1
|
Smith KK, Moreira JD, Wilson CR, Padera JO, Lamason AN, Xue L, Gopal DM, Flynn DB, Fetterman JL. A systematic review on the biochemical threshold of mitochondrial genetic variants. Genome Res 2024; 34:341-365. [PMID: 38627095 PMCID: PMC11067886 DOI: 10.1101/gr.278200.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Mitochondrial DNA (mtDNA) variants cause a range of diseases from severe pediatric syndromes to aging-related conditions. The percentage of mtDNA copies carrying a pathogenic variant, variant allele frequency (VAF), must reach a threshold before a biochemical defect occurs, termed the biochemical threshold. Whether the often-cited biochemical threshold of >60% VAF is similar across mtDNA variants and cell types is unclear. In our systematic review, we sought to identify the biochemical threshold of mtDNA variants in relation to VAF by human tissue/cell type. We used controlled vocabulary terms to identify articles measuring oxidative phosphorylation (OXPHOS) complex activities in relation to VAF. We identified 76 eligible publications, describing 69, 12, 16, and 49 cases for complexes I, III, IV, and V, respectively. Few studies evaluated OXPHOS activities in diverse tissue types, likely reflective of clinical access. A number of cases with similar VAFs for the same pathogenic variant had varying degrees of residual activity of the affected complex, alluding to the presence of modifying variants. Tissues and cells with VAFs <60% associated with low complex activities were described, suggesting the possibility of a biochemical threshold of <60%. Using Kendall rank correlation tests, the VAF of the m.8993T > G variant correlated with complex V activity in skeletal muscle (τ = -0.58, P = 0.01, n = 13); however, no correlation was observed in fibroblasts (P = 0.7, n = 9). Our systematic review highlights the need to investigate the biochemical threshold over a wider range of VAFs in disease-relevant cell types to better define the biochemical threshold for specific mtDNA variants.
Collapse
Affiliation(s)
- Karan K Smith
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Jesse D Moreira
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
- Programs in Human Physiology, Department of Health Sciences, Boston University Sargent College, Boston, Massachusetts 02215, USA
| | - Callum R Wilson
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - June O Padera
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Ashlee N Lamason
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Liying Xue
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Deepa M Gopal
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - David B Flynn
- Medical Sciences and Education, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Jessica L Fetterman
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA;
| |
Collapse
|
2
|
Talukdar PD, Chatterji U. Transcriptional co-activators: emerging roles in signaling pathways and potential therapeutic targets for diseases. Signal Transduct Target Ther 2023; 8:427. [PMID: 37953273 PMCID: PMC10641101 DOI: 10.1038/s41392-023-01651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/27/2023] [Accepted: 09/10/2023] [Indexed: 11/14/2023] Open
Abstract
Specific cell states in metazoans are established by the symphony of gene expression programs that necessitate intricate synergic interactions between transcription factors and the co-activators. Deregulation of these regulatory molecules is associated with cell state transitions, which in turn is accountable for diverse maladies, including developmental disorders, metabolic disorders, and most significantly, cancer. A decade back most transcription factors, the key enablers of disease development, were historically viewed as 'undruggable'; however, in the intervening years, a wealth of literature validated that they can be targeted indirectly through transcriptional co-activators, their confederates in various physiological and molecular processes. These co-activators, along with transcription factors, have the ability to initiate and modulate transcription of diverse genes necessary for normal physiological functions, whereby, deregulation of such interactions may foster tissue-specific disease phenotype. Hence, it is essential to analyze how these co-activators modulate specific multilateral processes in coordination with other factors. The proposed review attempts to elaborate an in-depth account of the transcription co-activators, their involvement in transcription regulation, and context-specific contributions to pathophysiological conditions. This review also addresses an issue that has not been dealt with in a comprehensive manner and hopes to direct attention towards future research that will encompass patient-friendly therapeutic strategies, where drugs targeting co-activators will have enhanced benefits and reduced side effects. Additional insights into currently available therapeutic interventions and the associated constraints will eventually reveal multitudes of advanced therapeutic targets aiming for disease amelioration and good patient prognosis.
Collapse
Affiliation(s)
- Priyanka Dey Talukdar
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Urmi Chatterji
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, West Bengal, India.
| |
Collapse
|
3
|
Sollazzo M, De Luise M, Lemma S, Bressi L, Iorio M, Miglietta S, Milioni S, Kurelac I, Iommarini L, Gasparre G, Porcelli AM. Respiratory Complex I dysfunction in cancer: from a maze of cellular adaptive responses to potential therapeutic strategies. FEBS J 2022; 289:8003-8019. [PMID: 34606156 PMCID: PMC10078660 DOI: 10.1111/febs.16218] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 01/14/2023]
Abstract
Mitochondria act as key organelles in cellular bioenergetics and biosynthetic processes producing signals that regulate different molecular networks for proliferation and cell death. This ability is also preserved in pathologic contexts such as tumorigenesis, during which bioenergetic changes and metabolic reprogramming confer flexibility favoring cancer cell survival in a hostile microenvironment. Although different studies epitomize mitochondrial dysfunction as a protumorigenic hit, genetic ablation or pharmacological inhibition of respiratory complex I causing a severe impairment is associated with a low-proliferative phenotype. In this scenario, it must be considered that despite the initial delay in growth, cancer cells may become able to resume proliferation exploiting molecular mechanisms to overcome growth arrest. Here, we highlight the current knowledge on molecular responses activated by complex I-defective cancer cells to bypass physiological control systems and to re-adapt their fitness during microenvironment changes. Such adaptive mechanisms could reveal possible novel molecular players in synthetic lethality with complex I impairment, thus providing new synergistic strategies for mitochondrial-based anticancer therapy.
Collapse
Affiliation(s)
- Manuela Sollazzo
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Monica De Luise
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Silvia Lemma
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Licia Bressi
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Iorio
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Stefano Miglietta
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Sara Milioni
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Ivana Kurelac
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Luisa Iommarini
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Giuseppe Gasparre
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Interdepartmental Center for Industrial Research (CIRI) Life Sciences and Technologies for Health, Alma Mater Studiorum-University of Bologna, Ozzano dell'Emilia, Italy
| |
Collapse
|
4
|
Mengozzi A, Costantino S, Paneni F, Duranti E, Nannipieri M, Mancini R, Lai M, La Rocca V, Puxeddu I, Antonioli L, Fornai M, Ghionzoli M, Georgiopoulos G, Ippolito C, Bernardini N, Ruschitzka F, Pugliese NR, Taddei S, Virdis* A, Masi S. Targeting SIRT1 Rescues Age- and Obesity-Induced Microvascular Dysfunction in Ex Vivo Human Vessels. Circ Res 2022; 131:476-491. [PMID: 35968712 PMCID: PMC9426744 DOI: 10.1161/circresaha.122.320888] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Experimental evidence suggests a key role of SIRT1 (silent information regulator 1) in age- and metabolic-related vascular dysfunction. Whether these effects hold true in the human microvasculature is unknown. We aimed to investigate the SIRT1 role in very early stages of age- and obesity-related microvascular dysfunction in humans. METHODS Ninety-five subjects undergoing elective laparoscopic surgery were recruited and stratified based on their body mass index status (above or below 30 kg/m2) and age (above or below 40 years) in 4 groups: Young Nonobese, Young Obese, Old Nonobese, and Old Obese. We measured small resistance arteries' endothelial function by pressurized micromyography before and after incubation with a SIRT1 agonist (SRT1720) and a mitochondria reactive oxygen species (mtROS) scavenger (MitoTEMPO). We assessed vascular levels of mtROS and nitric oxide availability by confocal microscopy and vascular gene expression of SIRT1 and mitochondrial proteins by qPCR. Chromatin immunoprecipitation assay was employed to investigate SIRT1-dependent epigenetic regulation of mitochondrial proteins. RESULTS Compared with Young Nonobese, obese and older patients showed lower vascular expression of SIRT1 and antioxidant proteins (FOXO3 [forkhead box protein O3] and SOD2) and higher expression of pro-oxidant and aging mitochondria proteins p66Shc and Arginase II. Old Obese, Young Obese and Old Nonobese groups endothelial dysfunction was rescued by SRT1720. The restoration was comparable to the one obtained with mitoTEMPO. These effects were explained by SIRT1-dependent chromatin changes leading to reduced p66Shc expression and upregulation of proteins involved in mitochondria respiratory chain. CONCLUSIONS SIRT1 is a novel central modulator of the earliest microvascular damage induced by age and obesity. Through a complex epigenetic control mainly involving p66Shc and Arginase II, it influences mtROS levels, NO availability, and the expression of proteins of the mitochondria respiratory chain. Therapeutic modulation of SIRT1 restores obesity- and age-related endothelial dysfunction. Early targeting of SIRT1 might represent a crucial strategy to prevent age- and obesity-related microvascular dysfunction.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy.,Scuola Superiore Sant’Anna, Pisa, Italy (A.M., V.L.R., N.B.)
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Switzerland (S.C., F.P.)
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Switzerland (S.C., F.P.).,Department of Cardiology, University Heart Center (F.P., F.R.), University Hospital Zurich, Switzerland.,Department of Research and Education (F.P.), University Hospital Zurich, Switzerland
| | - Emiliano Duranti
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Monica Nannipieri
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Rudj Mancini
- Unit of Bariatric Surgery, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy (R.M.)
| | - Michele Lai
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery (M.L., V.L.R.), University of Pisa, Italy
| | - Veronica La Rocca
- Retrovirus Center and Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery (M.L., V.L.R.), University of Pisa, Italy.,Scuola Superiore Sant’Anna, Pisa, Italy (A.M., V.L.R., N.B.)
| | - Ilaria Puxeddu
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Marco Ghionzoli
- Paediatric Surgery Unit, Meyer Children’s Hospital, Florence, Italy (M.G.)
| | - Georgios Georgiopoulos
- School of Biomedical Engineering and Imaging Sciences, King’s College London, United Kingdom (G.G.).,Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Greece (G.G.)
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy.,Scuola Superiore Sant’Anna, Pisa, Italy (A.M., V.L.R., N.B.)
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center (F.P., F.R.), University Hospital Zurich, Switzerland
| | - Nicola Riccardo Pugliese
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Agostino Virdis*
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine (A.M., E.D., M.N., I.P., L.A., M.F., C.I., N.B., N.R.P., S.T., A.V., S.M.), University of Pisa, Italy.,Institute of Cardiovascular Science, University College London, United Kingdom (S.M.)
| |
Collapse
|
5
|
De Luise M, Sollazzo M, Lama E, Coadă CA, Bressi L, Iorio M, Cavina B, D’Angelo L, Milioni S, Marchio L, Miglietta S, Coluccelli S, Tedesco G, Ghelli A, Lemma S, Perrone AM, Kurelac I, Iommarini L, Porcelli AM, Gasparre G. Inducing respiratory complex I impairment elicits an increase in PGC1α in ovarian cancer. Sci Rep 2022; 12:8020. [PMID: 35577908 PMCID: PMC9110394 DOI: 10.1038/s41598-022-11620-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/07/2022] [Indexed: 12/24/2022] Open
Abstract
AbstractAnticancer strategies aimed at inhibiting Complex I of the mitochondrial respiratory chain are increasingly being attempted in solid tumors, as functional oxidative phosphorylation is vital for cancer cells. Using ovarian cancer as a model, we show that a compensatory response to an energy crisis induced by Complex I genetic ablation or pharmacological inhibition is an increase in the mitochondrial biogenesis master regulator PGC1α, a pleiotropic coactivator of transcription regulating diverse biological processes within the cell. We associate this compensatory response to the increase in PGC1α target gene expression, setting the basis for the comprehension of the molecular pathways triggered by Complex I inhibition that may need attention as drawbacks before these approaches are implemented in ovarian cancer care.
Collapse
|
6
|
Lee YJ, Jang YN, Kim HM, Han YM, Seo HS, Eom Y, Song JS, Jeong JH, Jung TW. Stimulation of Alpha-1-Adrenergic Receptor Ameliorates Obesity-Induced Cataracts by Activating Glycolysis and Inhibiting Cataract-Inducing Factors. Endocrinol Metab (Seoul) 2022; 37:221-232. [PMID: 35316888 PMCID: PMC9081306 DOI: 10.3803/enm.2021.1237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/19/2021] [Accepted: 12/15/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Obesity, the prevalence of which is increasing due to the lack of exercise and increased consumption of Westernized diets, induces various complications, including ophthalmic diseases. For example, obesity is involved in the onset of cataracts. METHODS To clarify the effects and mechanisms of midodrine, an α1-adrenergic receptor agonist, in cataracts induced by obesity, we conducted various analytic experiments in Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a rat model of obesity. RESULTS Midodrine prevented cataract occurrence and improved lens clearance in OLETF rats. In the lenses of OLETF rats treated with midodrine, we observed lower levels of aldose reductase, tumor necrosis factor-α, and sorbitol, but higher levels of hexokinase, 5'-adenosine monophosphate-activated protein kinase-alpha, adenosine 5´-triphosphate, peroxisome proliferator-activated receptordelta, peroxisome proliferator-activated receptor gamma coactivator 1-alpha, superoxide dismutase, and catalase. CONCLUSION The ameliorating effects of midodrine on cataracts in the OLETF obesity rat model are exerted via the following three mechanisms: direct inhibition of the biosynthesis of sorbitol, which causes cataracts; reduction of reactive oxygen species and inflammation; and (3) stimulation of normal aerobic glycolysis.
Collapse
Affiliation(s)
- Yong-Jik Lee
- Cardiovascular Center, Korea University Guro Hospital, Seoul, Korea
- Cellvertics Co. Ltd., Seoul, Korea
- Department of Pharmacology, Chung-Ang University College of Medicine, Seoul, Korea
- Laboratory of Genomics and Translational Medicine, Department of Internal Medicine, Gachon University College of Medicine, Incheon, Korea
| | - Yoo-Na Jang
- Cardiovascular Center, Korea University Guro Hospital, Seoul, Korea
- Cellvertics Co. Ltd., Seoul, Korea
| | - Hyun-Min Kim
- Cardiovascular Center, Korea University Guro Hospital, Seoul, Korea
- Department of Medical Science, BK21 Plus KUMS Graduate Program, Korea University College of Medicine, Seoul, Korea
| | - Yoon-Mi Han
- Cardiovascular Center, Korea University Guro Hospital, Seoul, Korea
- Cellvertics Co. Ltd., Seoul, Korea
| | - Hong Seog Seo
- Cardiovascular Center, Korea University Guro Hospital, Seoul, Korea
- Cellvertics Co. Ltd., Seoul, Korea
- Department of Medical Science, BK21 Plus KUMS Graduate Program, Korea University College of Medicine, Seoul, Korea
| | - Youngsub Eom
- Department of Ophthalmology, Korea University Ansan Hospital, Ansan, Korea
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
| | - Jong-suk Song
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Korea
- Department of Ophthalmology, Korea University Guro Hospital, Seoul, Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Tae Woo Jung
- Department of Pharmacology, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
7
|
Mitochondrial biogenesis in organismal senescence and neurodegeneration. Mech Ageing Dev 2020; 191:111345. [DOI: 10.1016/j.mad.2020.111345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 12/19/2022]
|
8
|
Uittenbogaard M, Chiaramello A. Maternally inherited mitochondrial respiratory disorders: from pathogenetic principles to therapeutic implications. Mol Genet Metab 2020; 131:38-52. [PMID: 32624334 PMCID: PMC7749081 DOI: 10.1016/j.ymgme.2020.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023]
Abstract
Maternally inherited mitochondrial respiratory disorders are rare, progressive, and multi-systemic diseases that remain intractable, with no effective therapeutic interventions. Patients share a defective oxidative phosphorylation pathway responsible for mitochondrial ATP synthesis, in most cases due to pathogenic mitochondrial variants transmitted from mother to child or to a rare de novo mutation or large-scale deletion of the mitochondrial genome. The clinical diagnosis of these mitochondrial diseases is difficult due to exceptionally high clinical variability, while their genetic diagnosis has improved with the advent of next-generation sequencing. The mechanisms regulating the penetrance of the mitochondrial variants remain unresolved with the patient's nuclear background, epigenomic regulation, heteroplasmy, mitochondrial haplogroups, and environmental factors thought to act as rheostats. The lack of animal models mimicking the phenotypic manifestations of these disorders has hampered efforts toward curative therapies. Patient-derived cellular paradigms provide alternative models for elucidating the pathogenic mechanisms and screening pharmacological small molecules to enhance mitochondrial function. Recent progress has been made in designing promising approaches to curtail the negative impact of dysfunctional mitochondria and alleviate clinical symptoms: 1) boosting mitochondrial biogenesis; 2) shifting heteroplasmy; 3) reprogramming metabolism; and 4) administering hypoxia-based treatment. Here, we discuss their varying efficacies and limitations and provide an outlook on their therapeutic potential and clinical application.
Collapse
Affiliation(s)
- Martine Uittenbogaard
- George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, 2300 I Street N.W., Washington, DC 20037, USA
| | - Anne Chiaramello
- George Washington University School of Medicine and Health Sciences, Department of Anatomy and Cell Biology, 2300 I Street N.W., Washington, DC 20037, USA.
| |
Collapse
|
9
|
Ngoi NYL, Eu JQ, Hirpara J, Wang L, Lim JSJ, Lee SC, Lim YC, Pervaiz S, Goh BC, Wong ALA. Targeting Cell Metabolism as Cancer Therapy. Antioxid Redox Signal 2020; 32:285-308. [PMID: 31841375 DOI: 10.1089/ars.2019.7947] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Cancer cells exhibit altered metabolic pathways to keep up with biosynthetic and reduction-oxidation needs during tumor proliferation and metastasis. The common induction of metabolic pathways during cancer progression, regardless of cancer histio- or genotype, makes cancer metabolism an attractive target for therapeutic exploitation. Recent Advances: Emerging data suggest that these altered pathways may even result in resistance to anticancer therapies. Identifying specific metabolic dependencies that are unique to cancer cells has proved challenging in this field, limiting the therapeutic window for many candidate drug approaches. Critical Issues: Cancer cells display significant metabolic flexibility in nutrient-limited environments, hampering the longevity of suppressing cancer metabolism through any singular approach. Combinatorial "synthetic lethal" approaches may have a better chance for success and promising strategies are reviewed here. The dynamism of the immune system adds a level of complexity, as various immune populations in the tumor microenvironment often share metabolic pathways with cancer, with successive alterations during immune activation and quiescence. Decoding the reprogramming of metabolic pathways within cancer cells and stem cells, as well as examining metabolic symbiosis between components of the tumor microenvironment, would be essential to further meaningful drug development within the tumor's metabolic ecosystem. Future Directions: In this article, we examine evidence for the therapeutic potential of targeting metabolic alterations in cancer, and we discuss the drawbacks and successes that have stimulated this field.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Jie Qing Eu
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Jayshree Hirpara
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Joline S J Lim
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Soo-Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Yaw-Chyn Lim
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology and Medical Science Cluster Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| | - Andrea L A Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, Singapore, National University of Singapore, Singapore
| |
Collapse
|
10
|
Bokil A, Sancho P. Mitochondrial determinants of chemoresistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:634-646. [PMID: 35582564 PMCID: PMC8992520 DOI: 10.20517/cdr.2019.46] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/10/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022]
Abstract
Chemoresistance constitute nowadays the major contributor to therapy failure in most cancers. There are main factors that mitigate cell response to therapy, such as target organ, inherent sensitivity to the administered compound, its metabolism, drug efflux and influx or alterations on specific cellular targets, among others. We now know that intrinsic properties of cancer cells, including metabolic features, substantially contribute to chemoresistance. In fact, during the last years, numerous reports indicate that cancer cells resistant to chemotherapy demonstrate significant alterations in mitochondrial metabolism, membrane polarization and mass. Metabolic activity and expression of several mitochondrial proteins are modulated under treatment to cope with stress, making these organelles central players in the development of resistance to therapies. Here, we review the role of mitochondria in chemoresistant cells in terms of metabolic rewiring and function of key mitochondria-related proteins.
Collapse
Affiliation(s)
- Ansooya Bokil
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| | - Patricia Sancho
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| |
Collapse
|
11
|
Li Y, Chen J, Li Z, Li C. Mitochondrial OXPHOS is involved in the protective effects of L-arginine against heat-induced low sperm motility of boar. J Therm Biol 2019; 84:236-244. [DOI: 10.1016/j.jtherbio.2019.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/30/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022]
|
12
|
McMillan RP, Stewart S, Budnick JA, Caswell CC, Hulver MW, Mukherjee K, Srivastava S. Quantitative Variation in m.3243A > G Mutation Produce Discrete Changes in Energy Metabolism. Sci Rep 2019; 9:5752. [PMID: 30962477 PMCID: PMC6453956 DOI: 10.1038/s41598-019-42262-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/18/2019] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial DNA (mtDNA) 3243A > G tRNALeu(UUR) heteroplasmic mutation (m.3243A > G) exhibits clinically heterogeneous phenotypes. While the high mtDNA heteroplasmy exceeding a critical threshold causes mitochondrial encephalomyopathy, lactic acidosis with stroke-like episodes (MELAS) syndrome, the low mtDNA heteroplasmy causes maternally inherited diabetes with or without deafness (MIDD) syndrome. How quantitative differences in mtDNA heteroplasmy produces distinct pathological states has remained elusive. Here we show that despite striking similarities in the energy metabolic gene expression signature, the mitochondrial bioenergetics, biogenesis and fuel catabolic functions are distinct in cells harboring low or high levels of the m.3243 A > G mutation compared to wild type cells. We further demonstrate that the low heteroplasmic mutant cells exhibit a coordinate induction of transcriptional regulators of the mitochondrial biogenesis, glucose and fatty acid metabolism pathways that lack in near homoplasmic mutant cells compared to wild type cells. Altogether, these results shed new biological insights on the potential mechanisms by which low mtDNA heteroplasmy may progressively cause diabetes mellitus.
Collapse
Affiliation(s)
- Ryan P McMillan
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA.,Metabolic Phenotyping Core at Virginia Tech, Blacksburg, VA, 24061, USA
| | - Sidney Stewart
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.,Edward Via College of Osteopathic Medicine, Auburn, AL, 36832, USA
| | - James A Budnick
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Clayton C Caswell
- Department of Biomedical Sciences and Pathobiology, Center for One Health Research, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Matthew W Hulver
- Department of Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, 24061, USA.,Metabolic Phenotyping Core at Virginia Tech, Blacksburg, VA, 24061, USA
| | - Konark Mukherjee
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
| | - Sarika Srivastava
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
| |
Collapse
|
13
|
Gravel SP. Deciphering the Dichotomous Effects of PGC-1α on Tumorigenesis and Metastasis. Front Oncol 2018; 8:75. [PMID: 29629336 PMCID: PMC5876244 DOI: 10.3389/fonc.2018.00075] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022] Open
Abstract
Metabolic reprogramming confers cancer cells the ability to grow and survive under nutrient-depleted or stressful microenvironments. The amplification of oncogenes, the loss of tumor suppressors, as well as context- and lineage-specific determinants can converge and profoundly affect the metabolic status of cancer cells. Cumulating evidences suggest that highly glycolytic cells under the influence of oncogenes such as BRAF, or evolving in hypoxic microenvironments, will promote metastasis through modulation of multiple steps of tumorigenesis such as the epithelial-to-mesenchymal transition (EMT). On the contrary, increased reliance on mitochondrial respiration is associated with hyperplasic rather than metastatic disease. The PGC-1α transcriptional coactivator, a master regulator of mitochondrial biogenesis, has recently been shown to exert antimetastatic effects in cancer, notably through inhibition of EMT. Besides, PGC-1α has the opposite role in specific cancer subtypes, in which it appears to provide growth advantages. Thus, the regulation and role of PGC-1α in cancer is not univocal, and its use as a prognostic marker appears limited given its highly dynamic nature and its multifaceted regulation by transcriptional and posttranslational mechanisms. Herein, we expose key oncogenic and lineage-specific modules that finely regulate PGC-1α to promote or dampen the metastatic process. We propose a unifying model based on the systematic analysis of its controversial implication in cancer from cell proliferation to EMT and metastasis. This short review will provide a good understanding of current challenges associated with the study of PGC-1α.
Collapse
Affiliation(s)
- Simon-Pierre Gravel
- Laboratory of Metabolic Immunopharmacology, Faculty of Pharmacy, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
14
|
Peroxisome Proliferator-Activated Receptor γ and PGC-1 α in Cancer: Dual Actions as Tumor Promoter and Suppressor. PPAR Res 2018; 2018:6727421. [PMID: 29599799 PMCID: PMC5828371 DOI: 10.1155/2018/6727421] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/16/2017] [Accepted: 12/19/2017] [Indexed: 12/31/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is part of a nuclear receptor superfamily that regulates gene expression involved in cell differentiation, proliferation, immune/inflammation response, and lipid metabolism. PPARγ coactivator-1α (PGC-1α), initially identified as a PPARγ-interacting protein, is an important regulator of diverse metabolic pathways, such as oxidative metabolism and energy homeostasis. The role of PGC-1α in diabetes, neurodegeneration, and cardiovascular disease is particularly well known. PGC-1α is also now known to play important roles in cancer, independent of the role of PPARγ in cancer. Though many researchers have studied the expression and clinical implications of PPARγ and PGC-1α in cancer, there are still many controversies about the role of PPARγ and PGC-1α in cancer. This review examines and summarizes some recent data on the role and action mechanisms of PPARγ and PGC-1α in cancer, respectively, particularly the recent progress in understanding the role of PPARγ in several cancers since our review was published in 2012.
Collapse
|
15
|
Alterations in PGC1α expression levels are involved in colorectal cancer risk: a qualitative systematic review. BMC Cancer 2017; 17:731. [PMID: 29121859 PMCID: PMC5679491 DOI: 10.1186/s12885-017-3725-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 10/30/2017] [Indexed: 11/24/2022] Open
Abstract
Background Colorectal cancer (CRC) is a major global public health problem and the second leading cause of cancer-related death. Mitochondrial dysfunction has long been suspected to be involved in this type of tumorigenesis, as supported by an accumulating body of research evidence. However, little is known about how mitochondrial alterations contribute to tumorigenesis. Mitochondrial biogenesis is a fundamental cellular process required to maintain functional mitochondria and as an adaptive mechanism in response to changing energy requirements. Mitochondrial biogenesis is regulated by peroxisome proliferator-activated receptor gamma coactivator 1-α (PPARGC1A or PGC1α). In this paper, we report a systematic review to summarize current evidence on the role of PGC1α in the initiation and progression of CRC. The aim is to provide a basis for more comprehensive research. Methods The literature search, data extraction and quality assessment were performed according to the document Guidance on the Conduct of Narrative Synthesis in Systematic Reviews and the PRISMA declaration. Results The studies included in this review aimed to evaluate whether increased or decreased PGC1α expression affects the development of CRC. Each article proposes a possible molecular mechanism of action and we create two concept maps. Conclusion Our systematic review indicates that altered expression of PGC1α modifies CRC risk. Most studies showed that overexpression of this gene increases CRC risk, while some studies indicated that lower than normal expression levels could increase CRC risk. Thus, various authors propose PGC1α as a good candidate molecular target for cancer therapy. Reducing expression of this gene could help to reduce risk or progression of CRC. Electronic supplementary material The online version of this article (10.1186/s12885-017-3725-3) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Pereira CV, Moraes CT. Current strategies towards therapeutic manipulation of mtDNA heteroplasmy. Front Biosci (Landmark Ed) 2017; 22:991-1010. [PMID: 27814659 DOI: 10.2741/4529] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mitochondrial disease is a multifactorial disorder involving both nuclear and mitochondrial genomes. Over the past 20 years, great progress was achieved in the field of gene editing which raised the possibility of partial or complete elimination of mutant mtDNA that causes disease phenotypes. Each cell contains thousands of copies of mtDNA which can be either wild-type (WT) or mutant, a condition called heteroplasmy. As there are multiple copies of mtDNA inside a cell, the percentage of mutant mtDNA can vary and a directional shift in the heteroplasmy ratio towards an increase of WT mtDNA copies would have therapeutic value. Gene editing tools have been adapted to translocate to mitochondria and were able to change heteroplasmy in a predictable manner. These include mitochondrial targeted restriction endonucleases, Zinc-finger nucleases, and TAL-effector nucleases. These procedures could also be adapted to reduce the levels of mutant mtDNA in embryos, offering an option to the controversial mitochondrial replacement techniques during in vitro fertilization. The current strategies to induce heteroplasmy shift of mtDNA and its implications will be comprehensively discussed.
Collapse
Affiliation(s)
- Claudia V Pereira
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carlos T Moraes
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA,
| |
Collapse
|
17
|
Bromodomain Inhibitors Correct Bioenergetic Deficiency Caused by Mitochondrial Disease Complex I Mutations. Mol Cell 2016; 64:163-175. [PMID: 27666594 DOI: 10.1016/j.molcel.2016.08.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/01/2016] [Accepted: 08/16/2016] [Indexed: 12/20/2022]
Abstract
Mitochondrial diseases comprise a heterogeneous group of genetically inherited disorders that cause failures in energetic and metabolic function. Boosting residual oxidative phosphorylation (OXPHOS) activity can partially correct these failures. Herein, using a high-throughput chemical screen, we identified the bromodomain inhibitor I-BET 525762A as one of the top hits that increases COX5a protein levels in complex I (CI) mutant cybrid cells. In parallel, bromodomain-containing protein 4 (BRD4), a target of I-BET 525762A, was identified using a genome-wide CRISPR screen to search for genes whose loss of function rescues death of CI-impaired cybrids grown under conditions requiring OXPHOS activity for survival. We show that I-BET525762A or loss of BRD4 remodeled the mitochondrial proteome to increase the levels and activity of OXPHOS protein complexes, leading to rescue of the bioenergetic defects and cell death caused by mutations or chemical inhibition of CI. These studies show that BRD4 inhibition may have therapeutic implications for the treatment of mitochondrial diseases.
Collapse
|
18
|
Srivastava S. Emerging therapeutic roles for NAD(+) metabolism in mitochondrial and age-related disorders. Clin Transl Med 2016; 5:25. [PMID: 27465020 PMCID: PMC4963347 DOI: 10.1186/s40169-016-0104-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/26/2016] [Indexed: 12/12/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a central metabolic cofactor in eukaryotic cells that plays a critical role in regulating cellular metabolism and energy homeostasis. NAD+ in its reduced form (i.e. NADH) serves as the primary electron donor in mitochondrial respiratory chain, which involves adenosine triphosphate production by oxidative phosphorylation. The NAD+/NADH ratio also regulates the activity of various metabolic pathway enzymes such as those involved in glycolysis, Kreb’s cycle, and fatty acid oxidation. Intracellular NAD+ is synthesized de novo from l-tryptophan, although its main source of synthesis is through salvage pathways from dietary niacin as precursors. NAD+ is utilized by various proteins including sirtuins, poly ADP-ribose polymerases (PARPs) and cyclic ADP-ribose synthases. The NAD+ pool is thus set by a critical balance between NAD+ biosynthetic and NAD+ consuming pathways. Raising cellular NAD+ content by inducing its biosynthesis or inhibiting the activity of PARP and cADP-ribose synthases via genetic or pharmacological means lead to sirtuins activation. Sirtuins modulate distinct metabolic, energetic and stress response pathways, and through their activation, NAD+ directly links the cellular redox state with signaling and transcriptional events. NAD+ levels decline with mitochondrial dysfunction and reduced NAD+/NADH ratio is implicated in mitochondrial disorders, various age-related pathologies as well as during aging. Here, I will provide an overview of the current knowledge on NAD+ metabolism including its biosynthesis, utilization, compartmentalization and role in the regulation of metabolic homoeostasis. I will further discuss how augmenting intracellular NAD+ content increases oxidative metabolism to prevent bioenergetic and functional decline in multiple models of mitochondrial diseases and age-related disorders, and how this knowledge could be translated to the clinic for human relevance.
Collapse
Affiliation(s)
- Sarika Srivastava
- Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA.
| |
Collapse
|
19
|
Zandberg L, van Dyk HC, van der Westhuizen FH, van Dijk AA. A 3-methylcrotonyl-CoA carboxylase deficient human skin fibroblast transcriptome reveals underlying mitochondrial dysfunction and oxidative stress. Int J Biochem Cell Biol 2016; 78:116-129. [PMID: 27417235 DOI: 10.1016/j.biocel.2016.07.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 07/07/2016] [Accepted: 07/11/2016] [Indexed: 01/03/2023]
Abstract
Isolated 3-methylcrotonyl-CoA carboxylase (MCC) deficiency is an autosomal recessive inherited metabolic disease of leucine catabolism with a highly variable phenotype. Apart from extensive mutation analyses of the MCCC1 and MCCC2 genes encoding 3-methylcrotonyl-CoA carboxylase (EC 6.4.1.4), molecular data on MCC deficiency gene expression studies in human tissues is lacking. For IEMs, unbiased '-omics' approaches are starting to reveal the secondary cellular responses to defects in biochemical pathways. Here we present the first whole genome expression profile of immortalized cultured skin fibroblast cells of two clinically affected MCC deficient patients and two healthy individuals generated using Affymetrix(®)HuExST1.0 arrays. There were 16191 significantly differentially expressed transcript IDs of which 3591 were well annotated and present in the predefined knowledge database of Ingenuity Pathway Analysis software used for downstream functional analyses. The most noticeable feature of this MCCA deficient skin fibroblast transcriptome was the typical genetic hallmark of mitochondrial dysfunction, decreased antioxidant response and disruption of energy homeostasis, which was confirmed by mitochondrial functional analyses. The MCC deficient transcriptome seems to predict oxidative stress that could alter the complex secondary cellular response that involve genes of the glycolysis, the TCA cycle, OXPHOS, gluconeogenesis, β-oxidation and the branched-chain fatty acid metabolism. An important emerging insight from this human MCCA transcriptome in combination with previous reports is that chronic exposure to the primary and secondary metabolites of MCC deficiency and the resulting oxidative stress might impact adversely on the quality of life and energy levels, irrespective of whether MCC deficient individuals are clinically affected or asymptomatic.
Collapse
Affiliation(s)
- L Zandberg
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - H C van Dyk
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - F H van der Westhuizen
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - A A van Dijk
- Biochemistry Division, Centre for Human Metabolomics, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa.
| |
Collapse
|
20
|
PPARGC1B gene is associated with Kashin-Beck disease in Han Chinese. Funct Integr Genomics 2016; 16:459-63. [PMID: 27108113 DOI: 10.1007/s10142-016-0496-x] [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: 11/09/2015] [Revised: 04/08/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
Kashin-Beck disease (KBD) is a chronic osteochondropathy. The genetic basis of KBD remains elusive now. To investigate the relationship between PPARGC1B gene polymorphism and KBD, we conducted a two-stage association study using 2743 unrelated Han Chinese subjects. In the first stage, three SNPs rs1078324, rs4705372, and rs11743128 of PPARGC1B gene were genotyped in 559 KBD patients and 467 health controls using Sequenom MassARRAY platform. In the second stage, the association analysis results of PPARGC1B with KBD were replicated using an independent sample of 1717 subjects. SNP association analysis was conducted by PLINK software. Genotype imputation was conducted by IMPUTE 2.0 against the reference panel of the 1000 genome project. Bonferroni multiple testing correction was performed. We observed a significant association signal at rs4705372 (P = 0.0160) and a suggestive association signal at rs11743128 (P = 0.0290). Further replication study confirmed the association signals of rs4705372 (P = 0.0026) and rs11743128 (P = 0.0387) in the independent validation sample. Our study results suggest that PPARGC1B is a novel susceptibility gene of KBD.
Collapse
|
21
|
X-linked intellectual disability gene CASK regulates postnatal brain growth in a non-cell autonomous manner. Acta Neuropathol Commun 2016; 4:30. [PMID: 27036546 PMCID: PMC4818453 DOI: 10.1186/s40478-016-0295-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 01/07/2023] Open
Abstract
The phenotypic spectrum among girls with heterozygous mutations in the X-linked intellectual disability (XLID) gene CASK (calcium/calmodulin-dependent serine protein kinase) includes postnatal microcephaly, ponto-cerebellar hypoplasia, seizures, optic nerve hypoplasia, growth retardation and hypotonia. Although CASK knockout mice were previously reported to exhibit perinatal lethality and a 3-fold increased apoptotic rate in the brain, CASK deletion was not found to affect neuronal physiology and their electrical properties. The pathogenesis of CASK associated disorders and the potential function of CASK therefore remains unknown. Here, using Cre-LoxP mediated gene excision experiments; we demonstrate that deleting CASK specifically from mouse cerebellar neurons does not alter the cerebellar architecture or function. We demonstrate that the neuron-specific deletion of CASK in mice does not cause perinatal lethality but induces severe recurrent epileptic seizures and growth retardation before the onset of adulthood. Furthermore, we demonstrate that although neuron-specific haploinsufficiency of CASK is inconsequential, the CASK mutation associated human phenotypes are replicated with high fidelity in CASK heterozygous knockout female mice (CASK(+/-)). These data suggest that CASK-related phenotypes are not purely neuronal in origin. Surprisingly, the observed microcephaly in CASK(+/-) animals is not associated with a specific loss of CASK null brain cells indicating that CASK regulates postnatal brain growth in a non-cell autonomous manner. Using biochemical assay, we also demonstrate that CASK can interact with metabolic proteins. CASK knockdown in human cell lines cause reduced cellular respiration and CASK(+/-) mice display abnormalities in muscle and brain oxidative metabolism, suggesting a novel function of CASK in metabolism. Our data implies that some phenotypic components of CASK heterozygous deletion mutation associated disorders represent systemic manifestation of metabolic stress and therefore amenable to therapeutic intervention.
Collapse
|
22
|
Chen N, Cheng J, Zhou L, Lei T, Chen L, Shen Q, Qin L, Wan Z. Effects of treadmill running and rutin on lipolytic signaling pathways and TRPV4 protein expression in the adipose tissue of diet-induced obese mice. J Physiol Biochem 2015; 71:733-42. [PMID: 26424736 DOI: 10.1007/s13105-015-0437-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/23/2015] [Indexed: 01/14/2023]
Abstract
To explore the effects of rutin and exercise on high-fat diet (HFD)-induced disrupted lipolytic signaling, adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling, transient receptor potential cation channel subfamily V member 4 (TRPV4) and its associated protein expression, and whether depot-specific effects existed. C57BL/6J mice were randomized into five groups: chow group, HFD, HFD plus rutin intervention group (HR), HFD combined with treadmill running group (HE), and HFD combined with treadmill running and rutin intervention group (HRE). At the end of the 16-week intervention, lipolytic markers, AMPK signaling pathways, TRPV4, and peroxisome proliferator-activated receptor gamma coactivator 1α + β (PGC-1α + β) from adipose tissue were measured by western blotting. In epididymal adipose tissue, HFD resulted in significant reduction in the phosphorylation of hormone sensitive lipase at serine660 (p-HSL660), perilipin A, phosphoenolpyruvate carboxykinase (PEPCK), p-AMPK, and p-acetyl-CoA carboxylase (ACC) protein expression. Exercise intervention and exercise plus rutin completely restored p-HSL660, perilipin A, PEPCK, p-AMPK, and p-ACC protein expression to normal level. HFD and HR groups have reduced expression of PGC-1α + β, exercise, and exercise plus rutin completely restored PGC-1α + β expression to normal level. In subcutaneous adipose tissue, HFD elevated TRPV4, exercise, and exercise plus rutin completely reduced TRPV4 to normal level. HR, HE, and HRE group have increased PGC-1α + β. In conclusion, depot-specific effects existed in regards to how rutin and exercise affect lipolytic signaling and p-AMPK, as well as TRPV4 and PGC-1α + β expression.
Collapse
Affiliation(s)
- Neng Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Renai Road 199, Dushuhu Higher Education Town, Suzhou, China, 215123
| | - Jinbo Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Renai Road 199, Dushuhu Higher Education Town, Suzhou, China, 215123
| | - Lingmei Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Renai Road 199, Dushuhu Higher Education Town, Suzhou, China, 215123
| | - Ting Lei
- Suzhou Industrial Park Center Disease Control and Prevention, 58 Suqian Road, Suzhou, 215123, People's Republic of China
| | - Lihua Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Renai Road 199, Dushuhu Higher Education Town, Suzhou, China, 215123
| | - Qiang Shen
- Suzhou Center for Disease Control and Prevention, 72 Sanxiang Road, 215004, Suzhou, People's Republic of China
| | - Liqiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Renai Road 199, Dushuhu Higher Education Town, Suzhou, China, 215123.
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Renai Road 199, Dushuhu Higher Education Town, Suzhou, China, 215123. .,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, Soochow University, 199 Renai Road, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
23
|
Ripolone M, Ronchi D, Violano R, Vallejo D, Fagiolari G, Barca E, Lucchini V, Colombo I, Villa L, Berardinelli A, Balottin U, Morandi L, Mora M, Bordoni A, Fortunato F, Corti S, Parisi D, Toscano A, Sciacco M, DiMauro S, Comi GP, Moggio M. Impaired Muscle Mitochondrial Biogenesis and Myogenesis in Spinal Muscular Atrophy. JAMA Neurol 2015; 72:666-75. [PMID: 25844556 PMCID: PMC4944827 DOI: 10.1001/jamaneurol.2015.0178] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE The important depletion of mitochondrial DNA (mtDNA) and the general depression of mitochondrial respiratory chain complex levels (including complex II) have been confirmed, implying an increasing paucity of mitochondria in the muscle from patients with types I, II, and III spinal muscular atrophy (SMA-I, -II, and -III, respectively). OBJECTIVE To investigate mitochondrial dysfunction in a large series of muscle biopsy samples from patients with SMA. DESIGN, SETTING, AND PARTICIPANTS We studied quadriceps muscle samples from 24 patients with genetically documented SMA and paraspinal muscle samples from 3 patients with SMA-II undergoing surgery for scoliosis correction. Postmortem muscle samples were obtained from 1 additional patient. Age-matched controls consisted of muscle biopsy specimens from healthy children aged 1 to 3 years who had undergone analysis for suspected myopathy. Analyses were performed at the Neuromuscular Unit, Istituto di Ricovero e Cura a Carattere Scientifico Foundation Ca' Granda Ospedale Maggiore Policlinico-Milano, from April 2011 through January 2015. EXPOSURES We used histochemical, biochemical, and molecular techniques to examine the muscle samples. MAIN OUTCOMES AND MEASURES Respiratory chain activity and mitochondrial content. RESULTS Results of histochemical analysis revealed that cytochrome-c oxidase (COX) deficiency was more evident in muscle samples from patients with SMA-I and SMA-II. Residual activities for complexes I, II, and IV in muscles from patients with SMA-I were 41%, 27%, and 30%, respectively, compared with control samples (P < .005). Muscle mtDNA content and cytrate synthase activity were also reduced in all 3 SMA types (P < .05). We linked these alterations to downregulation of peroxisome proliferator-activated receptor coactivator 1α, the transcriptional activators nuclear respiratory factor 1 and nuclear respiratory factor 2, mitochondrial transcription factor A, and their downstream targets, implying depression of the entire mitochondrial biogenesis. Results of Western blot analysis confirmed the reduced levels of the respiratory chain subunits that included mitochondrially encoded COX1 (47.5%; P = .004), COX2 (32.4%; P < .001), COX4 (26.6%; P < .001), and succinate dehydrogenase complex subunit A (65.8%; P = .03) as well as the structural outer membrane mitochondrial porin (33.1%; P < .001). Conversely, the levels of expression of 3 myogenic regulatory factors-muscle-specific myogenic factor 5, myoblast determination 1, and myogenin-were higher in muscles from patients with SMA compared with muscles from age-matched controls (P < .05). CONCLUSIONS AND RELEVANCE Our results strongly support the conclusion that an altered regulation of myogenesis and a downregulated mitochondrial biogenesis contribute to pathologic change in the muscle of patients with SMA. Therapeutic strategies should aim at counteracting these changes.
Collapse
Affiliation(s)
- Michela Ripolone
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Dario Ronchi
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Raffaella Violano
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Dionis Vallejo
- Sien-Servicios Integrales en Neurologia, Universidad de Antioquia, Medellin, Colombia
| | - Gigliola Fagiolari
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Emanuele Barca
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Valeria Lucchini
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Irene Colombo
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Luisa Villa
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Angela Berardinelli
- Child Neuropsychiatry Unit, C. Mondino National Neurological Institute, Pavia, Italy
| | - Umberto Balottin
- Child Neuropsychiatry Unit, C. Mondino National Neurological Institute, Pavia, Italy
| | - Lucia Morandi
- Neuromuscular Diseases and Neuroimmunology Unit, Department of Clinical Neurosciences, IRCCS Foundation, Carlo Besta Neurological Institute, Milan, Italy
| | - Marina Mora
- Neuromuscular Diseases and Neuroimmunology Unit, Department of Clinical Neurosciences, IRCCS Foundation, Carlo Besta Neurological Institute, Milan, Italy
| | - Andreina Bordoni
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Francesco Fortunato
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Stefania Corti
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Daniela Parisi
- Reference Center for Rare Neuromuscular Disorders, Department of Neurosciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico G. Martino, Messina, Italy
| | - Antonio Toscano
- Reference Center for Rare Neuromuscular Disorders, Department of Neurosciences, University of Messina, Azienda Ospedaliera Universitaria Policlinico G. Martino, Messina, Italy
| | - Monica Sciacco
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Salvatore DiMauro
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Giacomo P Comi
- Neurology Unit, Neuroscience Section, Department of Pathophysiology and Transplantation, Dino Ferrari Centre, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Maurizio Moggio
- Neuromuscular Unit, Dino Ferrari Centre, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Fayzulin RZ, Perez M, Kozhukhar N, Spadafora D, Wilson GL, Alexeyev MF. A method for mutagenesis of mouse mtDNA and a resource of mouse mtDNA mutations for modeling human pathological conditions. Nucleic Acids Res 2015; 43:e62. [PMID: 25820427 PMCID: PMC4482060 DOI: 10.1093/nar/gkv140] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/10/2015] [Indexed: 12/23/2022] Open
Abstract
Mutations in human mitochondrial DNA (mtDNA) can cause mitochondrial disease and have been associated with neurodegenerative disorders, cancer, diabetes and aging. Yet our progress toward delineating the precise contributions of mtDNA mutations to these conditions is impeded by the limited availability of faithful transmitochondrial animal models. Here, we report a method for the isolation of mutations in mouse mtDNA and its implementation for the generation of a collection of over 150 cell lines suitable for the production of transmitochondrial mice. This method is based on the limited mutagenesis of mtDNA by proofreading-deficient DNA-polymerase γ followed by segregation of the resulting highly heteroplasmic mtDNA population by means of intracellular cloning. Among generated cell lines, we identify nine which carry mutations affecting the same amino acid or nucleotide positions as in human disease, including a mutation in the ND4 gene responsible for 70% of Leber Hereditary Optic Neuropathies (LHON). Similar to their human counterparts, cybrids carrying the homoplasmic mouse LHON mutation demonstrated reduced respiration, reduced ATP content and elevated production of mitochondrial reactive oxygen species (ROS). The generated resource of mouse mtDNA mutants will be useful both in modeling human mitochondrial disease and in understanding the mechanisms of ROS production mediated by mutations in mtDNA.
Collapse
Affiliation(s)
- Rafik Z Fayzulin
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, AL 36688, USA
| | - Michael Perez
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Natalia Kozhukhar
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Domenico Spadafora
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| | - Glenn L Wilson
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, AL 36688, USA
| | - Mikhail F Alexeyev
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
25
|
Uittenbogaard M, Chiaramello A. Mitochondrial biogenesis: a therapeutic target for neurodevelopmental disorders and neurodegenerative diseases. Curr Pharm Des 2014; 20:5574-93. [PMID: 24606804 PMCID: PMC4823001 DOI: 10.2174/1381612820666140305224906] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 03/03/2014] [Indexed: 11/22/2022]
Abstract
In the developing and mature brain, mitochondria act as central hubs for distinct but interwined pathways, necessary for neural development, survival, activity, connectivity and plasticity. In neurons, mitochondria assume diverse functions, such as energy production in the form of ATP, calcium buffering and generation of reactive oxygen species. Mitochondrial dysfunction contributes to a range of neurodevelopmental and neurodegenerative diseases, making mitochondria a potential target for pharmacological-based therapies. Pathogenesis associated with these diseases is accompanied by an increase in mitochondrial mass, a quantitative increase to overcome a qualitative deficiency due to mutated mitochondrial proteins that are either nuclear- or mitochondrial-encoded. This compensatory biological response is maladaptive, as it fails to sufficiently augment the bioenergetically functional mitochondrial mass and correct for the ATP deficit. Since regulation of neuronal mitochondrial biogenesis has been scantily investigated, our current understanding on the network of transcriptional regulators, co-activators and signaling regulators mainly derives from other cellular systems. The purpose of this review is to present the current state of our knowledge and understanding of the transcriptional and signaling cascades controlling neuronal mitochondrial biogenesis and the various therapeutic approaches to enhance the functional mitochondrial mass in the context of neurodevelopmental disorders and adult-onset neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Anne Chiaramello
- George Washington University School of Medicine and Health Sciences, Department of Anatomy and Regenerative Biology, 2300 I Street N.W., Washington DC 20037.
| |
Collapse
|
26
|
Hofer A, Noe N, Tischner C, Kladt N, Lellek V, Schauß A, Wenz T. Defining the action spectrum of potential PGC-1α activators on a mitochondrial and cellular level in vivo. Hum Mol Genet 2013; 23:2400-15. [PMID: 24334768 DOI: 10.1093/hmg/ddt631] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Previous studies have demonstrated a therapeutic benefit of pharmaceutical PGC-1α activation in cellular and murine model of disorders linked to mitochondrial dysfunction. While in some cases, this effect seems to be clearly associated with boosting of mitochondrial function, additional alterations as well as tissue- and cell-type-specific effects might play an important role. We initiated a comprehensive analysis of the effects of potential PGC-1α-activating drugs and pharmaceutically targeted the PPAR (bezafibrate, rosiglitazone), AMPK (AICAR, metformin) and Sirt1 (resveratrol) pathways in HeLa cells, neuronal cells and PGC-1α-deficient MEFs to get insight into cell type specificity and PGC-1α dependence of their working action. We used bezafibrate as a model drug to assess the effect on a tissue-specific level in a murine model. Not all analyzed drugs activate the PGC pathway or alter mitochondrial protein levels. However, they all affect supramolecular assembly of OXPHOS complexes and OXPHOS protein stability. In addition, a clear drug- and cell-type-specific influence on several cellular stress pathways as well as on post-translational modifications could be demonstrated, which might be relevant to fully understand the action of the analyzed drugs in the disease state. Importantly, the effect on the activation of mitochondrial biogenesis and stress response program upon drug treatment is PGC-1α dependent in MEFs demonstrating not only the pleiotropic effects of this molecule but points also to the working mechanism of the analyzed drugs. The definition of the action spectrum of the different drugs forms the basis for a defect-specific compensation strategy and a future personalized therapeutic approach.
Collapse
Affiliation(s)
- Annette Hofer
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, 50674 Cologne, Germany
| | | | | | | | | | | | | |
Collapse
|
27
|
Damiano M, Diguet E, Malgorn C, D'Aurelio M, Galvan L, Petit F, Benhaim L, Guillermier M, Houitte D, Dufour N, Hantraye P, Canals JM, Alberch J, Delzescaux T, Déglon N, Beal MF, Brouillet E. A role of mitochondrial complex II defects in genetic models of Huntington's disease expressing N-terminal fragments of mutant huntingtin. Hum Mol Genet 2013; 22:3869-82. [PMID: 23720495 PMCID: PMC3766181 DOI: 10.1093/hmg/ddt242] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by an abnormal expansion of a CAG repeat encoding a polyglutamine tract in the huntingtin (Htt) protein. The mutation leads to neuronal death through mechanisms which are still unknown. One hypothesis is that mitochondrial defects may play a key role. In support of this, the activity of mitochondrial complex II (C-II) is preferentially reduced in the striatum of HD patients. Here, we studied C-II expression in different genetic models of HD expressing N-terminal fragments of mutant Htt (mHtt). Western blot analysis showed that the expression of the 30 kDa Iron–Sulfur (Ip) subunit of C-II was significantly reduced in the striatum of the R6/1 transgenic mice, while the levels of the FAD containing catalytic 70 kDa subunit (Fp) were not significantly changed. Blue native gel analysis showed that the assembly of C-II in mitochondria was altered early in N171-82Q transgenic mice. Early loco-regional reduction in C-II activity and Ip protein expression was also demonstrated in a rat model of HD using intrastriatal injection of lentiviral vectors encoding mHtt. Infection of the rat striatum with a lentiviral vector coding the C-II Ip or Fp subunits induced a significant overexpression of these proteins that led to significant neuroprotection of striatal neurons against mHtt neurotoxicity. These results obtained in vivo support the hypothesis that structural and functional alterations of C-II induced by mHtt may play a critical role in the degeneration of striatal neurons in HD and that mitochondrial-targeted therapies may be useful in its treatment.
Collapse
Affiliation(s)
- Maria Damiano
- The first two authors contributed equally to the present study
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gong B, Pan Y, Vempati P, Zhao W, Knable L, Ho L, Wang J, Sastre M, Ono K, Sauve AA, Pasinetti GM. Nicotinamide riboside restores cognition through an upregulation of proliferator-activated receptor-γ coactivator 1α regulated β-secretase 1 degradation and mitochondrial gene expression in Alzheimer's mouse models. Neurobiol Aging 2013; 34:1581-8. [PMID: 23312803 DOI: 10.1016/j.neurobiolaging.2012.12.005] [Citation(s) in RCA: 267] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 12/05/2012] [Accepted: 12/09/2012] [Indexed: 12/23/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD)(+), a coenzyme involved in redox activities in the mitochondrial electron transport chain, has been identified as a key regulator of the lifespan-extending effects, and the activation of NAD(+) expression has been linked with a decrease in beta-amyloid (Aβ) toxicity in Alzheimer's disease (AD). Nicotinamide riboside (NR) is a NAD(+) precursor, it promotes peroxisome proliferator-activated receptor-γ coactivator 1 (PGC)-1α expression in the brain. Evidence has shown that PGC-1α is a crucial regulator of Aβ generation because it affects β-secretase (BACE1) degradation. In this study we tested the hypothesis that NR treatment in an AD mouse model could attenuate Aβ toxicity through the activation of PGC-1α-mediated BACE1 degradation. Using the Tg2576 AD mouse model, using in vivo behavioral analyses, biochemistry assays, small hairpin RNA (shRNA) gene silencing and electrophysiological recording, we found (1) dietary treatment of Tg2576 mice with 250 mg/kg/day of NR for 3 months significantly attenuates cognitive deterioration in Tg2576 mice and coincides with an increase in the steady-state levels of NAD(+) in the cerebral cortex; (2) application of NR to hippocampal slices (10 μM) for 4 hours abolishes the deficits in long-term potentiation recorded in the CA1 region of Tg2576 mice; (3) NR treatment promotes PGC-1α expression in the brain coinciding with enhanced degradation of BACE1 and the reduction of Aβ production in Tg2576 mice. Further in vitro studies confirmed that BACE1 protein content is decreased by NR treatment in primary neuronal cultures derived from Tg2576 embryos, in which BACE1 degradation was prevented by PGC-1α-shRNA gene silencing; and (4) NR treatment and PGC-1α overexpression enhance BACE1 ubiquitination and proteasomal degradation. Our studies suggest that dietary treatment with NR might benefit AD cognitive function and synaptic plasticity, in part by promoting PGC-1α-mediated BACE1 ubiquitination and degradation, thus preventing Aβ production in the brain.
Collapse
Affiliation(s)
- Bing Gong
- Department of Neurology, Center of Excellence for Novel Approaches to Neurotherapeutics, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pagliei B, Aquilano K, Baldelli S, Ciriolo MR. Garlic-derived diallyl disulfide modulates peroxisome proliferator activated receptor gamma co-activator 1 alpha in neuroblastoma cells. Biochem Pharmacol 2012. [PMID: 23178651 DOI: 10.1016/j.bcp.2012.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The peroxisome proliferator activated receptor gamma co-activator 1 alpha (PGC1α) is an inducible transcriptional co-activator with direct function in the induction of mitochondrial biogenesis. In the present report we show that, in SH-SY5Y neuroblastoma cells, garlic-derived diallyl disulfide (DADS) is able to increase PGC1α expression in a ROS-dependent manner and to induce mitochondrial biogenesis at early stage of treatment that precede cell cycle arrest and apoptosis outcome. In particular, we demonstrate that DADS elicits: i) the increase of PGC1α within nuclear compartment; ii) the decrease of PGC1α non-active acetylated form; iii) the induction of nuclear-encoded mitochondrial genes such as TFAM and TFBM1. We also show an accumulation of PGC1α within mitochondria along with an increased association with the regulatory D-Loop region of mtDNA and a concomitant augmented expression of mitochondrial RNA. Such events are related to a prompt elevation of mitochondrial mass, as assessed by evaluating the content of mtDNA. We show that the induction of mitochondrial biogenesis is directed to dampen the cytotoxic effect of DADS. Indeed, PGC1α overexpression or down-regulation prevents or exacerbates mtDNA loss and apoptosis. Overall the data highlight an anti-apoptotic role of PGC1α-mediated mitochondrial biogenesis in neuroblatoma cells and suggest PGC1α as a potential target for enhancing the effectiveness of therapy in aggressive neuroblastoma with high drug-resistance.
Collapse
Affiliation(s)
- Beatrice Pagliei
- Dept. Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | | | | | | |
Collapse
|
30
|
Read-through therapy for mitochondrial DNA nonsense mutations. Drug Discov Today 2012; 17:1063-7. [DOI: 10.1016/j.drudis.2012.04.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/27/2012] [Accepted: 04/23/2012] [Indexed: 12/11/2022]
|
31
|
Carter HN, Hood DA. Contractile activity-induced mitochondrial biogenesis and mTORC1. Am J Physiol Cell Physiol 2012; 303:C540-7. [PMID: 22700793 DOI: 10.1152/ajpcell.00156.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In response to exercise training, or chronic contractile activity, mitochondrial content is known to be enriched within skeletal muscle. However, the molecular mechanisms that mediate this adaptation are incompletely defined. Recently, the protein complex, mammalian target of rapamycin complex 1 (mTORC1), has been identified to facilitate the expression of nuclear genes encoding mitochondrial proteins (NUGEMPs) in resting muscle cells via the interaction of the mTORC1 components, mTOR and raptor, the transcription factor Yin Yang 1, and peroxisome proliferator-activated receptor-γ coactivator-1α. It is currently unknown if this mechanism is operative during the increase in mitochondrial content that occurs within skeletal muscle with chronic contractile activity (CCA). Thus we employed a cell culture model of murine skeletal muscle and subjected the myotubes to CCA for 3 h per day for 4 consecutive days in the presence or absence of the mTORC1 inhibitor rapamycin. CCA produced increases in the mitochondrial markers cytochrome oxidase (COX) IV (2.5-fold), Tfam (1.5-fold), and COX activity (1.6-fold). Rapamycin-mediated inhibition of mTORC1 did not suppress these CCA-induced increases in mitochondrial proteins and organelle content. mTORC1 inhibition alone produced a selective upregulation of mitochondrial proteins (COX IV, Tfam), but diminished organelle state 3 respiration. CCA restored this impairment to normal. Our results suggest that mTORC1 activity is not integral for the increase in mitochondrial content elicited by CCA, but is required to maintain mitochondrial function and homeostasis in resting muscle.
Collapse
Affiliation(s)
- Heather N. Carter
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - David A. Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Dillon LM, Rebelo AP, Moraes CT. The role of PGC-1 coactivators in aging skeletal muscle and heart. IUBMB Life 2012; 64:231-41. [PMID: 22279035 DOI: 10.1002/iub.608] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 12/05/2011] [Indexed: 12/11/2022]
Abstract
Aging is the progressive decline in cellular, tissue, and organ function. This complex process often manifests as loss of muscular strength, cardiovascular function, and cognitive ability. Mitochondrial dysfunction and decreased mitochondrial biogenesis are believed to participate in metabolic abnormalities and loss of organ function, which will eventually contribute to aging and decreased lifespan. In this review, we discuss what is currently known about mitochondrial dysfunction in the aging skeletal muscle and heart. We focused our discussion on the role of PGC-1 coactivators in the regulation of mitochondrial biogenesis and function and possible therapeutic benefits of increased mitochondrial biogenesis in compensating for mitochondrial dysfunction and circumventing aging and aging-related diseases.
Collapse
Affiliation(s)
- Lloye M Dillon
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, FL, USA
| | | | | |
Collapse
|
33
|
Nouws J, Nijtmans LGJ, Smeitink JA, Vogel RO. Assembly factors as a new class of disease genes for mitochondrial complex I deficiency: cause, pathology and treatment options. Brain 2011; 135:12-22. [DOI: 10.1093/brain/awr261] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
34
|
Jones AWE, Yao Z, Vicencio JM, Karkucinska-Wieckowska A, Szabadkai G. PGC-1 family coactivators and cell fate: roles in cancer, neurodegeneration, cardiovascular disease and retrograde mitochondria-nucleus signalling. Mitochondrion 2011; 12:86-99. [PMID: 21983689 DOI: 10.1016/j.mito.2011.09.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 09/08/2011] [Accepted: 09/16/2011] [Indexed: 12/29/2022]
Abstract
Over the past two decades, a complex nuclear transcriptional machinery controlling mitochondrial biogenesis and function has been described. Central to this network are the PGC-1 family coactivators, characterised as master regulators of mitochondrial biogenesis. Recent literature has identified a broader role for PGC-1 coactivators in both cell death and cellular adaptation under conditions of stress, here reviewed in the context of the pathology associated with cancer, neurodegeneration and cardiovascular disease. Moreover, we propose that these studies also imply a novel conceptual framework on the general role of mitochondrial dysfunction in disease. It is now well established that the complex nuclear transcriptional control of mitochondrial biogenesis allows for adaptation of mitochondrial mass and function to environmental conditions. On the other hand, it has also been suggested that mitochondria alter their function according to prevailing cellular energetic requirements and thus function as sensors that generate signals to adjust fundamental cellular processes through a retrograde mitochondria-nucleus signalling pathway. Therefore, altered mitochondrial function can affect cell fate not only directly by modifying cellular energy levels or redox state, but also indirectly, by altering nuclear transcriptional patterns. The current literature on such retrograde signalling in both yeast and mammalian cells is thus reviewed, with an outlook on its potential contribution to disease through the regulation of PGC-1 family coactivators. We propose that further investigation of these pathways will lead to the identification of novel pharmacological targets and treatment strategies to combat disease.
Collapse
Affiliation(s)
- Aleck W E Jones
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | | | | | | | | |
Collapse
|
35
|
Chiaradonna F, Moresco RM, Airoldi C, Gaglio D, Palorini R, Nicotra F, Messa C, Alberghina L. From cancer metabolism to new biomarkers and drug targets. Biotechnol Adv 2011; 30:30-51. [PMID: 21802503 DOI: 10.1016/j.biotechadv.2011.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 07/13/2011] [Indexed: 12/14/2022]
Abstract
Great interest is presently given to the analysis of metabolic changes that take place specifically in cancer cells. In this review we summarize the alterations in glycolysis, glutamine utilization, fatty acid synthesis and mitochondrial function that have been reported to occur in cancer cells and in human tumors. We then propose considering cancer as a system-level disease and argue how two hallmarks of cancer, enhanced cell proliferation and evasion from apoptosis, may be evaluated as system-level properties, and how this perspective is going to modify drug discovery. Given the relevance of the analysis of metabolism both for studies on the molecular basis of cancer cell phenotype and for clinical applications, the more relevant technologies for this purpose, from metabolome and metabolic flux analysis in cells by Nuclear Magnetic Resonance and Mass Spectrometry technologies to positron emission tomography on patients, are analyzed. The perspectives offered by specific changes in metabolism for a new drug discovery strategy for cancer are discussed and a survey of the industrial activity already going on in the field is reported.
Collapse
Affiliation(s)
- F Chiaradonna
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Gasparre G, Romeo G, Rugolo M, Porcelli AM. Learning from oncocytic tumors: Why choose inefficient mitochondria? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2011; 1807:633-42. [DOI: 10.1016/j.bbabio.2010.08.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 08/09/2010] [Accepted: 08/15/2010] [Indexed: 10/19/2022]
|
37
|
Sadikovic B, Wang J, El-Hattab AW, Landsverk M, Douglas G, Brundage EK, Craigen WJ, Schmitt ES, Wong LJC. Sequence homology at the breakpoint and clinical phenotype of mitochondrial DNA deletion syndromes. PLoS One 2010; 5:e15687. [PMID: 21187929 PMCID: PMC3004954 DOI: 10.1371/journal.pone.0015687] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 11/22/2010] [Indexed: 01/07/2023] Open
Abstract
Mitochondrial DNA (mtDNA) deletions are a common cause of mitochondrial disorders. Large mtDNA deletions can lead to a broad spectrum of clinical features with different age of onset, ranging from mild mitochondrial myopathies (MM), progressive external ophthalmoplegia (PEO), and Kearns-Sayre syndrome (KSS), to severe Pearson syndrome. The aim of this study is to investigate the molecular signatures surrounding the deletion breakpoints and their association with the clinical phenotype and age at onset. MtDNA deletions in 67 patients were characterized using array comparative genomic hybridization (aCGH) followed by PCR-sequencing of the deletion junctions. Sequence homology including both perfect and imperfect short repeats flanking the deletion regions were analyzed and correlated with clinical features and patients' age group. In all age groups, there was a significant increase in sequence homology flanking the deletion compared to mtDNA background. The youngest patient group (<6 years old) showed a diffused pattern of deletion distribution in size and locations, with a significantly lower sequence homology flanking the deletion, and the highest percentage of deletion mutant heteroplasmy. The older age groups showed rather discrete pattern of deletions with 44% of all patients over 6 years old carrying the most common 5 kb mtDNA deletion, which was found mostly in muscle specimens (22/41). Only 15% (3/20) of the young patients (<6 years old) carry the 5 kb common deletion, which is usually present in blood rather than muscle. This group of patients predominantly (16 out of 17) exhibit multisystem disorder and/or Pearson syndrome, while older patients had predominantly neuromuscular manifestations including KSS, PEO, and MM. In conclusion, sequence homology at the deletion flanking regions is a consistent feature of mtDNA deletions. Decreased levels of sequence homology and increased levels of deletion mutant heteroplasmy appear to correlate with earlier onset and more severe disease with multisystem involvement.
Collapse
Affiliation(s)
- Bekim Sadikovic
- Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Klomp JA, Petillo D, Niemi NM, Dykema KJ, Chen J, Yang XJ, Sääf A, Zickert P, Aly M, Bergerheim U, Nordenskjöld M, Gad S, Giraud S, Denoux Y, Yonneau L, Méjean A, Vasiliu V, Richard S, MacKeigan JP, Teh BT, Furge KA. Birt-Hogg-Dubé renal tumors are genetically distinct from other renal neoplasias and are associated with up-regulation of mitochondrial gene expression. BMC Med Genomics 2010; 3:59. [PMID: 21162720 PMCID: PMC3012009 DOI: 10.1186/1755-8794-3-59] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 12/16/2010] [Indexed: 12/24/2022] Open
Abstract
Background Germline mutations in the folliculin (FLCN) gene are associated with the development of Birt-Hogg-Dubé syndrome (BHDS), a disease characterized by papular skin lesions, a high occurrence of spontaneous pneumothorax, and the development of renal neoplasias. The majority of renal tumors that arise in BHDS-affected individuals are histologically similar to sporadic chromophobe renal cell carcinoma (RCC) and sporadic renal oncocytoma. However, most sporadic tumors lack FLCN mutations and the extent to which the BHDS-derived renal tumors share genetic defects associated with the sporadic tumors has not been well studied. Methods BHDS individuals were identified symptomatically and FLCN mutations were confirmed by DNA sequencing. Comparative gene expression profiling analyses were carried out on renal tumors isolated from individuals afflicted with BHDS and a panel of sporadic renal tumors of different subtypes using discriminate and clustering approaches. qRT-PCR was used to confirm selected results of the gene expression analyses. We further analyzed differentially expressed genes using gene set enrichment analysis and pathway analysis approaches. Pathway analysis results were confirmed by generation of independent pathway signatures and application to additional datasets. Results Renal tumors isolated from individuals with BHDS showed distinct gene expression and cytogenetic characteristics from sporadic renal oncocytoma and chromophobe RCC. The most prominent molecular feature of BHDS-derived kidney tumors was high expression of mitochondria-and oxidative phosphorylation (OXPHOS)-associated genes. This mitochondria expression phenotype was associated with deregulation of the PGC-1α-TFAM signaling axis. Loss of FLCN expression across various tumor types is also associated with increased nuclear mitochondrial gene expression. Conclusions Our results support a genetic distinction between BHDS-associated tumors and other renal neoplasias. In addition, deregulation of the PGC-1α-TFAM signaling axis is most pronounced in renal tumors that harbor FLCN mutations and in tumors from other organs that have relatively low expression of FLCN. These results are consistent with the recently discovered interaction between FLCN and AMPK and support a model in which FLCN is a regulator of mitochondrial function.
Collapse
Affiliation(s)
- Jeff A Klomp
- Laboratory of Computational Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Bayona-Bafaluy MP, López-Gallardo E, Montoya J, Ruiz-Pesini E. Maternally inherited susceptibility to cancer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2010; 1807:643-9. [PMID: 20732295 DOI: 10.1016/j.bbabio.2010.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 08/13/2010] [Accepted: 08/15/2010] [Indexed: 12/16/2022]
Abstract
Tumor microenvironment promotes mtDNA mutations. A number of these mutations will affect cell metabolism and increase cell survival. These mutations are positively selected and contribute to other tumor features, such as extracellular matrix remodeling and angiogenic processes, thus favoring metastases. Like somatic mutations, although with less marked effects, some mtDNA population polymorphisms will affect OXPHOS function, cell metabolism, and homeostasis. Thus, they could behave as inherited susceptibility factors for cancer. However, in addition to epidemiological evidence, other more direct clues are required. The cybrid approach can help to clarify this issue.
Collapse
Affiliation(s)
- María Pilar Bayona-Bafaluy
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain
| | | | | | | |
Collapse
|
40
|
Chang JC, Kou SJ, Lin WT, Liu CS. Regulatory role of mitochondria in oxidative stress and atherosclerosis. World J Cardiol 2010; 2:150-9. [PMID: 21160733 PMCID: PMC2999054 DOI: 10.4330/wjc.v2.i6.150] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 06/07/2010] [Accepted: 06/14/2010] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial physiology and biogenesis play a crucial role in the initiation and progression of cardiovascular disease following oxidative stress-induced damage such as atherosclerosis (AST). Dysfunctional mitochondria caused by an increase in mitochondrial reactive oxygen species (ROS) production, accumulation of mitochondrial DNA damage, and respiratory chain deficiency induces death of endothelial/smooth muscle cells and favors plaque formation/rupture via the regulation of mitochondrial biogenesis-related genes such as peroxisome proliferator-activated receptor γ coactivator (PGC-1), although more detailed mechanisms still need further study. Based on the effect of healthy mitochondria produced by mitochondrial biogenesis on decreasing ROS-mediated cell death and the recent finding that the regulation of PGC-1 involves mitochondrial fusion-related protein (mitofusin), we thus infer the regulatory role of mitochondrial fusion/fission balance in AST pathophysiology. In this review, the first section discusses the possible association between AST-inducing factors and the molecular regulatory mechanisms of mitochondrial biogenesis and dynamics, and explains the role of mitochondria-dependent regulation in cell apoptosis during AST development. Furthermore, nitric oxide has the Janus-faced effect by protecting vascular damage caused by AST while being a reactive nitrogen species (RNS) which act together with ROS to damage cells. Therefore, in the second section we discuss mitochondrial ATP-sensitive K(+) channels, which regulate mitochondrial ion transport to maintain mitochondrial physiology, involved in the regulation of ROS/RNS production and their influence on AST/cardiovascular diseases (CVD). Through this review, we can further appreciate the multi-regulatory functions of the mitochondria involved in AST development. The understanding of these related mechanisms will benefit drug development in treating AST/CVD through targeted biofunctions of mitochondria.
Collapse
Affiliation(s)
- Jui-Chih Chang
- Jui-Chih Chang, Wei-Ting Lin, Chin-San Liu, Department of Neurology, Vascular and Genomic Center, Changhua Christian Hospital, Changhua 50094, Taiwan, China
| | | | | | | |
Collapse
|
41
|
Wallace DC, Fan W, Procaccio V. Mitochondrial energetics and therapeutics. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2010; 5:297-348. [PMID: 20078222 DOI: 10.1146/annurev.pathol.4.110807.092314] [Citation(s) in RCA: 506] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mitochondrial dysfunction has been linked to a wide range of degenerative and metabolic diseases, cancer, and aging. All these clinical manifestations arise from the central role of bioenergetics in cell biology. Although genetic therapies are maturing as the rules of bioenergetic genetics are clarified, metabolic therapies have been ineffectual. This failure results from our limited appreciation of the role of bioenergetics as the interface between the environment and the cell. A systems approach, which, ironically, was first successfully applied over 80 years ago with the introduction of the ketogenic diet, is required. Analysis of the many ways that a shift from carbohydrate glycolytic metabolism to fatty acid and ketone oxidative metabolism may modulate metabolism, signal transduction pathways, and the epigenome gives us an appreciation of the ketogenic diet and the potential for bioenergetic therapeutics.
Collapse
Affiliation(s)
- Douglas C Wallace
- Center for Molecular and Mitochondrial Medicine and Genetics and Departments of Biological Chemistry, Ecology and Evolutionary Biology, and Pediatrics, University of California at Irvine, Irvine, California 92697-3940, USA.
| | | | | |
Collapse
|
42
|
Cormio A, Guerra F, Cormio G, Pesce V, Fracasso F, Loizzi V, Cantatore P, Selvaggi L, Gadaleta MN. The PGC-1α-dependent pathway of mitochondrial biogenesis is upregulated in type I endometrial cancer. Biochem Biophys Res Commun 2009; 390:1182-5. [DOI: 10.1016/j.bbrc.2009.10.114] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Accepted: 10/22/2009] [Indexed: 12/22/2022]
|
43
|
Soto IC, Fontanesi F, Valledor M, Horn D, Singh R, Barrientos A. Synthesis of cytochrome c oxidase subunit 1 is translationally downregulated in the absence of functional F1F0-ATP synthase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1776-86. [PMID: 19735676 DOI: 10.1016/j.bbamcr.2009.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 08/31/2009] [Accepted: 09/01/2009] [Indexed: 10/20/2022]
Abstract
The mitochondrial F(1)F(0)-ATP synthase or ATPase is a key enzyme for aerobic energy production in eukaryotic cells. Mutations in ATPase structural and assembly genes are the primary cause of severe human encephalomyopathies, frequently associated with a pleiotropic decrease in cytochrome c oxidase (COX) activity. We have studied the structural and functional constraints underlying the COX defect using Saccharomyces cerevisiae genetic and pharmacological models of ATPase deficiency. In both yeast Deltaatp10 and oligomycin-treated wild type cells, COX assembly is selectively impaired in the absence of functional ATPase. The COX biogenesis defect does not involve a primary alteration in the expression of the COX subunits as previously suggested but in their maturation and/or assembly. Expression of COX subunit 1, however, is translationally regulated as in most bona fide COX assembly mutants. Additionally, the COX defect in oligomycin-inhibited ATPase-deficient yeast cells, but not in atp10 cells could be partially prevented by partially dissipating the mitochondrial membrane potential using the uncoupler CCCP. Similar results were obtained with oligomycin-treated and ATP12-deficient human fibroblasts respectively. Our findings imply that fully assembled ATPase and its proton pumping function are both required for COX biogenesis in yeast and mammalian cells through a mechanism independent of Cox1p synthesis.
Collapse
Affiliation(s)
- Ileana C Soto
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine. Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
44
|
Wareski P, Vaarmann A, Choubey V, Safiulina D, Liiv J, Kuum M, Kaasik A. PGC-1{alpha} and PGC-1{beta} regulate mitochondrial density in neurons. J Biol Chem 2009; 284:21379-85. [PMID: 19542216 DOI: 10.1074/jbc.m109.018911] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recent studies indicate that regulation of cellular oxidative capacity through enhancing mitochondrial biogenesis may be beneficial for neuronal recovery and survival in human neurodegenerative disorders. The peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) has been shown to be a master regulator of mitochondrial biogenesis and cellular energy metabolism in muscle and liver. The aim of our study was to establish whether PGC-1alpha and PGC-1beta control mitochondrial density also in neurons and if these coactivators could be up-regulated by deacetylation. The results demonstrate that PGC-1alpha and PGC-1beta control mitochondrial capacity in an additive and independent manner. This effect was observed in all studied subtypes of neurons, in cortical, midbrain, and cerebellar granule neurons. We also observed that endogenous neuronal PGC-1alpha but not PGC-1beta could be activated through its repressor domain by suppressing it. Results demonstrate also that overexpression of SIRT1 deacetylase or suppression of GCN5 acetyltransferase activates transcriptional activity of PGC-1alpha in neurons and increases mitochondrial density. These effects were mediated exclusively via PGC-1alpha, since overexpression of SIRT1 or suppression of GCN5 was ineffective where PGC-1alpha was suppressed by short hairpin RNA. Moreover, the results demonstrate that overexpression of PGC-1beta or PGC-1alpha or activation of the latter by SIRT1 protected neurons from mutant alpha-synuclein- or mutant huntingtin-induced mitochondrial loss. These evidences demonstrate that activation or overexpression of the PGC-1 family of coactivators could be used to compensate for neuronal mitochondrial loss and suggest that therapeutic agents activating PGC-1 would be valuable for treating neurodegenerative diseases in which mitochondrial dysfunction and oxidative damage play an important pathogenic role.
Collapse
Affiliation(s)
- Przemyslaw Wareski
- Department of Pharmacology, University of Tartu, Ravila 19, 51014 Tartu, Estonia
| | | | | | | | | | | | | |
Collapse
|
45
|
Srivastava S, Diaz F, Iommarini L, Aure K, Lombes A, Moraes CT. PGC-1alpha/beta induced expression partially compensates for respiratory chain defects in cells from patients with mitochondrial disorders. Hum Mol Genet 2009; 18:1805-12. [PMID: 19297390 DOI: 10.1093/hmg/ddp093] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Members of the peroxisome proliferator-activated receptor gamma coactivator (PGC) family are potent inducers of mitochondrial biogenesis. We have tested the potential effect of increased mitochondrial biogenesis in cells derived from patients harboring oxidative phosphorylation defects due to either nuclear or mitochondrial DNA mutations. We found that the PGC-1alpha and/or PGC-1beta expression improved mitochondrial respiration in cells harboring a complex III or IV deficiency as well as in transmitochondrial cybrids harboring mitochondrial encephalomyopathy lactic acidosis and stroke A3243G tRNA((Leu)UUR) gene mutation. The respiratory function improvement was found to be associated with increased levels of mitochondrial components per cell, although this increase was not homogeneous. These results reinforce the concept that increased mitochondrial biogenesis is a promising venue for the treatment of mitochondrial diseases.
Collapse
Affiliation(s)
- Sarika Srivastava
- Department of Neurology, University of Miami School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
46
|
Gasparre G, Iommarini L, Porcelli AM, Lang M, Ferri GG, Kurelac I, Zuntini R, Mariani E, Pennisi LF, Pasquini E, Pasquinelli G, Ghelli A, Bonora E, Ceccarelli C, Rugolo M, Salfi N, Romeo G, Carelli V. An inherited mitochondrial DNA disruptive mutation shifts to homoplasmy in oncocytic tumor cells. Hum Mutat 2009; 30:391-6. [DOI: 10.1002/humu.20870] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Dieteren CEJ, Koopman WJH, Nijtmans LGJ. Chapter 7 Tracing human mitochondrial complex I assembly by use of GFP-tagged subunits. Methods Enzymol 2009; 456:133-51. [PMID: 19348886 DOI: 10.1016/s0076-6879(08)04407-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Disturbances in the assembly of mitochondrial complex I (CI) are a frequent cause of mitochondrial disorders. Several lines of evidence hint at a semi-sequential assembly pathway, in which the 45 individual subunits that form the holoenzyme are pieced together by means of smaller intermediates. To understand this process, it is necessary to explain the exact order, the rate-limiting steps, and the dynamics of subunit incorporation. In this chapter, we describe an approach to regulate the expression levels of an AcGFP(1)-tagged subunit (NDUFS3) in mammalian cells by means of a tetracycline-inducible promoter. This strategy allows the study of the dynamics of CI assembly intermediates in living cells on native gels. After establishing that the AcGFP(1) tag does not interfere with the activity and assembly of the enzyme, we show how this system can be used to trace the labeled subunit in an induction pulse-chase experiment or to study its accumulation in specific assembly intermediates after inhibition of mitochondrial translation.
Collapse
Affiliation(s)
- Cindy E J Dieteren
- Department of Paediatrics, Nijmegen Centre for Mitochondrial Disorders, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | |
Collapse
|
48
|
Nolte F, Hofmann WK. Myelodysplastic syndromes: molecular pathogenesis and genomic changes. Ann Hematol 2008; 87:777-95. [PMID: 18516602 DOI: 10.1007/s00277-008-0502-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Accepted: 04/15/2008] [Indexed: 01/27/2023]
Abstract
Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis presenting with peripheral cytopenias in combination with a hyperplastic bone marrow and an increased risk of evolution to acute myeloid leukemia. The classification systems such as the WHO classification mainly rely on morphological criteria and are supplemented by the International Prognostic Scoring System which takes cytogenetical changes into consideration when determining the prognosis of MDS but wide intra-subtype variations do exist. The pathomechanisms causing primary MDS require further work. Development and progression of MDS is suggested to be a multistep alteration to hematopoietic stem cells. Different molecular alterations have been described, affecting genes involved in cell-cycle control, mitotic checkpoints, and growth factor receptors. Secondary signal proteins and transcription factors, which gives the cell a growth advantage over its normal counterpart, may be affected as well. The accumulation of such defects may finally cause the leukemic transformation of MDS.
Collapse
Affiliation(s)
- Florian Nolte
- Department of Hematology and Oncology, University Hospital Benjamin Franklin, Charité, Hindenburgdamm 30, 12203, Berlin, Germany.
| | | |
Collapse
|
49
|
Barrientos A, Gouget K, Horn D, Soto IC, Fontanesi F. Suppression mechanisms of COX assembly defects in yeast and human: insights into the COX assembly process. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:97-107. [PMID: 18522805 DOI: 10.1016/j.bbamcr.2008.05.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 04/29/2008] [Accepted: 05/05/2008] [Indexed: 12/11/2022]
Abstract
Eukaryotic cytochrome c oxidase (COX) is the terminal enzyme of the mitochondrial respiratory chain. COX is a multimeric enzyme formed by subunits of dual genetic origin whose assembly is intricate and highly regulated. In addition to the structural subunits, a large number of accessory factors are required to build the holoenzyme. The function of these factors is required in all stages of the assembly process. They are relevant to human health because devastating human disorders have been associated with mutations in nuclear genes encoding conserved COX assembly factors. The study of yeast strains and human cell lines from patients carrying mutations in structural subunits and COX assembly factors has been invaluable to attain the current state of knowledge, even if still fragmentary, of the COX assembly process. After the identification of the genes involved, the isolation and characterization of genetic and metabolic suppressors of COX assembly defects, reviewed here, have become a profitable strategy to gain insight into their functions and the pathways in which they operate. Additionally, they have the potential to provide useful information for devising therapeutic approaches to combat human disorders associated with COX deficiency.
Collapse
Affiliation(s)
- Antoni Barrientos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | | | | | | | | |
Collapse
|
50
|
Diaz F, Moraes CT. Mitochondrial biogenesis and turnover. Cell Calcium 2008; 44:24-35. [PMID: 18395251 DOI: 10.1016/j.ceca.2007.12.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2007] [Revised: 12/10/2007] [Accepted: 12/12/2007] [Indexed: 11/17/2022]
Abstract
Mitochondrial biogenesis is a complex process involving the coordinated expression of mitochondrial and nuclear genes, the import of the products of the latter into the organelle and turnover. The mechanisms associated with these events have been intensively studied in the last 20 years and our understanding of their details is much improved. Mitochondrial biogenesis requires the participation of calcium signaling that activates a series of calcium-dependent protein kinases that in turn activate transcription factors and coactivators such as PGC-1alpha that regulates the expression of genes coding for mitochondrial components. In addition, mitochondrial biogenesis involves the balance of mitochondrial fission-fusion. Mitochondrial malfunction or defects in any of the many pathways involved in mitochondrial biogenesis can lead to degenerative diseases and possibly play an important part in aging.
Collapse
Affiliation(s)
- Francisca Diaz
- Department of Neurology, University of Miami, Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL 33136, USA.
| | | |
Collapse
|