in the pathogenesis of major neurodegenerative diseases, including Parkinson’s disease, atypical parkinsonisms, and Huntington disease. Disruption of mitochondrial structure and dynamic is linked to increased levels of reactive oxygen species production, abnormal intracellular calcium levels, and reduced mitochondrial ATP production. However, recent research has uncovered a much more complex involvement of mitochondria in such disorders than has previously been appreciated, and a remarkable number of genes and proteins that contribute to the neurodegeneration cascade interact with mitochondria or affect mitochondrial function. In this review, we aim to summarize and discuss the deep interconnections between mitochondrial dysfunction and basal ganglia disorders, with an emphasis into the molecular triggers to the disease process. Understanding the regulation of mitochondrial pathways may be beneficial in finding pharmacological or non-pharmacological interventions to delay the onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Valentina Nicoletti
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Palermo
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Del Prete
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michelangelo Mancuso
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
Erskine D, Koss D, Korolchuk VI, Outeiro TF, Attems J, McKeith I. Lipids, lysosomes and mitochondria: insights into Lewy body formation from rare monogenic disorders. Acta Neuropathol 2021; 141:511-526. [PMID: 33515275 PMCID: PMC7952289 DOI: 10.1007/s00401-021-02266-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
Accumulation of the protein α-synuclein into insoluble intracellular deposits termed Lewy bodies (LBs) is the characteristic neuropathological feature of LB diseases, such as Parkinson's disease (PD), Parkinson's disease dementia (PDD) and dementia with LB (DLB). α-Synuclein aggregation is thought to be a critical pathogenic event in the aetiology of LB disease, based on genetic analyses, fundamental studies using model systems, and the observation of LB pathology in post-mortem tissue. However, some monogenic disorders not traditionally characterised as synucleinopathies, such as lysosomal storage disorders, iron storage disorders and mitochondrial diseases, appear disproportionately vulnerable to the deposition of LBs, perhaps suggesting the process of LB formation may be a result of processes perturbed as a result of these conditions. The present review discusses biological pathways common to monogenic disorders associated with LB formation, identifying catabolic processes, particularly related to lipid homeostasis, autophagy and mitochondrial function, as processes that could contribute to LB formation. These findings are discussed in the context of known mediators of α-synuclein aggregation, highlighting the potential influence of impairments to these processes in the aetiology of LB formation.
Collapse
Affiliation(s)
- Daniel Erskine
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
- Wellcome Centre for Mitochondrial Research, Newcastle upon Tyne, UK.
| | - David Koss
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Viktor I Korolchuk
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tiago F Outeiro
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Max Planck Institute for Experimental Medicine, Goettingen, Germany
- Scientific Employee With an Honorary Contract at Deutsches Zentrum Für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Johannes Attems
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ian McKeith
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
20
|
Western Pacific ALS-PDC: Evidence implicating cycad genotoxins. J Neurol Sci 2020; 419:117185. [PMID: 33190068 DOI: 10.1016/j.jns.2020.117185] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/20/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022]
Abstract
Amyotrophic Lateral Sclerosis and Parkinsonism-Dementia Complex (ALS-PDC) is a disappearing neurodegenerative disorder of apparent environmental origin formerly hyperendemic among Chamorros of Guam-USA, Japanese residents of the Kii Peninsula, Honshu Island, Japan and Auyu-Jakai linguistic groups of Papua-Indonesia on the island of New Guinea. The most plausible etiology is exposure to genotoxins in seed of neurotoxic cycad plants formerly used for food and/or medicine. Primary suspicion falls on methylazoxymethanol (MAM), the aglycone of cycasin and on the non-protein amino acid β-N-methylamino-L-alanine, both of which are metabolized to formaldehyde. Human and animal studies suggest: (a) exposures occurred early in life and sometimes during late fetal brain development, (b) clinical expression of neurodegenerative disease appeared years or decades later, and (c) pathological changes in various tissues indicate the disease was not confined to the CNS. Experimental evidence points to toxic molecular mechanisms involving DNA damage, epigenetic changes, transcriptional mutagenesis, neuronal cell-cycle reactivation and perturbation of the ubiquitin-proteasome system that led to polyproteinopathy and culminated in neuronal degeneration. Lessons learned from research on ALS-PDC include: (a) familial disease may reflect common toxic exposures across generations, (b) primary disease prevention follows cessation of exposure to culpable environmental triggers; and (c) disease latency provides a prolonged period during which to intervene therapeutically. Exposure to genotoxic chemicals ("slow toxins") in the early stages of life should be considered in the search for the etiology of ALS-PDC-related neurodegenerative disorders, including sporadic forms of ALS, progressive supranuclear palsy and Alzheimer's disease.
Collapse
|
21
|
Tan AH, Lohmann K, Tay YW, Lim JL, Ahmad-Annuar A, Ramli N, Chin YT, Mawardi AS, Azmi K, Aziz ZA, Puvanarajah SD, Bauer P, Klein C, Rolfs A, Lim SY. PINK1 p.Leu347Pro mutations in Malays: Prevalence and illustrative cases. Parkinsonism Relat Disord 2020; 79:34-39. [DOI: 10.1016/j.parkreldis.2020.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/30/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
|
22
|
Neuropathological findings in PINK1-associated Parkinson's disease. Parkinsonism Relat Disord 2020; 78:105-108. [PMID: 32814227 DOI: 10.1016/j.parkreldis.2020.07.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Biallelic mutations in PTEN-induced putative kinase 1 (PINK1) is a relatively common cause of autosomal recessive early-onset Parkinson's disease (PD). However, only three PINK1 patients with brain autopsy have been reported in the literature. METHODS We describe the clinical and pathological characteristics of a patient with early-onset PD. We screened for copy number variants SNCA, PRKN, PINK1, DJ-1, ATP13A2, LPA and TNFRSF9 by multiplex ligation-dependent probe amplification (MLPA), and subsequently we performed whole-exome sequencing. RESULTS Clinically the patient presented with typical parkinsonism that responded well to levodopa. After 23 years of disease she had a bilateral GPi deep brain stimulation (DBS) surgery. Genetic analyses revealed a heterozygous exon 4-5 deletion and a homozygous exon 1 [c. 230T > C (p.Leu77Pro)] mutation in PINK1. Post-mortem neuropathological examination after more than 30 years of disease revealed gliosis and a large loss of melanin-containing neurons in the substantia nigra. Lewy body pathology was evident in substantia nigra, temporal cortex, locus coeruleus and the parahippocampal region. CONCLUSION We describe the first clinical and pathological characterization of a PINK1 patient with a typical disease presentation and long disease duration. Previous reports describe two patients with Lewy-related pathologies, albeit with differential distribution, and one patient with no Lewy-related pathology. Hence, it seems that only two patients with parkinsonism due to mutations in PINK1 are consistent with α-synucleinopathy distribution like that seen in the majority of cases with sporadic PD. Our data further extend the clinicopathological characterization of PINK1-associated PD.
Collapse
|
23
|
Aasly JO. Long-Term Outcomes of Genetic Parkinson's Disease. J Mov Disord 2020; 13:81-96. [PMID: 32498494 PMCID: PMC7280945 DOI: 10.14802/jmd.19080] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/23/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder that affects 1–2% of people by the age of 70 years. Age is the most important risk factor, and most cases are sporadic without any known environmental or genetic causes. Since the late 1990s, mutations in the genes SNCA, PRKN, LRRK2, PINK1, DJ-1, VPS35, and GBA have been shown to be important risk factors for PD. In addition, common variants with small effect sizes are now recognized to modulate the risk for PD. Most studies in genetic PD have focused on finding new genes, but few have studied the long-term outcome of patients with the specific genetic PD forms. Patients with known genetic PD have now been followed for more than 20 years, and we see that they may have distinct and different prognoses. New therapeutic possibilities are emerging based on the genetic cause underlying the disease. Future medication may be based on the pathophysiology individualized to the patient’s genetic background. The challenge is to find the biological consequences of different genetic variants. In this review, the clinical patterns and long-term prognoses of the most common genetic PD variants are presented.
Collapse
Affiliation(s)
- Jan O Aasly
- Department of Neurology, St. Olav's Hospital, Trondheim, Norway.,Department of Neuroscience, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
24
|
Oluwole OG, Kuivaniemi H, Abrahams S, Haylett WL, Vorster AA, van Heerden CJ, Kenyon CP, Tabb DL, Fawale MB, Sunmonu TA, Ajose A, Olaogun MO, Rossouw AC, van Hillegondsberg LS, Carr J, Ross OA, Komolafe MA, Tromp G, Bardien S. Targeted next-generation sequencing identifies novel variants in candidate genes for Parkinson's disease in Black South African and Nigerian patients. BMC MEDICAL GENETICS 2020; 21:23. [PMID: 32019516 PMCID: PMC7001245 DOI: 10.1186/s12881-020-0953-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 01/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The prevalence of Parkinson's disease (PD) is increasing in sub-Saharan Africa, but little is known about the genetics of PD in these populations. Due to their unique ancestry and diversity, sub-Saharan African populations have the potential to reveal novel insights into the pathobiology of PD. In this study, we aimed to characterise the genetic variation in known and novel PD genes in a group of Black South African and Nigerian patients. METHODS We recruited 33 Black South African and 14 Nigerian PD patients, and screened them for sequence variants in 751 genes using an Ion AmpliSeq™ Neurological Research panel. We used bcftools to filter variants and annovar software for the annotation. Rare variants were prioritised using MetaLR and MetaSVM prediction scores. The effect of a variant on ATP13A2's protein structure was investigated by molecular modelling. RESULTS We identified 14,655 rare variants with a minor allele frequency ≤ 0.01, which included 2448 missense variants. Notably, no common pathogenic mutations were identified in these patients. Also, none of the known PD-associated mutations were found highlighting the need for more studies in African populations. Altogether, 54 rare variants in 42 genes were considered deleterious and were prioritized, based on MetaLR and MetaSVM scores, for follow-up studies. Protein modelling showed that the S1004R variant in ATP13A2 possibly alters the conformation of the protein. CONCLUSIONS We identified several rare variants predicted to be deleterious in sub-Saharan Africa PD patients; however, further studies are required to determine the biological effects of these variants and their possible role in PD. Studies such as these are important to elucidate the genetic aetiology of this disorder in patients of African ancestry.
Collapse
Affiliation(s)
- Oluwafemi G Oluwole
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Shameemah Abrahams
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - William L Haylett
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Alvera A Vorster
- DNA Sequencing Unit, Central Analytical Facility, Stellenbosch University, Stellenbosch, South Africa
| | - Carel J van Heerden
- DNA Sequencing Unit, Central Analytical Facility, Stellenbosch University, Stellenbosch, South Africa
| | - Colin P Kenyon
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- Bioinformatics Unit, South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - David L Tabb
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- Bioinformatics Unit, South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Stellenbosch, South Africa
| | - Michael B Fawale
- Neurology Unit, Department of Medicine, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Taofiki A Sunmonu
- Neurology Unit, Department of Medicine, Federal Medical Centre, Owo, Nigeria
| | - Abiodun Ajose
- Department of Chemical Pathology, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Matthew O Olaogun
- Department of Medical Rehabilitation, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Anastasia C Rossouw
- Division of Neurology, Department of Medicine, Faculty of Health Sciences, Walter Sisulu University, East London, South Africa
| | - Ludo S van Hillegondsberg
- Division of Neurology, Department of Medicine, Faculty of Health Sciences, Walter Sisulu University, East London, South Africa
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jonathan Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
- Department of Clinical Genomics, Mayo Clinic College of Medicine, Jacksonville, Florida, USA
| | - Morenikeji A Komolafe
- Neurology Unit, Department of Medicine, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.
- Bioinformatics Unit, South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa.
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa.
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa.
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Stellenbosch, South Africa.
| | - Soraya Bardien
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
25
|
Barodia SK, McMeekin LJ, Creed RB, Quinones EK, Cowell RM, Goldberg MS. PINK1 phosphorylates ubiquitin predominantly in astrocytes. NPJ PARKINSONS DISEASE 2019; 5:29. [PMID: 31840043 PMCID: PMC6906478 DOI: 10.1038/s41531-019-0101-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022]
Abstract
Loss-of-function mutations in PINK1 are causally linked to recessively inherited Parkinson’s disease (PD), with marked loss of dopaminergic neurons in the substantia nigra that are required for normal movement. PINK1 is a nuclear-encoded mitochondrial-targeted kinase that phosphorylates a conserved serine at amino acid 65 (pS65) in ubiquitin as well as Parkin, another gene with loss-of-function mutations linked to recessive parkinsonism. The steady-state levels of PINK1 protein are very low, even in cells that express PINK1, because PINK1 is normally targeted for degradation after mitochondrial import by a process that is dependent upon mitochondrial membrane potential. Dissipation of the mitochondrial membrane potential with ionophores, such as CCCP and valinomycin, causes the accumulation of PINK1 on the outer mitochondrial membrane, a marked increase of pS65-ubiquitin and the recruitment of Parkin, which targets dysfunctional mitochondria for degradation by autophagy. While the high penetrance of PINK1 mutations establish its critical function for maintaining neurons, the activity of PINK1 in primary neurons has been difficult to detect. Mounting evidence implicates non-neuronal cells, including astrocytes and microglia, in the pathogenesis of both idiopathic and inherited PD. Herein we used both western analysis and immunofluorescence of pS65-ubiquitin to directly compare the activity of PINK1 in primary neurons, astrocytes, microglia, and oligodendrocyte progenitor cells cultured from the brains of wild-type (WT) and PINK1 knockout (KO) rat pups. Our findings that PINK1-dependent ubiquitin phosphorylation is predominantly in astrocytes supports increased priority for research on the function of PINK1 in astrocytes and the contribution of astrocyte dysfunction to PD pathogenesis.
Collapse
Affiliation(s)
- Sandeep K Barodia
- 1Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Laura J McMeekin
- 2Department of Neuroscience, Southern Research, Birmingham, AL 35205 USA.,3Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Rose B Creed
- 1Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Elijah K Quinones
- 1Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Rita M Cowell
- 2Department of Neuroscience, Southern Research, Birmingham, AL 35205 USA.,3Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Matthew S Goldberg
- 1Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA.,4Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| |
Collapse
|
26
|
Pathophysiology of and therapeutic options for a GABRA1 variant linked to epileptic encephalopathy. Mol Brain 2019; 12:92. [PMID: 31707987 PMCID: PMC6842544 DOI: 10.1186/s13041-019-0513-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/14/2019] [Indexed: 01/07/2023] Open
Abstract
We report the identification of a de novo GABRA1 (R214C) variant in a child with epileptic encephalopathy (EE), describe its functional characterization and pathophysiology, and evaluate its potential therapeutic options. The GABRA1 (R214C) variant was identified using whole exome sequencing, and the pathogenic effect of this mutation was investigated by comparing wild-type (WT) α1 and R214C α1 GABAA receptor-expressing HEK cells. GABA-evoked currents in these cells were recorded using whole-cell, outside-out macro-patch and cell-attached single-channel patch-clamp recordings. Changes to surface and total protein expression levels of WT α1 and R214C α1 were quantified using surface biotinylation assay and western blotting, respectively. Finally, potential therapeutic options were explored by determining the effects of modulators, including diazepam, insulin, and verapamil, on channel gating and receptor trafficking of WT and R214C GABAA receptors. We found that the GABRA1 (R214C) variant decreased whole-cell GABA-evoked currents by reducing single channel open time and both surface and total GABAA receptor expression levels. The GABA-evoked currents in R214C GABAA receptors could only be partially restored with benzodiazepine (diazepam) and insulin. However, verapamil treatment for 24 h fully restored the function of R214C mutant receptors, primarily by increasing channel open time. We conclude that the GABRA1 (R214C) variant reduces channel activity and surface expression of mutant receptors, thereby contributing to the pathogenesis of genetic EE. The functional restoration by verapamil suggests that it is a potentially new therapeutic option for patients with the R214C variant and highlights the value of precision medicine in the treatment of genetic EEs.
Collapse
|
27
|
Niemann N, Jankovic J. Juvenile parkinsonism: Differential diagnosis, genetics, and treatment. Parkinsonism Relat Disord 2019; 67:74-89. [DOI: 10.1016/j.parkreldis.2019.06.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/24/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022]
|
28
|
Mishima T, Fujioka S, Tsuboi Y. Perry disease: recent advances and perspectives. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1625766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
| | | | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
29
|
Ryan M, Zaldívar Vaillant T, McLaughlin RL, Doherty MA, Rooney J, Heverin M, Gutierrez J, Lara-Fernández GE, Pita Rodríguez M, Hackembruch J, Perna A, Vazquez MC, Musio M, Ketzoian CN, Logroscino G, Hardiman O. Comparison of the clinical and genetic features of amyotrophic lateral sclerosis across Cuban, Uruguayan and Irish clinic-based populations. J Neurol Neurosurg Psychiatry 2019; 90:659-665. [PMID: 30846540 DOI: 10.1136/jnnp-2018-319838] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/19/2018] [Accepted: 01/15/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVES This study compares the clinical characteristics of patients with amyotrophic lateral sclerosis (ALS) within three clinic-based populations from Cuba, Uruguay and Ireland and determines the impact of known ALS-associated genetic variants on phenotypic manifestations within the Cuban population. METHODS Demographic and clinical information was collected on 115 Cuban, 220 Uruguayan and 1038 Irish patients with ALS attending national specialist clinics through 1996-2017. All Cuban patients and 676 Irish patients underwent next-generation DNA sequencing and were screened for the pathogenic C9orf72 repeat expansion. RESULTS The mean age of onset was younger in the Cuban (53.0 years, 95% CI 50.4 to 55.6) and Uruguayan (58.2 years, 95% CI 56.5 to 60.0) populations compared with the Irish population (61.6 years, 95% CI 60.9 to 62.4). No differences in survival between populations were observed. 1.7 % (95% CI 0.6 to 4.1) of Cubans with ALS carried the C9orf72 repeat expansion compared with 9.9% (95% CI 7.8 to 12.0) of Irish patients with ALS (p=0.004). Other known variants identified in the Cuban population included ANG (one patient), CHCHD10 (one patient) and DCTN1 (three patients). CONCLUSIONS AND RELEVANCE This study is the first to describe the clinical characteristics of ALS in Cuban and Uruguayan populations and report differences between the Cuban and Irish genetic signature in terms of known ALS-associated genetic variants. These novel clinical and genetic data add to our understanding of ALS across different and understudied populations.
Collapse
Affiliation(s)
- Marie Ryan
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | | | | | - Mark A Doherty
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - James Rooney
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | - Mark Heverin
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland
| | | | | | | | - Jochen Hackembruch
- Institute of Neurology, Hospital de Clínicas, School of Medicine, University of the Republic, Montevideo, Uruguay
| | - Abayubá Perna
- Institute of Neurology, Hospital de Clínicas, School of Medicine, University of the Republic, Montevideo, Uruguay
| | - Maria Cristina Vazquez
- Institute of Neurology, Hospital de Clínicas, School of Medicine, University of the Republic, Montevideo, Uruguay
| | - Marco Musio
- Unit of Neurodegenerative Diseases, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro', Pia Fondazione Cardinale G Panico, Lecce, Italy
| | - Carlos N Ketzoian
- Institute of Neurology, Hospital de Clínicas, School of Medicine, University of the Republic, Montevideo, Uruguay
| | - Giancarlo Logroscino
- Unit of Neurodegenerative Diseases, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro', Pia Fondazione Cardinale G Panico, Lecce, Italy.,Department of Basic Medical Sciences, Neurosciences and Sense Organs, Universita degli Studi di Bari Aldo Moro, Bari, Italy
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity College Dublin, Dublin, Ireland.,Neurology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
Demos M, Guella I, DeGuzman C, McKenzie MB, Buerki SE, Evans DM, Toyota EB, Boelman C, Huh LL, Datta A, Michoulas A, Selby K, Bjornson BH, Horvath G, Lopez-Rangel E, van Karnebeek CDM, Salvarinova R, Slade E, Eydoux P, Adam S, Van Allen MI, Nelson TN, Bolbocean C, Connolly MB, Farrer MJ. Diagnostic Yield and Treatment Impact of Targeted Exome Sequencing in Early-Onset Epilepsy. Front Neurol 2019; 10:434. [PMID: 31164858 PMCID: PMC6536592 DOI: 10.3389/fneur.2019.00434] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
Targeted whole-exome sequencing (WES) is a powerful diagnostic tool for a broad spectrum of heterogeneous neurological disorders. Here, we aim to examine the impact on diagnosis, treatment and cost with early use of targeted WES in early-onset epilepsy. WES was performed on 180 patients with early-onset epilepsy (≤5 years) of unknown cause. Patients were classified as Retrospective (epilepsy diagnosis >6 months) or Prospective (epilepsy diagnosis <6 months). WES was performed on an Ion Proton™ and variant reporting was restricted to the sequences of 620 known epilepsy genes. Diagnostic yield and time to diagnosis were calculated. An analysis of cost and impact on treatment was also performed. A molecular diagnoses (pathogenic/likely pathogenic variants) was achieved in 59/180 patients (33%). Clinical management changed following WES findings in 23 of 59 diagnosed patients (39%) or 13% of all patients. A possible diagnosis was identified in 21 additional patients (12%) for whom supporting evidence is pending. Time from epilepsy onset to a genetic diagnosis was faster when WES was performed early in the diagnostic process (mean: 145 days Prospective vs. 2,882 days Retrospective). Costs of prior negative tests averaged $8,344 per patient in the Retrospective group, suggesting savings of $5,110 per patient using WES. These results highlight the diagnostic yield, clinical utility and potential cost-effectiveness of using targeted WES early in the diagnostic workup of patients with unexplained early-onset epilepsy. The costs and clinical benefits are likely to continue to improve. Advances in precision medicine and further studies regarding impact on long-term clinical outcome will be important.
Collapse
Affiliation(s)
- Michelle Demos
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Ilaria Guella
- Department of Medical Genetics, Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Conrado DeGuzman
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Marna B McKenzie
- Department of Medical Genetics, Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Sarah E Buerki
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada.,Division of Neuropediatrics, University Children's Hospital Zurich, Zurich, Switzerland
| | - Daniel M Evans
- Department of Medical Genetics, Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Eric B Toyota
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Cyrus Boelman
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Linda L Huh
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Anita Datta
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Aspasia Michoulas
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Kathryn Selby
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Bruce H Bjornson
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Gabriella Horvath
- Division of Biochemical Diseases, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Elena Lopez-Rangel
- Division of Developmental Pediatrics, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Clara D M van Karnebeek
- Department of Pediatrics, Centre for Molecular Medicine and Therapeutics, BCCHRI, University of British Columbia, Vancouver, BC, Canada.,Department of Pediatrics, Academic Medical Centre, Amsterdam, Netherlands
| | - Ramona Salvarinova
- Division of Biochemical Diseases, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Erin Slade
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Patrice Eydoux
- Division of Genome Diagnostics, Department of Pathology and Laboratory Medicine, BC Children's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Shelin Adam
- Department of Medical Genetics, BC Children's and BC's Women's Hospitals, University of British Columbia, Vancouver, BC, Canada
| | - Margot I Van Allen
- Department of Medical Genetics, BC Children's and BC's Women's Hospitals, University of British Columbia, Vancouver, BC, Canada
| | - Tanya N Nelson
- Division of Genome Diagnostics, Department of Pathology and Laboratory Medicine, BC Children's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Corneliu Bolbocean
- University of Tennessee Health Science Center, Memphis, TN, United States.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Mary B Connolly
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Matthew J Farrer
- Department of Medical Genetics, Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
31
|
Kunath T, Natalwala A, Chan C, Chen Y, Stecher B, Taylor M, Khan S, Muqit MMK. Are PARKIN patients ideal candidates for dopaminergic cell replacement therapies? Eur J Neurosci 2019; 49:453-462. [PMID: 30586214 PMCID: PMC6492143 DOI: 10.1111/ejn.14314] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 12/12/2022]
Abstract
Parkinson's is a heterogeneous, complex condition. Stratification of Parkinson's subtypes will be essential to identify those that will benefit most from a cell replacement therapy. Foetal mesencephalic grafts can alleviate motor symptoms in some Parkinson's patients. However, on-going synucleinopathy results in the grafts eventually developing Lewy bodies, and they begin to fail. We propose that Parkinson's patients with PARKIN mutations may benefit most from a cell replacement therapy because (a) they often lack synucleinopathy, and (b) their neurodegeneration is often confined to the nigrostriatal pathway. While patients with PARKIN mutations exhibit clinical signs of Parkinson's, post-mortem studies to date indicate the majority lack Lewy bodies suggesting the nigral dopaminergic neurons are lost in a cell autonomous manner independent of α-synuclein mechanisms. Furthermore, these patients are usually younger, slow progressing and typically do not suffer from complex non-nigral symptoms that are unlikely to be ameliorated by a cell replacement therapy. Transplantation of dopaminergic cells into the putamen of these patients will provide neurons with wild-type PARKIN expression to re-innervate the striatum. The focal nature of PARKIN-mediated neurodegeneration and lack of active synucleinopathy in most young-onset cases makes these patients ideal candidates for a dopaminergic cell replacement therapy. Strategies to improve the outcome of cell replacement therapies for sporadic Parkinson's include the use of adjunct therapeutics that target α-synuclein spreading and the use of genetically engineered grafts that are resistant to synucleinopathy.
Collapse
Affiliation(s)
- Tilo Kunath
- MRC Centre for Regenerative MedicineInstitute for Stem Cell ResearchSchool of Biological SciencesThe University of EdinburghEdinburghUK
| | - Ammar Natalwala
- MRC Centre for Regenerative MedicineInstitute for Stem Cell ResearchSchool of Biological SciencesThe University of EdinburghEdinburghUK
- Translational Neurosurgery GroupWestern General HospitalEdinburghUK
| | - Claire Chan
- MRC Centre for Regenerative MedicineInstitute for Stem Cell ResearchSchool of Biological SciencesThe University of EdinburghEdinburghUK
| | - Yixi Chen
- MRC Centre for Regenerative MedicineInstitute for Stem Cell ResearchSchool of Biological SciencesThe University of EdinburghEdinburghUK
| | | | - Martin Taylor
- Edinburgh Research Interest GroupParkinson's UKEdinburghUK
| | - Sadaquate Khan
- Translational Neurosurgery GroupWestern General HospitalEdinburghUK
| | - Miratul M. K. Muqit
- MRC Protein Phosphorylation and Ubiquitylation UnitSchool of Life SciencesUniversity of DundeeDD1 5EHDundeeUK
| |
Collapse
|
32
|
Wang S, Wang Q, Zhang X, Liao X, Wang G, Yu L, Zhang W, Zhou Q, Hu S, Yuan W. Distinct prognostic value of dynactin subunit 4 (DCTN4) and diagnostic value of DCTN1, DCTN2, and DCTN4 in colon adenocarcinoma. Cancer Manag Res 2018; 10:5807-5824. [PMID: 30510450 PMCID: PMC6248376 DOI: 10.2147/cmar.s183062] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Colon adenocarcinoma (COAD) is ranked as the third most commonly diagnosed cancer in both women and men, and it is the most frequently occurring malignant tumor. Dynactin is a protein compound based on multiple subunits, including dynactin 1–6 (DCTN1–6), in most categories of cytoplasmic dynein performance in eukaryotes. Nevertheless, correlations between the DCTN family and the prognosis and diagnosis of COAD remain unidentified. Methods Statistics for DCTN mRNA expression in patients with COAD were acquired from The Cancer Genome Atlas. Kaplan–Meier analyses and a Cox regression model were applied to determine overall survival, with computation of HRs and 95% CIs. Several online data portals were used to assess the biological process, and pathway examination was performed using the Kyoto Encyclopedia of Genes and Genomes to predict the biological functionality of DCTN genes. Results We found that high expression of DCTN4 was linked with satisfactory results for overall survival (P=0.042, HR=0.650, 95% CI 0.429–0.985). The expression of DCTN1, DATN2, and DCTN4 was closely correlated with the frequency of colon tumors (P<0.001, area under the curve [AUC]=0.8811, 95% CI 0.8311–0.9312; P<0.001, AUC=0.870, 96% CI 0.833–0.9071; and P=0.0051, AUC=0.6317, 95% CI 0.5725–0.6908, respectively). In the enrichment examination, the level of gene expression was related to the cell cycle, cell apoptosis, and the cell metastasis pathway. Conclusion The expression levels of DCTN1, DCTN2, and DCTN4 could allow differentiation between cancer-bearing tissues and paracancerous tissue. These genes can be applied as biomarkers to predict the prognosis and diagnosis of COAD.
Collapse
Affiliation(s)
- Shijun Wang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China, ;
| | - Qiaoqi Wang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Xiqian Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Henan Province, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Guixian Wang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China, ;
| | - Long Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Henan Province, China
| | - Wei Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China, ;
| | - Quanbo Zhou
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China, ;
| | - Shengyun Hu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China, ;
| | - Weitang Yuan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China, ;
| |
Collapse
|
33
|
Cobb MM, Ravisankar A, Skibinski G, Finkbeiner S. iPS cells in the study of PD molecular pathogenesis. Cell Tissue Res 2018; 373:61-77. [PMID: 29234887 PMCID: PMC5997490 DOI: 10.1007/s00441-017-2749-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and its pathogenic mechanisms are poorly understood. The majority of PD cases are sporadic but a number of genes are associated with familial PD. Sporadic and familial PD have many molecular and cellular features in common, suggesting some shared pathogenic mechanisms. Induced pluripotent stem cells (iPSCs) have been derived from patients harboring a range of different mutations of PD-associated genes. PD patient-derived iPSCs have been differentiated into relevant cell types, in particular dopaminergic neurons and used as a model to study PD. In this review, we describe how iPSCs have been used to improve our understanding of the pathogenesis of PD. We describe what cellular and molecular phenotypes have been observed in neurons derived from iPSCs harboring known PD-associated mutations and what common pathways may be involved.
Collapse
Affiliation(s)
- Melanie M Cobb
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Abinaya Ravisankar
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Gaia Skibinski
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA
| | - Steven Finkbeiner
- Gladstone Institutes, the Taube/Koret Center for Neurodegenerative Disease, San Francisco, CA, 94158, USA.
- Department of Neurology, University of California, San Francisco, CA, 94143, USA.
- Department Physiology, University of California, San Francisco, CA, 94143, USA.
- Graduate Programs in Neuroscience and Biomedical Sciences, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
34
|
Wang Q, Wang X, Liang Q, Wang S, Liao X, Li D, Pan F. Prognostic Value of Dynactin mRNA Expression in Cutaneous Melanoma. Med Sci Monit 2018; 24:3752-3763. [PMID: 29864111 PMCID: PMC6016438 DOI: 10.12659/msm.910566] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/15/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Dynactin (DCTN) is a multi-subunit protein encoded by DCTN genes for 6 subunits. In different diseases the DCTN genes may have different roles; therefore, we investigated the prognostic potential of DCTN mRNA expression in cutaneous melanoma (CM). MATERIAL AND METHODS Data for DCTN mRNA expression in CM patients were obtained from the OncoLnc database, which contains updated gene expression data for 459 CM patients based on the Cancer Genome Atlas. Kaplan-Meier analysis and a Cox regression model were used to determine overall survival (OS) with calculation of hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS The multivariate survival analysis showed that individually low expression of DCTN1, DCTN2, and DCTN5 and high expression of DCTN6 were associated with favorable OS (adjusted P=0.008, HR=0.676, 95% CI=0.506-0.903; adjusted P=0.004, HR=0.648, 95% CI=0.485-0.867; adjusted P=0.011, HR=0.686, 95% CI=0.514-0.916; and adjusted P=0.018, HR=0.706, 95% CI=0.530-0.942, respectively). In a joint-effects analysis, combinations of low expression of DCTN1, DCTN2, and DCTN5 and high expression of DCTN6 were found to be more highly correlated with favorable OS (all P<0.05). CONCLUSIONS Our findings suggest that downregulated DCTN1, DCTN2, and DCTN5 and upregulated DCTN6 mRNA expression in CM are associated with favorable prognosis and may represent potential prognostic biomarkers. Moreover, use of the 4 genes in combination can improve the sensitivity for predicting OS in CM patients.
Collapse
Affiliation(s)
- Qiaoqi Wang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Qian Liang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Shijun Wang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Dong Li
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| | - Fuqiang Pan
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| |
Collapse
|
35
|
Mishima T, Fujioka S, Tomiyama H, Yabe I, Kurisaki R, Fujii N, Neshige R, Ross OA, Farrer MJ, Dickson DW, Wszolek ZK, Hattori N, Tsuboi Y. Establishing diagnostic criteria for Perry syndrome. J Neurol Neurosurg Psychiatry 2018; 89:482-487. [PMID: 29089398 PMCID: PMC5909757 DOI: 10.1136/jnnp-2017-316864] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/19/2017] [Accepted: 10/18/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To establish international diagnostic criteria for Perry syndrome, a disorder characterised by clinical signs of parkinsonism, depression/apathy, weight loss, respiratory symptoms, mutations in the DCTN1 gene and TAR DNA-binding protein 43 (TDP-43) pathology. METHODS Data from the published literature and newly identified patients were gathered and analysed during and after the International Symposium on Perry syndrome in Tokyo to identify diagnostic criteria for Perry syndrome. RESULTS Eighty-seven patients with Perry syndrome carrying DCTN1 mutations from 20 families were included in this study, and common signs of the disorder were identified, including parkinsonism (95.2% of patients), depression/apathy (71.4%), respiratory symptoms (66.7%) and weight loss (49.2%). CONCLUSIONS Based on our findings, we propose the following definitive diagnostic criteria for Perry syndrome: the presence of four cardinal signs of Perry syndrome, accompanied by a mutation in DCTN1; or a family history of the disease, parkinsonism and a mutation in DCTN1; or the presence of four cardinal signs and pathological findings that include nigral neuronal loss and TDP-43 pathology. As patients with Perry syndrome present with uniform clinical, genetic and pathological features, we further propose the disorder be termed 'Perry disease.'
Collapse
Affiliation(s)
- Takayasu Mishima
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan.,Department of Neuroscience, Mayo Clinic, Jackonsville, Florida, USA
| | - Shinsuke Fujioka
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Hiroyuki Tomiyama
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Neuroscience for Neurodegenerative Disorders, Juntendo University School of Medicine, Tokyo, Japan
| | - Ichiro Yabe
- Department of Neurology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Ryoichi Kurisaki
- Department of Neurology, National Hospital Organization Kumamoto Minami National Hospital, Kumamoto, Japan
| | - Naoki Fujii
- Department of Neurology, National Hospital Organization Omuta Hospital, Fukuoka, Japan
| | | | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jackonsville, Florida, USA.,Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida, USA
| | - Matthew J Farrer
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jackonsville, Florida, USA
| | | | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Neuroscience for Neurodegenerative Disorders, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University School of Medicine, Fukuoka, Japan
| |
Collapse
|
36
|
Verheijen BM, Oyanagi K, van Leeuwen FW. Dysfunction of Protein Quality Control in Parkinsonism-Dementia Complex of Guam. Front Neurol 2018; 9:173. [PMID: 29615966 PMCID: PMC5869191 DOI: 10.3389/fneur.2018.00173] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
Guam parkinsonism–dementia complex (G-PDC) is an enigmatic neurodegenerative disease that is endemic to the Pacific island of Guam. G-PDC patients are clinically characterized by progressive cognitive impairment and parkinsonism. Neuropathologically, G-PDC is characterized by abundant neurofibrillary tangles, which are composed of hyperphosphorylated tau, marked deposition of 43-kDa TAR DNA-binding protein, and neuronal loss. Although both genetic and environmental factors have been implicated, the etiology and pathogenesis of G-PDC remain unknown. Recent neuropathological studies have provided new clues about the pathomechanisms involved in G-PDC. For example, deposition of abnormal components of the protein quality control system in brains of G-PDC patients indicates a role for proteostasis imbalance in the disease. This opens up promising avenues for new research on G-PDC and could have important implications for the study of other neurodegenerative disorders.
Collapse
Affiliation(s)
- Bert M Verheijen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Kiyomitsu Oyanagi
- Division of Neuropathology, Department of Brain Disease Research, Shinshu University School of Medicine, Nagano, Japan.,Brain Research Laboratory, Hatsuishi Hospital, Chiba, Japan
| | - Fred W van Leeuwen
- Department of Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
37
|
Creed RB, Goldberg MS. New Developments in Genetic rat models of Parkinson's Disease. Mov Disord 2018; 33:717-729. [PMID: 29418019 DOI: 10.1002/mds.27296] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/04/2017] [Accepted: 12/10/2017] [Indexed: 12/12/2022] Open
Abstract
Preclinical research on Parkinson's disease has relied heavily on mouse and rat animal models. Initially, PD animal models were generated primarily by chemical neurotoxins that induce acute loss of dopaminergic neurons in the substantia nigra. On the discovery of genetic mutations causally linked to PD, mice were used more than rats to generate laboratory animals bearing PD-linked mutations because mutagenesis was more difficult in rats. Recent advances in technology for mammalian genome engineering and optimization of viral expression vectors have increased the use of genetic rat models of PD. Emerging research tools include "knockout" rats with disruption of genes in which mutations have been causally linked to PD, including LRRK2, α-synuclein, Parkin, PINK1, and DJ-1. Rats have also been increasingly used for transgenic and viral-mediated overexpression of genes relevant to PD, particularly α-synuclein. It may not be realistic to obtain a single animal model that completely reproduces every feature of a human disease as complex as PD. Nevertheless, compared with mice with the same mutations, many genetic rat animal models of PD better reproduce key aspects of PD including progressive loss of dopaminergic neurons in the substantia nigra, locomotor behavior deficits, and age-dependent formation of abnormal α-synuclein protein aggregates. Here we briefly review new developments in genetic rat models of PD that may have greater potential for identifying underlying mechanisms, for discovering novel therapeutic targets, and for developing greatly needed treatments to slow or halt disease progression. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Rose B Creed
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Matthew S Goldberg
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
38
|
Matarazzo M, Wile D, Mackenzie M, Stoessl AJ. PET Molecular Imaging in Familial Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 142:177-223. [DOI: 10.1016/bs.irn.2018.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Mishima T, Koga S, Lin WL, Kasanuki K, Castanedes-Casey M, Wszolek ZK, Oh SJ, Tsuboi Y, Dickson DW. Perry Syndrome: A Distinctive Type of TDP-43 Proteinopathy. J Neuropathol Exp Neurol 2017; 76:676-682. [PMID: 28789478 DOI: 10.1093/jnen/nlx049] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Perry syndrome is a rare atypical parkinsonism with depression, apathy, weight loss, and central hypoventilation caused by mutations in dynactin p150glued (DCTN1). A rare distal hereditary motor neuropathy, HMN7B, also has mutations in DCTN1. Perry syndrome has TAR DNA-binding protein of 43 kDa (TDP-43) inclusions as a defining feature. Other TDP-43 proteinopathies include amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) with and without motor neuron disease (FTLD-MND). TDP-43 forms aggregates in neuronal cytoplasmic inclusions (NCIs), neuronal intranuclear inclusions, dystrophic neurites (DNs), as well as axonal spheroids, oligodendroglial cytoplasmic inclusions, and perivascular astrocytic inclusions (PVIs). We performed semiquantitative assessment of these lesions and presence of dynactin subunit p50 lesions in 3 cases of Perry syndrome and one of HMN7B. We compared them with 3 cases of FTLD-MND, 3 of ALS, and 3 of hippocampal sclerosis (HpScl). Perry syndrome had NCIs, DNs, and frequent PVIs and spheroids. Perry syndrome cases were similar, but different from ALS, FTLD-MND, and HpScl. TDP-43 pathology was not detected in HMN7B. Dynactin p50 inclusions were observed in both Perry syndrome and HMN7B, but not in the other conditions. These results suggest that Perry syndrome may be distinctive type of TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Takayasu Mishima
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Wen-Lang Lin
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Koji Kasanuki
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Monica Castanedes-Casey
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Zbigniew K Wszolek
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Shin J Oh
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Yoshio Tsuboi
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida (TM, SK, W-LL, KK, MC-C, DWD); Department of Neurology, Fukuoka University, Fukuoka, Japan (TM, YT); Department of Neurology, Mayo Clinic, Jacksonville, Florida (ZKW); and Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama (SJO)
| |
Collapse
|
40
|
Liu X, Yang L, Tang L, Chen L, Liu X, Fan D. DCTN1 gene analysis in Chinese patients with sporadic amyotrophic lateral sclerosis. PLoS One 2017; 12:e0182572. [PMID: 28792508 PMCID: PMC5549744 DOI: 10.1371/journal.pone.0182572] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/20/2017] [Indexed: 01/13/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder. Missense mutations of DCTN1 have been identified as a possible genetic risk factor for ALS. Here, we tested the DCTN1 protein-coding exons in 510 sporadic ALS patients in whom SOD1, TARDBP, FUS, and C9orf72 genes were screened before. Polymerase chain reaction and Sanger sequencing were used for mutation discovery. The results revealed two rare heterozygous missense variants, c.1867C>T (p.R623W) and c.2798C>T (p.A933V). These two patients exhibited spinal disease onset without cognitive impairment, and their onset age and diagnosis delay was within the average range of Chinese ALS patients. Our results suggested that variants in DCTN1 are not common risk factors for Chinese sporadic ALS and that the frequency of variants of unknown significance in the cohort study was 0.39%.
Collapse
Affiliation(s)
- Xiangyi Liu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Lipeng Yang
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Lu Tang
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Lu Chen
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Xiaolu Liu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
- * E-mail:
| |
Collapse
|
41
|
Yeo T, Tan LC. 'Hummingbird' Sign in a Patient with Guam Parkinsonism-Dementia Complex. J Mov Disord 2017; 10:145-148. [PMID: 28782342 PMCID: PMC5615172 DOI: 10.14802/jmd.17025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/31/2017] [Accepted: 06/16/2017] [Indexed: 12/12/2022] Open
Abstract
We present a case of a 71-year-old male Chamorro patient from Guam who presented with progressive supranuclear palsy (PSP)-Richardson's syndrome. Considering his strong family history of parkinsonism and a PSP phenotype, he was clinically diagnosed with Guam parkinsonism-dementia complex (PDC). Magnetic resonance imaging (MRI) of the brain revealed prominent midbrain atrophy with preserved pontine volume, forming the 'hummingbird' sign, which has not been described before in Guam PDC. Molecular analysis of the chromosome 9 open reading frame 72 gene (C9orf72) showed only 6 GGGGCC repeats. We discuss the clinico-pathological similarities and differences between PSP and Guam PDC, and highlight the topography of neuropathological changes seen in Guam PDC to explain the appearance of the 'hummingbird' sign on MRI.
Collapse
Affiliation(s)
- Tianrong Yeo
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| | - Louis Cs Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
42
|
Konno T, Ross OA, Teive HAG, Sławek J, Dickson DW, Wszolek ZK. DCTN1-related neurodegeneration: Perry syndrome and beyond. Parkinsonism Relat Disord 2017. [PMID: 28625595 DOI: 10.1016/j.parkreldis.2017.06.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Perry syndrome (PS) is a rare hereditary neurodegenerative disease characterized by autosomal dominant parkinsonism, psychiatric symptoms, weight loss, central hypoventilation, and distinct TDP-43 pathology. The mutated causative gene for PS is DCTN1, which encodes the dynactin subunit p150Glued. Dynactin is a motor protein involved in axonal transport; the p150Glued subunit has a critical role in the overall function. Since the discovery of DCTN1 in PS, it has been increasingly recognized that DCTN1 mutations can exhibit more diverse phenotypes than previously thought. Progressive supranuclear palsy- and/or frontotemporal dementia-like phenotypes have been associated with the PS phenotypes. In addition, DCTN1 mutations were identified in a family with motor-neuron disease before the discovery in PS. In this review, we analyze the clinical and genetic aspects of DCTN1-related neurodegeneration and discuss its pathogenesis. We also describe three families with PS, Canadian, Polish, and Brazilian. DCTN1 mutation was newly identified in two of them, the Canadian and Polish families. The Canadian family was first described in late 1970's but was never genetically tested. We recently had the opportunity to evaluate this family and to test the gene status of an affected family member. The Polish family is newly identified and is the first PS family in Poland. Although still rare, DCTN1-related neurodegeneration needs to be considered in a differential diagnosis of parkinsonian disorders, frontotemporal dementia, and motor-neuron diseases, especially if there is family history.
Collapse
Affiliation(s)
- Takuya Konno
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Hélio A G Teive
- Movement Disorders Unit, Neurology Service, Hospital de Clínicas, Federal University of Paraná, Rua General Carneiro 1103/102, Centro, Curitiba, PR, 80060-150, Brazil
| | - Jarosław Sławek
- Department of Neurological-Psychiatric Nursing, Medical University of Gdansk, Poland; Department of Neurology, St. Adalbert Hospital, Copernicus Ltd., Gdansk, Poland
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Zbigniew K Wszolek
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
43
|
Ubiquitin and Parkinson's disease through the looking glass of genetics. Biochem J 2017; 474:1439-1451. [PMID: 28408429 PMCID: PMC5390927 DOI: 10.1042/bcj20160498] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 12/12/2022]
Abstract
Biochemical alterations found in the brains of Parkinson's disease (PD) patients indicate that cellular stress is a major driver of dopaminergic neuronal loss. Oxidative stress, mitochondrial dysfunction, and ER stress lead to impairment of the homeostatic regulation of protein quality control pathways with a consequent increase in protein misfolding and aggregation and failure of the protein degradation machinery. Ubiquitin signalling plays a central role in protein quality control; however, prior to genetic advances, the detailed mechanisms of how impairment in the ubiquitin system was linked to PD remained mysterious. The discovery of mutations in the α-synuclein gene, which encodes the main protein misfolded in PD aggregates, together with mutations in genes encoding ubiquitin regulatory molecules, including PTEN-induced kinase 1 (PINK1), Parkin, and FBX07, has provided an opportunity to dissect out the molecular basis of ubiquitin signalling disruption in PD, and this knowledge will be critical for developing novel therapeutic strategies in PD that target the ubiquitin system.
Collapse
|
44
|
Lichtenstein ML, Dwosh E, Roy Chowdhury A, Farrer MJ, McKenzie MB, Guella I, Evans DM, Nygaard HB, Shewchuk JR, Hayden S, Barton JJS, Feldman HH. Neurobehavioral characterization of adult-onset Alexander disease: A family study. Neurol Clin Pract 2017; 7:425-429. [PMID: 29620072 DOI: 10.1212/cpj.0000000000000356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 02/13/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Maya L Lichtenstein
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Emily Dwosh
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Anupama Roy Chowdhury
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Matthew J Farrer
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Marna B McKenzie
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Ilaria Guella
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Daniel M Evans
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Haakon B Nygaard
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Jason R Shewchuk
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Sherri Hayden
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Jason J S Barton
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| | - Howard H Feldman
- Division of Neurology, Department of Medicine (MLL, HBN, SH, JJSB, HHF), Department of Medical Genetics, Faculty of Medicine (ED), Centre for Applied Neurogenetics, Department of Medical Genetics (MJF, MBM, IG, DME), Division of Neuroradiology, Department of Radiology (JRS), and Department of Ophthalmology and Visual Sciences (JJSB), University of British Columbia, Vancouver, Canada; Department of Geriatric Medicine (ARC), Khoo Teck Puat Hospital, Singapore; and Department of Neuroscience (HHF), University of California San Diego
| |
Collapse
|
45
|
Kasten M, Marras C, Klein C. Nonmotor Signs in Genetic Forms of Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 133:129-178. [DOI: 10.1016/bs.irn.2017.05.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Truban D, Hou X, Caulfield TR, Fiesel FC, Springer W. PINK1, Parkin, and Mitochondrial Quality Control: What can we Learn about Parkinson's Disease Pathobiology? JOURNAL OF PARKINSON'S DISEASE 2017; 7:13-29. [PMID: 27911343 PMCID: PMC5302033 DOI: 10.3233/jpd-160989] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/10/2016] [Indexed: 12/12/2022]
Abstract
The first clinical description of Parkinson's disease (PD) will embrace its two century anniversary in 2017. For the past 30 years, mitochondrial dysfunction has been hypothesized to play a central role in the pathobiology of this devastating neurodegenerative disease. The identifications of mutations in genes encoding PINK1 (PTEN-induced kinase 1) and Parkin (E3 ubiquitin ligase) in familial PD and their functional association with mitochondrial quality control provided further support to this hypothesis. Recent research focused mainly on their key involvement in the clearance of damaged mitochondria, a process known as mitophagy. It has become evident that there are many other aspects of this complex regulated, multifaceted pathway that provides neuroprotection. As such, numerous additional factors that impact PINK1/Parkin have already been identified including genes involved in other forms of PD. A great pathogenic overlap amongst different forms of familial, environmental and even sporadic disease is emerging that potentially converges at the level of mitochondrial quality control. Tremendous efforts now seek to further detail the roles and exploit PINK1 and Parkin, their upstream regulators and downstream signaling pathways for future translation. This review summarizes the latest findings on PINK1/Parkin-directed mitochondrial quality control, its integration and cross-talk with other disease factors and pathways as well as the implications for idiopathic PD. In addition, we highlight novel avenues for the development of biomarkers and disease-modifying therapies that are based on a detailed understanding of the PINK1/Parkin pathway.
Collapse
Affiliation(s)
- Dominika Truban
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Xu Hou
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Thomas R. Caulfield
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Fabienne C. Fiesel
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Wolfdieter Springer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| |
Collapse
|
47
|
Barodia SK, Creed RB, Goldberg MS. Parkin and PINK1 functions in oxidative stress and neurodegeneration. Brain Res Bull 2016; 133:51-59. [PMID: 28017782 DOI: 10.1016/j.brainresbull.2016.12.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/07/2016] [Accepted: 12/15/2016] [Indexed: 12/12/2022]
Abstract
Loss-of-function mutations in the genes encoding Parkin and PINK1 are causally linked to autosomal recessive Parkinson's disease (PD). Parkin, an E3 ubiquitin ligase, and PINK1, a mitochondrial-targeted kinase, function together in a common pathway to remove dysfunctional mitochondria by autophagy. Presumably, deficiency for Parkin or PINK1 impairs mitochondrial autophagy and thereby increases oxidative stress due to the accumulation of dysfunctional mitochondria that release reactive oxygen species. Parkin and PINK1 likely have additional functions that may be relevant to the mechanisms by which mutations in these genes cause neurodegeneration, such as regulating inflammation, apoptosis, or dendritic morphogenesis. Here we briefly review what is known about functions of Parkin and PINK1 related to oxidative stress and neurodegeneration.
Collapse
Affiliation(s)
- Sandeep K Barodia
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Rose B Creed
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Matthew S Goldberg
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States; Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
48
|
Guella I, Huh L, McKenzie MB, Toyota EB, Bebin EM, Thompson ML, Cooper GM, Evans DM, Buerki SE, Adam S, Van Allen MI, Nelson TN, Connolly MB, Farrer MJ, Demos M. De novo FGF12 mutation in 2 patients with neonatal-onset epilepsy. NEUROLOGY-GENETICS 2016; 2:e120. [PMID: 27872899 PMCID: PMC5113095 DOI: 10.1212/nxg.0000000000000120] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/29/2016] [Indexed: 12/12/2022]
Abstract
Objective: We describe 2 additional patients with early-onset epilepsy with a de novo FGF12 mutation. Methods: Whole-exome sequencing was performed in 2 unrelated patients with early-onset epilepsy and their unaffected parents. Genetic variants were assessed by comparative trio analysis. Clinical evolution, EEG, and neuroimaging are described. The phenotype and response to treatment was reviewed and compared to affected siblings in the original report. Results: We identified the same FGF12 de novo mutation reported previously (c.G155A, p.R52H) in 2 additional patients with early-onset epilepsy. Similar to the original brothers described, both presented with tonic seizures in the first month of life. In the first patient, seizures responded to sodium channel blockers and her development was normal at 11 months. Patient 2 is a 15-year-old girl with treatment-resistant focal epilepsy, moderate intellectual disability, and autism. Carbamazepine (sodium channel blocker) was tried later in her course but not continued due to an allergic reaction. Conclusions: The identification of a recurrent de novo mutation in 2 additional unrelated probands with early-onset epilepsy supports the role of FGF12 p.R52H in disease pathogenesis. Affected carriers presented with similar early clinical phenotypes; however, this report expands the phenotype associated with this mutation which contrasts with the progressive course and early mortality of the siblings in the original report.
Collapse
Affiliation(s)
- Ilaria Guella
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Linda Huh
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Marna B McKenzie
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Eric B Toyota
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - E Martina Bebin
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Michelle L Thompson
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Gregory M Cooper
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Daniel M Evans
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Sarah E Buerki
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Shelin Adam
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Margot I Van Allen
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Tanya N Nelson
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Mary B Connolly
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Matthew J Farrer
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| | - Michelle Demos
- Centre for Applied Neurogenetics (CAN), Department of Medical Genetics (I.G., M.B.M., D.M.E., M.J.F.), Division of Neurology (L.H., E.B.T., S.E.B., M.B.C., M.D.), Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, Canada; Department of Neurology (E.M.B.), University of Alabama at Birmingham; HudsonAlpha Institute for Biotechnology (M.L.T., G.M.C.), Huntsville, AL; Department of Medical Genetics (S.A., M.I.V.A.), University of British Columbia, Vancouver, Canada; and Departments of Pathology and Laboratory Medicine (T.N.N.), University of British Columbia and BC Children's Hospital, Vancouver, Canada
| |
Collapse
|
49
|
Seeking environmental causes of neurodegenerative disease and envisioning primary prevention. Neurotoxicology 2016; 56:269-283. [DOI: 10.1016/j.neuro.2016.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/23/2016] [Indexed: 12/12/2022]
|
50
|
Ishiura H, Tsuji S. Epidemiology and molecular mechanism of frontotemporal lobar degeneration/amyotrophic lateral sclerosis with repeat expansion mutation in C9orf72. J Neurogenet 2016; 29:85-94. [PMID: 26540641 DOI: 10.3109/01677063.2015.1085980] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GGGGCC hexanucleotide repeat expansions in C9orf72 were identified in 2011 as the genetic cause of frontotemporal lobar degeneration (FTLD)/amyotrophic lateral sclerosis (ALS) linked to chromosome 9. Since then, a number of studies have been conducted to delineate the molecular epidemiology of the repeat expansions and the molecular pathophysiology of the disease. The frequency of the repeat expansions considerably varied among countries. The frequency of the repeat expansions was high in European populations and populations of European descent and a substantial proportion of sporadic FTLD or ALS patients also have the mutations in these populations. On the other hand, the frequency was extremely low in Asia or Oceania except for limited regions including Kii Peninsula of Japan. A founder effect seems to strongly influence the regional differences in the frequency, but there is no definitive evidence that supports the notion that the repeat expansions arose in a single founder or multiple founders. As a disease-causing mechanism, several molecular mechanisms have been proposed, including conformational changes of DNA (G-quadruplex formation and hypermethylation) or RNA (G-quadruplex formation) molecules, altered transcriptional levels of C9orf72, sequestration of RNA-binding proteins, bidirectional transcription, formation of RNA foci, and neurotoxicity of dipeptide repeat proteins generated by repeat-associated non-ATG-initiated translation. Further investigations on the molecular mechanisms of neurodegeneration are expected to lead to the development of therapeutic interventions for this disease as well as for other diseases associated with non-coding repeat expansions.
Collapse
Affiliation(s)
- Hiroyuki Ishiura
- a Department of Neurology , The University of Tokyo , Tokyo , Japan
| | - Shoji Tsuji
- a Department of Neurology , The University of Tokyo , Tokyo , Japan
| |
Collapse
|