1
|
Chen J, Li H, Liang R, Huang Y, Tang Q. Aging through the lens of mitochondrial DNA mutations and inheritance paradoxes. Biogerontology 2024; 26:33. [PMID: 39729246 DOI: 10.1007/s10522-024-10175-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Mitochondrial DNA encodes essential components of the respiratory chain complexes, serving as the foundation of mitochondrial respiratory function. Mutations in mtDNA primarily impair energy metabolism, exerting far-reaching effects on cellular physiology, particularly in the context of aging. The intrinsic vulnerability of mtDNA is increasingly recognized as a key driver in the initiation of aging and the progression of its related diseases. In the field of aging research, it is critical to unravel the intricate mechanisms underpinning mtDNA mutations in living organisms and to elucidate the pathological consequences they trigger. Interestingly, certain effects, such as oxidative stress and apoptosis, may not universally accelerate aging as traditionally perceived. These phenomena demand deeper investigation and a more nuanced reinterpretation of current findings to address persistent scientific uncertainties. By synthesizing recent insights, this review seeks to clarify how pathogenic mtDNA mutations drive cellular senescence and systemic health deterioration, while also exploring the complex dynamics of mtDNA inheritance that may propagate these mutations. Such a comprehensive understanding could ultimately inform the development of innovative therapeutic strategies to counteract mitochondrial dysfunctions associated with aging.
Collapse
Affiliation(s)
- Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongyu Li
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yongyin Huang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiang Tang
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
2
|
Abati E, Gagliardi D, Manini A, Del Bo R, Ronchi D, Meneri M, Beretta F, Sarno A, Rizzo F, Monfrini E, Di Fonzo A, Pellecchia MT, Brusati A, Silani V, Comi GP, Ratti A, Verde F, Ticozzi N, Corti S. Investigating the prevalence of MFN2 mutations in amyotrophic lateral sclerosis: insights from an Italian cohort. Brain Commun 2024; 6:fcae312. [PMID: 39315308 PMCID: PMC11417610 DOI: 10.1093/braincomms/fcae312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/11/2024] [Accepted: 09/19/2024] [Indexed: 09/25/2024] Open
Abstract
The MFN2 gene encodes mitofusin 2, a key protein for mitochondrial fusion, transport, maintenance and cell communication. MFN2 mutations are primarily linked to Charcot-Marie-Tooth disease type 2A. However, a few cases of amyotrophic lateral sclerosis and amyotrophic lateral sclerosis/frontotemporal dementia phenotypes with concomitant MFN2 mutations have been previously reported. This study examines the clinical and genetic characteristics of an Italian cohort of amyotrophic lateral sclerosis patients with rare, non-synonymous MFN2 mutations. A group of patients (n = 385) diagnosed with amyotrophic lateral sclerosis at our Neurology Units between 2008 and 2023 underwent comprehensive molecular testing, including MFN2. After excluding pathogenic mutations in the main amyotrophic lateral sclerosis-related genes (i.e. C9orf72, SOD1, FUS and TARDBP), MFN2 variants were classified based on the American College of Medical Genetics and Genomics guidelines, and demographic and clinical data of MFN2-mutated patients were retrieved. We identified 12 rare, heterozygous, non-synonymous MFN2 variants in 19 individuals (4.9%). Eight of these variants, carried by nine patients (2.3%), were either pathogenic, likely pathogenic or variants of unknown significance according to the American College of Medical Genetics and Genomics guidelines. Among these patients, four exhibited a familial pattern of inheritance. The observed phenotypes included classic and bulbar amyotrophic lateral sclerosis, amyotrophic lateral sclerosis/frontotemporal dementia, flail arm, flail leg and progressive muscular atrophy. Median survival after disease onset was extremely variable, ranging from less than 1 to 13 years. This study investigates the prevalence of rare, non-synonymous MFN2 variants within an Italian cohort of amyotrophic lateral sclerosis patients, who have been extensively investigated, enhancing our knowledge of the underlying phenotypic spectrum. Further research is needed to understand whether MFN2 mutations contribute to motor neuron disease and to what extent. Improving our knowledge regarding the genetic basis of amyotrophic lateral sclerosis is crucial both in a diagnostic and therapeutic perspective.
Collapse
Affiliation(s)
- Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Delia Gagliardi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
| | - Arianna Manini
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
| | - Roberto Del Bo
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
| | - Dario Ronchi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
| | - Megi Meneri
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Beretta
- Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), Università degli Studi di Firenze, 50139 Firenze, Italy
| | - Annalisa Sarno
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
| | - Federica Rizzo
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Edoardo Monfrini
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Alessio Di Fonzo
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Maria Teresa Pellecchia
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, Neuroscience Section, Università degli Studi di Salerno, 84081Salerno, Italy
| | - Alberto Brusati
- Department of Brain and Behavioral Sciences, Università degli Studi di Pavia, 27100 Pavia, Italy
| | - Vincenzo Silani
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, 20133 Milan, Italy
| | - Federico Verde
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Nicola Ticozzi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, 20122 Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan 20122 Milan, Italy
| |
Collapse
|
3
|
Lopriore P, Palermo G, Meli A, Bellini G, Benevento E, Montano V, Siciliano G, Mancuso M, Ceravolo R. Mitochondrial Parkinsonism: A Practical Guide to Genes and Clinical Diagnosis. Mov Disord Clin Pract 2024; 11:948-965. [PMID: 38943319 PMCID: PMC11329577 DOI: 10.1002/mdc3.14148] [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: 01/07/2024] [Revised: 04/19/2024] [Accepted: 06/01/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Primary mitochondrial diseases (PMDs) are the most common inborn errors of energy metabolism, with a combined prevalence of 1 in 4300. They can result from mutations in either nuclear DNA (nDNA) or mitochondrial DNA (mtDNA). These disorders are multisystemic and mainly affect high energy-demanding tissues, such as muscle and the central nervous system (CNS). Among many clinical features of CNS involvement, parkinsonism is one of the most common movement disorders in PMDs. METHODS This review provides a pragmatic educational overview of the most recent advances in the field of mitochondrial parkinsonism, from pathophysiology and genetic etiologies to phenotype and diagnosis. RESULTS mtDNA maintenance and mitochondrial dynamics alterations represent the principal mechanisms underlying mitochondrial parkinsonism. It can be present in isolation, alongside other movement disorders or, more commonly, as part of a multisystemic phenotype. Mutations in several nuclear-encoded genes (ie, POLG, TWNK, SPG7, and OPA1) and, more rarely, mtDNA mutations, are responsible for mitochondrial parkinsonism. Progressive external opthalmoplegia and optic atrophy may guide genetic etiology identification. CONCLUSION A comprehensive deep-phenotyping approach is needed to reach a diagnosis of mitochondrial parkinsonism, which lacks distinctive clinical features and exemplifies the intricate genotype-phenotype interplay of PMDs.
Collapse
Affiliation(s)
- Piervito Lopriore
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Unit of Neurology, Department of Clinical and Experimental Medicine, Center for Neurodegenerative Diseases–Parkinson's Disease and Movement DisordersUniversity of PisaPisaItaly
| | - Adriana Meli
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Gabriele Bellini
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Unit of Neurology, Department of Clinical and Experimental Medicine, Center for Neurodegenerative Diseases–Parkinson's Disease and Movement DisordersUniversity of PisaPisaItaly
| | - Elena Benevento
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Unit of Neurology, Department of Clinical and Experimental Medicine, Center for Neurodegenerative Diseases–Parkinson's Disease and Movement DisordersUniversity of PisaPisaItaly
| | - Vincenzo Montano
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Gabriele Siciliano
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Michelangelo Mancuso
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Unit of Neurology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Unit of Neurology, Department of Clinical and Experimental Medicine, Center for Neurodegenerative Diseases–Parkinson's Disease and Movement DisordersUniversity of PisaPisaItaly
| |
Collapse
|
4
|
Wang Y, Hang L, Shou W, Li C, Dong F, Feng X, Jin R, Li B, Xiao S. Case Report: A novel RRM2B variant in a Chinese infant with mitochondrial DNA depletion syndrome and collective analyses of RRM2B variants for disease etiology. Front Pediatr 2024; 12:1363728. [PMID: 38737634 PMCID: PMC11084280 DOI: 10.3389/fped.2024.1363728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/10/2024] [Indexed: 05/14/2024] Open
Abstract
Background There are few reports of infantile mitochondrial DNA depletion syndrome (MDDS) caused by variants in RRM2B and the correlation between genotype and phenotype has rarely been analyzed in detail. This study investigated an infantile patient with MDDS, from clinical characteristics to genetic causes. Methods Routine physical examinations, laboratory assays, which included gas chromatography-mass spectrometry of blood and urine, and MRI scans were performed to obtain an exact diagnosis. Whole-exome sequencing was used to pinpoint the abnormal gene and bioinformatic analyses were performed on the identified variant. Results The case presented with progressive neurologic deterioration, failure to thrive, respiratory distress and lactic acidosis. Sequencing revealed that the patient had a homozygous novel missense variant, c.155T>C (p.Ile52Thr), in exon 2 of the RRM2B gene. Multiple lines of bioinformatic evidence suggested that this was a likely detrimental variant. In addition, reported RRM2B variants were compiled from the relevant literature to analyze disease etiology. We found a distinctive distribution of genotypes across disease manifestations of different severity. Pathogenic alleles of RRM2B were significantly enriched in MDDS cases. Conclusion The novel variant is a likely genetic cause of MDDS. It expands our understanding of the pathogenic variant spectrum and the contribution of the RRM2B gene to the disease spectrum of MDDS.
Collapse
Affiliation(s)
- Yanjun Wang
- Pediatric Intensive Care Unit, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Ling Hang
- Pediatric Intensive Care Unit, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Weihua Shou
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children’s Major Disease Research, Yunnan Medical Center for Pediatric Diseases, Yunnan Institute of Pediatrics, Kunming Children’s Hospital, Kunming, China
| | - Cuifen Li
- Pediatric Intensive Care Unit, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Fangling Dong
- Pediatric Intensive Care Unit, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Xingxing Feng
- Department of Clinical Laboratory, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Ruohong Jin
- Pediatric Intensive Care Unit, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Bin Li
- Pediatric Intensive Care Unit, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Shufang Xiao
- Pediatric Intensive Care Unit, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, China
| |
Collapse
|
5
|
Li H, Zeng F, Huang C, Pu Q, Thomas ER, Chen Y, Li X. The potential role of glucose metabolism, lipid metabolism, and amino acid metabolism in the treatment of Parkinson's disease. CNS Neurosci Ther 2024; 30:e14411. [PMID: 37577934 PMCID: PMC10848100 DOI: 10.1111/cns.14411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023] Open
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is a common neurodegenerative disease, which can cause progressive deterioration of motor function causing muscle stiffness, tremor, and bradykinesia. In this review, we hope to describe approaches that can improve the life of PD patients through modifications of energy metabolism. RECENT FINDINGS The main pathological features of PD are the progressive loss of nigrostriatal dopaminergic neurons and the production of Lewy bodies. Abnormal aggregation of α-synuclein (α-Syn) leading to the formation of Lewy bodies is closely associated with neuronal dysfunction and degeneration. The main causes of PD are said to be mitochondrial damage, oxidative stress, inflammation, and abnormal protein aggregation. Presence of abnormal energy metabolism is another cause of PD. Many studies have found significant differences between neurodegenerative diseases and metabolic decompensation, which has become a biological hallmark of neurodegenerative diseases. SUMMARY In this review, we highlight the relationship between abnormal energy metabolism (Glucose metabolism, lipid metabolism, and amino acid metabolism) and PD. Improvement of key molecules in glucose metabolism, fat metabolism, and amino acid metabolism (e.g., glucose-6-phosphate dehydrogenase, triglycerides, and levodopa) might be potentially beneficial in PD. Some of these metabolic indicators may serve well during the diagnosis of PD. In addition, modulation of these metabolic pathways may be a potential target for the treatment and prevention of PD.
Collapse
Affiliation(s)
- Hangzhen Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Cancan Huang
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Qiqi Pu
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | | | - Yan Chen
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
6
|
Rizzo F, Bono S, Ruepp MD, Salani S, Ottoboni L, Abati E, Melzi V, Cordiglieri C, Pagliarani S, De Gioia R, Anastasia A, Taiana M, Garbellini M, Lodato S, Kunderfranco P, Cazzato D, Cartelli D, Lonati C, Bresolin N, Comi G, Nizzardo M, Corti S. Combined RNA interference and gene replacement therapy targeting MFN2 as proof of principle for the treatment of Charcot-Marie-Tooth type 2A. Cell Mol Life Sci 2023; 80:373. [PMID: 38007410 PMCID: PMC10676309 DOI: 10.1007/s00018-023-05018-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/27/2023]
Abstract
Mitofusin-2 (MFN2) is an outer mitochondrial membrane protein essential for mitochondrial networking in most cells. Autosomal dominant mutations in the MFN2 gene cause Charcot-Marie-Tooth type 2A disease (CMT2A), a severe and disabling sensory-motor neuropathy that impacts the entire nervous system. Here, we propose a novel therapeutic strategy tailored to correcting the root genetic defect of CMT2A. Though mutant and wild-type MFN2 mRNA are inhibited by RNA interference (RNAi), the wild-type protein is restored by overexpressing cDNA encoding functional MFN2 modified to be resistant to RNAi. We tested this strategy in CMT2A patient-specific human induced pluripotent stem cell (iPSC)-differentiated motor neurons (MNs), demonstrating the correct silencing of endogenous MFN2 and replacement with an exogenous copy of the functional wild-type gene. This approach significantly rescues the CMT2A MN phenotype in vitro, stabilizing the altered axonal mitochondrial distribution and correcting abnormal mitophagic processes. The MFN2 molecular correction was also properly confirmed in vivo in the MitoCharc1 CMT2A transgenic mouse model after cerebrospinal fluid (CSF) delivery of the constructs into newborn mice using adeno-associated virus 9 (AAV9). Altogether, our data support the feasibility of a combined RNAi and gene therapy strategy for treating the broad spectrum of human diseases associated with MFN2 mutations.
Collapse
Affiliation(s)
- Federica Rizzo
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Bono
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marc David Ruepp
- United Kingdom Dementia Research Institute Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Sabrina Salani
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linda Ottoboni
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Abati
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Cordiglieri
- Istituto Di Genetica Molecolare "Romeo Ed Enrica Invernizzi", Milan, Italy
| | - Serena Pagliarani
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberta De Gioia
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessia Anastasia
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michela Taiana
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, 20089, Milan, Italy
| | - Paolo Kunderfranco
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, 20089, Milan, Italy
| | - Daniele Cazzato
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy
| | - Nereo Bresolin
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Giacomo Comi
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Monica Nizzardo
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy.
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Milan, Italy.
| |
Collapse
|
7
|
Du J, Zhang C, Liu F, Liu X, Wang D, Zhao D, Shui G, Zhao Y, Yan C. Distinctive metabolic remodeling in TYMP deficiency beyond mitochondrial dysfunction. J Mol Med (Berl) 2023; 101:1237-1253. [PMID: 37603049 DOI: 10.1007/s00109-023-02358-9] [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: 01/02/2023] [Revised: 07/09/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is caused by mutations in the TYMP gene, which encodes thymidine phosphorylase (TP). As a cytosolic metabolic enzyme, TP defects affect biological processes that are thought to not be limited to the abnormal replication of mitochondrial DNA. This study aimed to elucidate the characteristic metabolic alterations and associated homeostatic regulation caused by TYMP deficiency. The pathogenicity of novel TYMP variants was evaluated in terms of clinical features, genetic analysis, and structural instability. We analyzed plasma samples from three patients with MNGIE; three patients with m.3243A > G mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS); and four healthy controls (HC) using both targeted and untargeted metabolomics techniques. Transcriptomics analysis and bioenergetic studies were performed on skin fibroblasts from participants in these three groups. A TYMP overexpression experiment was conducted to rescue the observed changes. Compared with controls, specific alterations in nucleosides, bile acids, and steroid metabolites were identified in the plasma of MNGIE patients. Comparable mitochondrial dysfunction was present in fibroblasts from patients with TYMP deficiency and in those from patients with the m.3243A > G mutation. Distinctively decreased sterol regulatory element binding protein (SREBP) regulated cholesterol metabolism and fatty acid (FA) biosynthesis as well as reduced FA degradation were revealed in fibroblasts with TYMP deficiency. The restoration of thymidine phosphorylase activity rescued the observed changes in MNGIE fibroblasts. Our findings indicated that more widespread metabolic disturbance may be caused by TYMP deficiency in addition to mitochondrial dysfunction, which expands our knowledge of the biochemical outcome of TYMP deficiency. KEY MESSAGES: Distinct metabolic profiles in patients with TYMP deficiency compared to those with m.3243A > G mutation. TYMP deficiency leads to a global disruption of nucleoside metabolism. Cholesterol and fatty acid metabolism are inhibited in individuals with MNGIE. TYMP is functionally related to SREBP-regulated pathways. Potential metabolite biomarkers that could be valuable clinical tools to improve the diagnosis of MNGIE.
Collapse
Affiliation(s)
- Jixiang Du
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chao Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fuchen Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xihan Liu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dongdong Wang
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dandan Zhao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 101408, China
| | - Yuying Zhao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Qingdao Key Lab of Mitochondrial Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, 266103, China.
| |
Collapse
|
8
|
Hsu YL, Chen HJ, Gao JX, Yang MY, Fu RH. Chiisanoside Mediates the Parkin/ZNF746/PGC-1α Axis by Downregulating MiR-181a to Improve Mitochondrial Biogenesis in 6-OHDA-Caused Neurotoxicity Models In Vitro and In Vivo: Suggestions for Prevention of Parkinson's Disease. Antioxidants (Basel) 2023; 12:1782. [PMID: 37760085 PMCID: PMC10525196 DOI: 10.3390/antiox12091782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The degeneration of dopamine (DA) neurons is known to be associated with defects in mitochondrial biogenesis caused by aging, environmental factors, or mutations in genes, leading to Parkinson's disease (PD). As PD has not yet been successfully cured, the strategy of using small molecule drugs to protect and restore mitochondrial biogenesis is a promising direction. This study evaluated the efficacy of synthetic chiisanoside (CSS) identified in the leaves of Acanthopanax sessiliflorus to prevent PD symptoms. The results show that in the 6-hydroxydopamine (6-OHDA) model, CSS pretreatment can effectively alleviate the reactive oxygen species generation and apoptosis of SH-SY5Y cells, thereby lessening the defects in the C. elegans model including DA neuron degeneration, dopamine-mediated food sensitivity behavioral disorders, and shortened lifespan. Mechanistically, we found that CSS could restore the expression of proliferator-activated receptor gamma coactivator-1-alpha (PGC-1α), a key molecule in mitochondrial biogenesis, and its downstream related genes inhibited by 6-OHDA. We further confirmed that this is due to the enhanced activity of parkin leading to the ubiquitination and degradation of PGC-1α inhibitor protein Zinc finger protein 746 (ZNF746). Parkin siRNA treatment abolished this effect of CSS. Furthermore, we found that CSS inhibited 6-OHDA-induced expression of miR-181a, which targets parkin. The CSS's ability to reverse the 6-OHDA-induced reduction in mitochondrial biogenesis and activation of apoptosis was abolished after the transfection of anti-miR-181a and miR-181a mimics. Therefore, the neuroprotective effect of CSS mainly promotes mitochondrial biogenesis by regulating the miR-181a/Parkin/ZNF746/PGC-1α axis. CSS potentially has the opportunity to be developed into PD prevention agents.
Collapse
Affiliation(s)
- Yu-Ling Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Hui-Jye Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
| | - Jia-Xin Gao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
| | - Ming-Yang Yang
- Ph.D. Program for Aging, China Medical University, Taichung 40402, Taiwan;
| | - Ru-Huei Fu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; (Y.-L.H.); (H.-J.C.); (J.-X.G.)
- Ph.D. Program for Aging, China Medical University, Taichung 40402, Taiwan;
- Translational Medicine Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| |
Collapse
|
9
|
Scott KS, Chelette B, Chidomere C, Phillip West A, Dantzer R. Cisplatin decreases voluntary wheel-running activity but does not impair food-motivated behavior in mice. Brain Behav Immun 2023; 111:169-176. [PMID: 37076053 PMCID: PMC10330347 DOI: 10.1016/j.bbi.2023.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023] Open
Abstract
Cisplatin is a chemotherapeutic agent that is still commonly used to treat solid tumors. However, it has several toxic side effects due in large part to the mitochondrial damage that it induces. As this mitochondrial damage is likely to result in a decrease in the amount of metabolic energy that is available for behavioral activities, it is not surprising that fatigue develops in cancer patients treated with cisplatin. The present preclinical study was initiated to determine whether the detrimental effects of cisplatin were more pronounced on physical effort requiring a lot of energy versus effort that not only requires less energy but also procures energy in the form of food. For this purpose, mice were either trained to run in a wheel or to work for food in various schedules of food reinforcement before being treated with cisplatin. The experiments were carried out only in male mice as we had already reported that sex differences in cisplatin-induced neurotoxicities are minimal. Cisplatin was administered daily for one cycle of five days, or two cycles separated by a five-day rest. As observed in previous experiments, cisplatin drastically reduced voluntary wheel running. In contrast, when cisplatin was administered to food-restricted mice trained to work for a food reward in a progressive ratio schedule or in a fixed-interval schedule, it tended to increase the number of responses emitted to obtain the food rewards. This increase was not associated with any change in the temporal distribution of responses during the interval between two reinforcements in mice submitted to the fixed interval schedule of food reinforcement. When cisplatin was administered to food-restricted mice trained in an effort-based decision-making task in which they had to choose between working for a grain pellet with little effort and working for a preferred chocolate pellet with more effort, it decreased the total number of responses emitted to obtain food rewards. However, this effect was much less marked than the decrease in wheel running induced by cisplatin. The decrease in the effort invested in the procurement of food rewards was not associated with any change in the relative distribution of effort between low reward and high reward during the time course of the test session. These findings show that cisplatin decreases energy-consuming activities but not energy-procuring activities unless they require a choice between options differing in their cost-benefit ratio. Furthermore, they indicate that the physical dimension of fatigue is more likely to develop in cisplatin-treated individuals than the motivational dimension of fatigue.
Collapse
Affiliation(s)
- Kiersten S Scott
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Brandon Chelette
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Chinenye Chidomere
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Robert Dantzer
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA.
| |
Collapse
|
10
|
Gupta R, Kumari S, Tripathi R, Ambasta RK, Kumar P. Unwinding the modalities of necrosome activation and necroptosis machinery in neurological diseases. Ageing Res Rev 2023; 86:101855. [PMID: 36681250 DOI: 10.1016/j.arr.2023.101855] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/09/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
Necroptosis, a regulated form of cell death, is involved in the genesis and development of various life-threatening diseases, including cancer, neurological disorders, cardiac myopathy, and diabetes. Necroptosis initiates with the formation and activation of a necrosome complex, which consists of RIPK1, RIPK2, RIPK3, and MLKL. Emerging studies has demonstrated the regulation of the necroptosis cell death pathway through the implication of numerous post-translational modifications, namely ubiquitination, acetylation, methylation, SUMOylation, hydroxylation, and others. In addition, the negative regulation of the necroptosis pathway has been shown to interfere with brain homeostasis through the regulation of axonal degeneration, mitochondrial dynamics, lysosomal defects, and inflammatory response. Necroptosis is controlled by the activity and expression of signaling molecules, namely VEGF/VEGFR, PI3K/Akt/GSK-3β, c-Jun N-terminal kinases (JNK), ERK/MAPK, and Wnt/β-catenin. Herein, we briefly discussed the implication and potential of necrosome activation in the pathogenesis and progression of neurological manifestations, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, traumatic brain injury, and others. Further, we present a detailed picture of natural compounds, micro-RNAs, and chemical compounds as therapeutic agents for treating neurological manifestations.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India.
| |
Collapse
|
11
|
Jiménez-Salvador I, Meade P, Iglesias E, Bayona-Bafaluy P, Ruiz-Pesini E. Developmental origins of Parkinson disease: Improving the rodent models. Ageing Res Rev 2023; 86:101880. [PMID: 36773760 DOI: 10.1016/j.arr.2023.101880] [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: 10/20/2022] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Numerous pesticides are inhibitors of the oxidative phosphorylation system. Oxidative phosphorylation dysfunction adversely affects neurogenesis and often accompanies Parkinson disease. Since brain development occurs mainly in the prenatal period, early exposure to pesticides could alter the development of the nervous system and increase the risk of Parkinson disease. Different rodent models have been used to confirm this hypothesis. However, more precise considerations of the selected strain, the xenobiotic, its mode of administration, and the timing of animal analysis, are necessary to resemble the model to the human clinical condition and obtain more reliable results.
Collapse
Affiliation(s)
- Irene Jiménez-Salvador
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain.
| | - Patricia Meade
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza, 50018 Zaragoza, Spain.
| | - Eldris Iglesias
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Facultad de Ciencias de la Salud, Universidad San Jorge, 50830 Villanueva de Gállego, Zaragoza, Spain.
| | - Pilar Bayona-Bafaluy
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza, 50018 Zaragoza, Spain.
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| |
Collapse
|
12
|
Park J, Baruch-Torres N, Yin YW. Structural and Molecular Basis for Mitochondrial DNA Replication and Transcription in Health and Antiviral Drug Toxicity. Molecules 2023; 28:1796. [PMID: 36838782 PMCID: PMC9961925 DOI: 10.3390/molecules28041796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Human mitochondrial DNA (mtDNA) is a 16.9 kbp double-stranded, circular DNA, encoding subunits of the oxidative phosphorylation electron transfer chain and essential RNAs for mitochondrial protein translation. The minimal human mtDNA replisome is composed of the DNA helicase Twinkle, DNA polymerase γ, and mitochondrial single-stranded DNA-binding protein. While the mitochondrial RNA transcription is carried out by mitochondrial RNA polymerase, mitochondrial transcription factors TFAM and TFB2M, and a transcription elongation factor, TEFM, both RNA transcriptions, and DNA replication machineries are intertwined and control mtDNA copy numbers, cellular energy supplies, and cellular metabolism. In this review, we discuss the mechanisms governing these main pathways and the mtDNA diseases that arise from mutations in transcription and replication machineries from a structural point of view. We also address the adverse effect of antiviral drugs mediated by mitochondrial DNA and RNA polymerases as well as possible structural approaches to develop nucleoside reverse transcriptase inhibitor and ribonucleosides analogs with reduced toxicity.
Collapse
Affiliation(s)
- Joon Park
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Pharmacology and Toxicology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Noe Baruch-Torres
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Pharmacology and Toxicology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Y. Whitney Yin
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Pharmacology and Toxicology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
13
|
Feng J, Chen Z, Liang W, Wei Z, Ding G. Roles of Mitochondrial DNA Damage in Kidney Diseases: A New Biomarker. Int J Mol Sci 2022; 23:ijms232315166. [PMID: 36499488 PMCID: PMC9735745 DOI: 10.3390/ijms232315166] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/27/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
The kidney is a mitochondria-rich organ, and kidney diseases are recognized as mitochondria-related pathologies. Intact mitochondrial DNA (mtDNA) maintains normal mitochondrial function. Mitochondrial dysfunction caused by mtDNA damage, including impaired mtDNA replication, mtDNA mutation, mtDNA leakage, and mtDNA methylation, is involved in the progression of kidney diseases. Herein, we review the roles of mtDNA damage in different setting of kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD). In a variety of kidney diseases, mtDNA damage is closely associated with loss of kidney function. The level of mtDNA in peripheral serum and urine also reflects the status of kidney injury. Alleviating mtDNA damage can promote the recovery of mitochondrial function by exogenous drug treatment and thus reduce kidney injury. In short, we conclude that mtDNA damage may serve as a novel biomarker for assessing kidney injury in different causes of renal dysfunction, which provides a new theoretical basis for mtDNA-targeted intervention as a therapeutic option for kidney diseases.
Collapse
Affiliation(s)
- Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan 430060, China
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan 430060, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan 430060, China
| | - Zhongping Wei
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan 430060, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Nephrology and Urology Research Institute of Wuhan University, Wuhan 430060, China
- Correspondence:
| |
Collapse
|
14
|
Xing J, Qi L, Liu X, Shi G, Sun X, Yang Y. Roles of mitochondrial fusion and fission in breast cancer progression: a systematic review. World J Surg Oncol 2022; 20:331. [PMID: 36192752 PMCID: PMC9528125 DOI: 10.1186/s12957-022-02799-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/24/2022] [Indexed: 12/02/2022] Open
Abstract
Background Mitochondria play critical roles in cellular physiological activity as cellular organelles. Under extracellular stimulation, mitochondria undergo constant fusion and fission to meet different cellular demands. Mitochondrial dynamics, which are involved in mitochondrial fusion and fission, are regulated by specialized proteins and lipids, and their dysregulation causes human diseases, such as cancer. The advanced literature about the crucial role of mitochondrial dynamics in breast cancer is performed. Methods All related studies were systematically searched through online databases (PubMed, Web of Science, and EMBASE) using keywords (e.g., breast cancer, mitochondrial, fission, and fusion), and these studies were then screened through the preset inclusion and exclusion criteria. Results Eligible studies (n = 19) were evaluated and discussed in the systematic review. These advanced studies established the roles of mitochondrial fission and fusion of breast cancer in the metabolism, proliferation, survival, and metastasis. Importantly, the manipulating of mitochondrial dynamic is significant for the progresses of breast cancer. Conclusion Understanding the mechanisms underlying mitochondrial fission and fusion during tumorigenesis is important for improving breast cancer treatments.
Collapse
Affiliation(s)
- Jixiang Xing
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Luyao Qi
- The Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Xiaofei Liu
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Guangxi Shi
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaohui Sun
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yi Yang
- Department of Breast and Thyroid, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
15
|
A novel RRM2B mutation associated with mitochondrial DNA depletion syndrome. Mol Genet Metab Rep 2022; 32:100887. [PMID: 35756861 PMCID: PMC9218228 DOI: 10.1016/j.ymgmr.2022.100887] [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: 04/21/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial DNA (mtDNA) depletion syndromes are disorders characterized by infantile-onset, severe progression, and the drastic loss of mtDNA content in affected tissues. In a patient who showed severe hypotonia, proximal tubulopathy and sensorineural hearing loss after birth, we observed severe mtDNA depletion and impaired respiratory chain activity in muscle due to heterozygous variants c.686G > T and c.551-2A > G in RRM2B, encoding the p53R2 subunit of the ribonucleotide reductase.
Collapse
|
16
|
NADPH and Mitochondrial Quality Control as Targets for a Circadian-Based Fasting and Exercise Therapy for the Treatment of Parkinson's Disease. Cells 2022; 11:cells11152416. [PMID: 35954260 PMCID: PMC9367803 DOI: 10.3390/cells11152416] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 02/01/2023] Open
Abstract
Dysfunctional mitochondrial quality control (MQC) is implicated in the pathogenesis of Parkinson's disease (PD). The improper selection of mitochondria for mitophagy increases reactive oxygen species (ROS) levels and lowers ATP levels. The downstream effects include oxidative damage, failure to maintain proteostasis and ion gradients, and decreased NAD+ and NADPH levels, resulting in insufficient energy metabolism and neurotransmitter synthesis. A ketosis-based metabolic therapy that increases the levels of (R)-3-hydroxybutyrate (BHB) may reverse the dysfunctional MQC by partially replacing glucose as an energy source, by stimulating mitophagy, and by decreasing inflammation. Fasting can potentially raise cytoplasmic NADPH levels by increasing the mitochondrial export and cytoplasmic metabolism of ketone body-derived citrate that increases flux through isocitrate dehydrogenase 1 (IDH1). NADPH is an essential cofactor for nitric oxide synthase, and the nitric oxide synthesized can diffuse into the mitochondrial matrix and react with electron transport chain-synthesized superoxide to form peroxynitrite. Excessive superoxide and peroxynitrite production can cause the opening of the mitochondrial permeability transition pore (mPTP) to depolarize the mitochondria and activate PINK1-dependent mitophagy. Both fasting and exercise increase ketogenesis and increase the cellular NAD+/NADH ratio, both of which are beneficial for neuronal metabolism. In addition, both fasting and exercise engage the adaptive cellular stress response signaling pathways that protect neurons against the oxidative and proteotoxic stress implicated in PD. Here, we discuss how intermittent fasting from the evening meal through to the next-day lunch together with morning exercise, when circadian NAD+/NADH is most oxidized, circadian NADP+/NADPH is most reduced, and circadian mitophagy gene expression is high, may slow the progression of PD.
Collapse
|
17
|
Percetti M, Franco G, Monfrini E, Caporali L, Minardi R, La Morgia C, Valentino ML, Liguori R, Palmieri I, Ottaviani D, Vizziello M, Ronchi D, Di Berardino F, Cocco A, Macao B, Falkenberg M, Comi GP, Albanese A, Giometto B, Valente EM, Carelli V, Di Fonzo A.
TWNK
in Parkinson's Disease: A Movement Disorder and Mitochondrial Disease Center Perspective Study. Mov Disord 2022; 37:1938-1943. [PMID: 35792653 PMCID: PMC9544864 DOI: 10.1002/mds.29139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/17/2022] [Accepted: 05/30/2022] [Indexed: 11/06/2022] Open
Abstract
Background Objectives Methods Results Conclusions
Collapse
Affiliation(s)
- Marco Percetti
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Neurology Unit, San Gerardo Hospital ASST Monza, Monza Italy
| | - Giulia Franco
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Neurology Unit Milan Italy
| | - Edoardo Monfrini
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Neurology Unit Milan Italy
| | | | | | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna Bologna Italy
| | - Maria Lucia Valentino
- IRCCS Istituto delle Scienze Neurologiche di Bologna Bologna Italy
- Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM) University of Bologna Bologna Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna Bologna Italy
- Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM) University of Bologna Bologna Italy
| | - Ilaria Palmieri
- Neurogenetics Research Center IRCCS Mondino Foundation Pavia Italy
| | - Donatella Ottaviani
- Neurology Unit Rovereto Hospital, Azienda Provinciale per i Servizi Sanitari (APSS) di Trento Trento Italy
| | - Maria Vizziello
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Neurology Unit Milan Italy
| | - Dario Ronchi
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation University of Milan Milan Italy
| | - Federica Di Berardino
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Audiology Unit Milan Italy
| | - Antoniangela Cocco
- University of Milan Milan Italy
- Department of Neurology Istituto di Ricovero e Cura a Carattere Scientifico Humanitas, Research Hospital Milan Italy
| | - Bertil Macao
- Department of Medical Biochemistry and Cell Biology University of Gothenburg Gothenburg Sweden
| | - Maria Falkenberg
- Department of Medical Biochemistry and Cell Biology University of Gothenburg Gothenburg Sweden
| | - Giacomo Pietro Comi
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Neurology Unit Milan Italy
| | - Alberto Albanese
- Department of Neurology Istituto di Ricovero e Cura a Carattere Scientifico Humanitas, Research Hospital Milan Italy
| | - Bruno Giometto
- Neurology Unit Rovereto Hospital, Azienda Provinciale per i Servizi Sanitari (APSS) di Trento Trento Italy
| | - Enza Maria Valente
- Neurogenetics Research Center IRCCS Mondino Foundation Pavia Italy
- Department of Molecular Medicine University of Pavia Pavia Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna Bologna Italy
- Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM) University of Bologna Bologna Italy
| | - Alessio Di Fonzo
- Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation University of Milan Milan Italy
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico Neurology Unit Milan Italy
| |
Collapse
|
18
|
Manini A, Caporali L, Meneri M, Zanotti S, Piga D, Arena IG, Corti S, Toscano A, Comi GP, Musumeci O, Carelli V, Ronchi D. Case Report: Rare Homozygous RNASEH1 Mutations Associated With Adult-Onset Mitochondrial Encephalomyopathy and Multiple Mitochondrial DNA Deletions. Front Genet 2022; 13:906667. [PMID: 35711919 PMCID: PMC9194440 DOI: 10.3389/fgene.2022.906667] [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] [Received: 03/28/2022] [Accepted: 04/25/2022] [Indexed: 12/02/2022] Open
Abstract
Mitochondrial DNA (mtDNA) maintenance disorders embrace a broad range of clinical syndromes distinguished by the evidence of mtDNA depletion and/or deletions in affected tissues. Among the nuclear genes associated with mtDNA maintenance disorders, RNASEH1 mutations produce a homogeneous phenotype, with progressive external ophthalmoplegia (PEO), ptosis, limb weakness, cerebellar ataxia, and dysphagia. The encoded enzyme, ribonuclease H1, is involved in mtDNA replication, whose impairment leads to an increase in replication intermediates resulting from mtDNA replication slowdown. Here, we describe two unrelated Italian probands (Patient 1 and Patient 2) affected by chronic PEO, ptosis, and muscle weakness. Cerebellar features and severe dysphagia requiring enteral feeding were observed in one patient. In both cases, muscle biopsy revealed diffuse mitochondrial abnormalities and multiple mtDNA deletions. A targeted next-generation sequencing analysis revealed the homozygous RNASEH1 mutations c.129-3C>G and c.424G>A in patients 1 and 2, respectively. The c.129-3C>G substitution has never been described as disease-related and resulted in the loss of exon 2 in Patient 1 muscle RNASEH1 transcript. Overall, we recommend implementing the use of high-throughput sequencing approaches in the clinical setting to reach genetic diagnosis in case of suspected presentations with impaired mtDNA homeostasis.
Collapse
Affiliation(s)
- Arianna Manini
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Leonardo Caporali
- Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| | - Megi Meneri
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Department of Neuroscience, Milan, Italy
| | - Simona Zanotti
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
| | - Daniela Piga
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Department of Neuroscience, Milan, Italy
| | - Ignazio Giuseppe Arena
- Unit of Neurology and Neuromuscular disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Stefania Corti
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Department of Neuroscience, Milan, Italy
| | - Antonio Toscano
- Unit of Neurology and Neuromuscular disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giacomo Pietro Comi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
| | - Olimpia Musumeci
- Unit of Neurology and Neuromuscular disorders, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Valerio Carelli
- Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy.,Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy
| | - Dario Ronchi
- Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Department of Neuroscience, Milan, Italy
| |
Collapse
|