1
|
Zhang F, Zhang L, Hu G, Chen X, Liu H, Li C, Guo X, Huang C, Sun F, Li T, Cui Z, Guo Y, Yan W, Xia Y, Liu Z, Lin Z, Duan W, Lu L, Wang X, Wang Z, Wang S, Tao L. Rectifying METTL4-Mediated N 6-Methyladenine Excess in Mitochondrial DNA Alleviates Heart Failure. Circulation 2024; 150:1441-1458. [PMID: 38686562 DOI: 10.1161/circulationaha.123.068358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/08/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Myocardial mitochondrial dysfunction underpins the pathogenesis of heart failure (HF), yet therapeutic options to restore myocardial mitochondrial function are scarce. Epigenetic modifications of mitochondrial DNA (mtDNA), such as methylation, play a pivotal role in modulating mitochondrial homeostasis. However, their involvement in HF remains unclear. METHODS Experimental HF models were established through continuous angiotensin II and phenylephrine (AngII/PE) infusion or prolonged myocardial ischemia/reperfusion injury. The landscape of N6-methyladenine (6mA) methylation within failing cardiomyocyte mtDNA was characterized using high-resolution mass spectrometry and methylated DNA immunoprecipitation sequencing. A tamoxifen-inducible cardiomyocyte-specific Mettl4 knockout mouse model and adeno-associated virus vectors designed for cardiomyocyte-targeted manipulation of METTL4 (methyltransferase-like protein 4) expression were used to ascertain the role of mtDNA 6mA and its methyltransferase METTL4 in HF. RESULTS METTL4 was predominantly localized within adult cardiomyocyte mitochondria. 6mA modifications were significantly more abundant in mtDNA than in nuclear DNA. Postnatal cardiomyocyte maturation presented with a reduction in 6mA levels within mtDNA, coinciding with a decrease in METTL4 expression. However, an increase in both mtDNA 6mA level and METTL4 expression was observed in failing adult cardiomyocytes, suggesting a shift toward a neonatal-like state. METTL4 preferentially targeted mtDNA promoter regions, which resulted in interference with transcription initiation complex assembly, mtDNA transcriptional stalling, and ultimately mitochondrial dysfunction. Amplifying cardiomyocyte mtDNA 6mA through METTL4 overexpression led to spontaneous mitochondrial dysfunction and HF phenotypes. The transcription factor p53 was identified as a direct regulator of METTL4 transcription in response to HF-provoking stress, thereby revealing a stress-responsive mechanism that controls METTL4 expression and mtDNA 6mA. Cardiomyocyte-specific deletion of the Mettl4 gene eliminated mtDNA 6mA excess, preserved mitochondrial function, and mitigated the development of HF upon continuous infusion of AngII/PE. In addition, specific silencing of METTL4 in cardiomyocytes restored mitochondrial function and offered therapeutic relief in mice with preexisting HF, irrespective of whether the condition was induced by AngII/PE infusion or myocardial ischemia/reperfusion injury. CONCLUSIONS Our findings identify a pivotal role of cardiomyocyte mtDNA 6mA and the corresponding methyltransferase, METTL4, in the pathogenesis of mitochondrial dysfunction and HF. Targeted suppression of METTL4 to rectify mtDNA 6mA excess emerges as a promising strategy for developing mitochondria-focused HF interventions.
Collapse
Affiliation(s)
- Fuyang Zhang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Zhang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guangyu Hu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiyao Chen
- Geriatrics (X.C.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Liu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Congye Li
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiong Guo
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chong Huang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fangfang Sun
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tongzheng Li
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhe Cui
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yongzhen Guo
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenjun Yan
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yunlong Xia
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhiyuan Liu
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen Lin
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weixun Duan
- Cardiovascular Surgery (W.D., L.L.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Linhe Lu
- Cardiovascular Surgery (W.D., L.L.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinyi Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhengyang Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shan Wang
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Tao
- Departments of Cardiology (F.Z., L.Z., G.H., H.L., C.L., X.G., C.H., F.S., T.L., Z.C., Y.G., W.Y., Y.X., Z. Liu, Z. Lin, X.W., Z.W., S.W., L.T.), Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
2
|
Bhattacharya J, Nitnavare RB, Bhatnagar-Mathur P, Reddy PS. Cytoplasmic male sterility-based hybrids: mechanistic insights. PLANTA 2024; 260:100. [PMID: 39302508 DOI: 10.1007/s00425-024-04532-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
MAIN CONCLUSION A comprehensive understanding of the nucleocytoplasmic interactions that occur between genes related to the restoration of fertility and cytoplasmic male sterility (CMS) provides insight into the development of hybrids of important crop species. Modern biotechnological techniques allow this to be achieved in an efficient and quick manner. Heterosis is paramount for increasing the yield and quality of a crop. The development of hybrids for achieving heterosis has been well-studied and proven to be robust and efficient. Cytoplasmic male sterility (CMS) has been explored extensively in the production of hybrids. The underlying mechanisms of CMS include the role of cytotoxic proteins, PCD of tapetal cells, and improper RNA editing of restoration factors. On the other hand, the restoration of fertility is caused by the presence of restorer-of-fertility (Rf) genes or restorer genes, which inhibit the effects of sterility-causing genes. The interaction between mitochondria and the nuclear genome is crucial for several regulatory pathways, as observed in the CMS-Rf system and occurs at the genomic, transcriptional, post-transcriptional, translational, and post-translational levels. These CMS-Rf mechanisms have been validated in several crop systems. This review aims to summarize the nucleo-mitochondrial interaction mechanism of the CMS-Rf system. It also sheds light on biotechnological interventions, such as genetic engineering and genome editing, to achieve CMS-based hybrids.
Collapse
Affiliation(s)
- Joorie Bhattacharya
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru, Hyderabad, Telangana, 502324, India
- Department of Genetics, Osmania University, Hyderabad, Telangana, 500007, India
| | - Rahul B Nitnavare
- Division of Plant and Crop Sciences, School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire, Nottingham, LE12 5RD, UK
| | - Pooja Bhatnagar-Mathur
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru, Hyderabad, Telangana, 502324, India.
- Plant Breeding & Genetics Laboratory of United Nation, International Atomic Energy Agency, 1400, Vienna, Austria.
| | - Palakolanu Sudhakar Reddy
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Patancheru, Hyderabad, Telangana, 502324, India.
| |
Collapse
|
3
|
Aslamkhan AG, Michna L, Podtelezhnikov A, Vlasakova K, Suemizu H, Ohnishi Y, Liu L, Lane P, Xu Q, Kuhls MC, Wang Z, Pacchione S, Erdos Z, Tracy RW, Koeplinger K, Muniappa N, Valentine J, Galijatovic-Idrizbegovic A, Glaab WE, Sistare FD, Lebron J. A mechanistic biomarker investigation of fialuridine hepatotoxicity using the chimeric TK-NOG Hu-liver mouse model and in vitro micropatterned hepatocyte cocultures. Toxicol Res (Camb) 2024; 13:tfad120. [PMID: 38223529 PMCID: PMC10784659 DOI: 10.1093/toxres/tfad120] [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: 05/31/2023] [Revised: 10/20/2023] [Accepted: 12/01/2023] [Indexed: 01/16/2024] Open
Abstract
Fialuridine (FIAU) is a nucleoside-based drug that caused liver failure and deaths in a human clinical trial that were not predicted by nonclinical safety studies. A recent report concluded that a TK-NOG humanized liver (hu-liver) mouse model detected human-specific FIAU liver toxicity, and broader use of that model could improve drug safety testing. We further evaluated this model at similar dose levels to assess FIAU sensitivity and potential mechanistic biomarkers. Although we were unable to reproduce the marked acute liver toxicity with two separate studies (including one with a "sensitized" donor), we identified molecular biomarkers reflecting the early stages of FIAU mitochondrial toxicity, which were not seen with its stereoisomer (FIRU). Dose dependent FIAU-induced changes in hu-liver mice included more pronounced reductions in mitochondrial to nuclear DNA (mtDNA/nucDNA) ratios in human hepatocytes compared to mouse hepatocytes and kidneys of the same animals. FIAU treatment also triggered a p53 transcriptional response and opposing changes in transcripts of nuclear- and mitochondrial-encoded mitochondrial proteins. The time dependent accumulation of FIAU into mtDNA is consistent with the ≥9-week latency of liver toxicity observed for FIAU in the clinic. Similar changes were observed in an in vitro micro-patterned hepatocyte coculture system. In addition, FIAU-dependent mtDNA/nucDNA ratio and transcriptional alterations, especially reductions in mitochondrially encoded transcripts, were seen in livers of non-engrafted TK-NOG and CD-1 mice dosed for a shorter period. Conclusion: These mechanistic biomarker findings can be leveraged in an in vitro model and in a more routine preclinical model (CD-1 mice) to identify nucleosides with such a FIAU-like mitochondrial toxicity mechanistic liability potential. Further optimization of the TK-NOG hu-liver mouse model is necessary before broader adoption for drug safety testing.
Collapse
Affiliation(s)
- Amy G Aslamkhan
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Laura Michna
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Alexei Podtelezhnikov
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Katerina Vlasakova
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Hiroshi Suemizu
- Laboratory Animal Research, Central Institute for Experimental Animals, 210-0821 Kawasaki-ku, Kawasaki 3-25-12 Tonomachi, Japan
| | - Yasuyuki Ohnishi
- Laboratory Animal Research, Central Institute for Experimental Animals, 210-0821 Kawasaki-ku, Kawasaki 3-25-12 Tonomachi, Japan
| | - Liping Liu
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Pamela Lane
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Qiuwei Xu
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Matthew C Kuhls
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Zhibin Wang
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Stephen Pacchione
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Zoltan Erdos
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Rodger William Tracy
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, United States
| | - Kenneth Koeplinger
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, United States
| | - Nagaraja Muniappa
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - John Valentine
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA, United States
| | | | - Warren E Glaab
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Frank D Sistare
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| | - Jose Lebron
- Nonclinical Drug Safety, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, United States
| |
Collapse
|
4
|
Nagarajan‐Radha V, Cordina N, Beekman M. Diet and mitonuclear haplotype interactions affect growth rate in a slime mould. Ecol Evol 2023; 13:e10508. [PMID: 37674651 PMCID: PMC10477482 DOI: 10.1002/ece3.10508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
Trait expression in metazoans is strongly influenced by the balance of macronutrients (i.e. protein, carbohydrate and fat) in the diet. At the same time, an individual's genetic background seems to regulate the magnitude of phenotypic response to a particular diet. It needs to be better understood whether interactions between diet, genetic background and trait expression are found in unicellular eukaryotes. A protist-the slime mould, Physarum polycephalum can choose diets based on protein-to-carbohydrate (P:C) content to support optimal growth rate. Yet, the role of genetic background (variation in the mitochondrial and nuclear DNAs) in mediating growth rate response to dietary P:C ratios in the slime mould is unknown. Here, we studied the effects of interactions between mitochondrial and nuclear DNA haplotypes and diet (i.e. G × G × E interactions) on the growth rate of P. polycephalum. A genetic panel of six distinct strains of P. polycephalum that differ in their mitochondrial and nuclear DNA haplotypes was used to measure growth rate across five diets that varied in their P:C ratio and total calories. We first determined the strains' growth rate (total biomass and surface area) when grown on a set menu with access to a particular diet. We then assessed whether the growth rate of strains increased on a buffet menu with access to all diets. Our findings show that the growth rate of P. polycephalum is generally higher on diets containing more carbohydrates than protein and that total calories negatively affect the growth rate. Three-way interactions between mitochondrial, nuclear haplotypes and dietary P:C ratios affected the strains' surface area of growth but not biomass. Intriguingly, strains did not increase their surface area and biomass when they had access to all diets on the buffet menu. Our findings have broad implications for our understanding of the effect of mitonuclear interactions on trait expression across diverse eukaryotic lineages.
Collapse
Affiliation(s)
- Venkatesh Nagarajan‐Radha
- Behaviour, Ecology and Evolution Lab, School of Life and Environmental SciencesThe University of SydneyCamperdownNew South WalesAustralia
| | - Natalie Cordina
- Behaviour, Ecology and Evolution Lab, School of Life and Environmental SciencesThe University of SydneyCamperdownNew South WalesAustralia
| | - Madeleine Beekman
- Behaviour, Ecology and Evolution Lab, School of Life and Environmental SciencesThe University of SydneyCamperdownNew South WalesAustralia
| |
Collapse
|
5
|
Mitochondrial DNA Polymorphism in HV1 and HV2 Regions and 12S rDNA in Perimenopausal Hypertensive Women. Biomedicines 2023; 11:biomedicines11030823. [PMID: 36979802 PMCID: PMC10044999 DOI: 10.3390/biomedicines11030823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Estrogens enhance cellular mitochondrial activity. The diminution of female hormones during menopause may have an effect on the mitochondrial genome and the expression of mitochondrial proteins. Hence, oxidative stress and the pro-inflammatory state contribute to the formation of systemic illnesses including arterial hypertension (AH). This study aimed to determine the types and frequency of mutations in the mitochondrial DNA (mtDNA) nucleotide sequence in the hypervariable regions 1 and 2 (HV1 and HV2) and the 12S RNA coding sequence of the D-loop in postmenopausal women with hypertension. In our study, 100 women were investigated, 53 of whom were postmenopausal and 47 of whom were premenopausal (53.9 ± 3.7 years vs. 47.7 ± 4.2 years, respectively). Of those studied, 35 premenopausal and 40 postmenopausal women were diagnosed with AH. A medical checkup with 24 h monitoring of blood pressure (RR) and heart rate was undertaken (HR). The polymorphism of the D-loop and 12S rDNA region of mtDNA was examined. Changes in the nucleotide sequence of mtDNA were observed in 23% of the group of 100 women. The changes were identified in 91.3% of HV1 and HV2 regions, 60.9% of HV1 segments, 47.5% of HV2 regions, and 43.5% of 12S rDNA regions. The frequency of nucleotide sequence alterations in mtDNA was substantially higher in postmenopausal women (34%) than in premenopausal women (10.6%), p = 0.016. A higher frequency of changes in HV1 + HV2 sections in postmenopausal women (30.2%) compared to the premenopausal group (10.6%) was detected, p = 0.011. Only postmenopausal women were found to have modifications to the HV2 segment and the 12S rDNA region. After menopause, polymorphism in the mtDNA region was substantially more frequent in women with arterial hypertension than before menopause (p = 0.030; 37.5% vs. 11.5%). Comparable findings were observed in the HV2 and HV1 regions of the AH group (35% vs. 11.5%), p = 0.015, in the HV1 segment (25% vs. 11.5%), p = 0.529, and in the HV2 segment, 12S rDNA (25% vs. 0%). More than 80% of all changes in nucleotide sequence were homoplasmic. The mtDNA polymorphisms of the nucleotide sequence in the HV1 and HV2 regions, the HV2 region alone, and the 12S RNA coding sequence were associated with estrogen deficiency and a more severe course of arterial hypertension, accompanied by symptoms of adrenergic stimulation.
Collapse
|
6
|
Chen J, Li R, Knapp S, Zhu G, Whitener RL, Leiter EH, Mathews CE. Intergenomic and epistatic interactions control free radical mediated pancreatic β-cell damage. Front Genet 2022; 13:994501. [PMID: 36276935 PMCID: PMC9585181 DOI: 10.3389/fgene.2022.994501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Alloxan (AL)-generated Reactive Oxygen Species (ROS) selectively destroy insulin-producing pancreatic β-cells. A previous genome-wide scan (GWS) using a cohort of 296 F2 hybrids between NOD (AL-sensitive) and ALR (AL-resistant) mice identified linkages contributing to β-cell susceptibility or resistance to AL-induced diabetes on Chromosomes (Chr) 2, 3, 8, and a single nucleotide polymorphism in mt-Nd2 of the mitochondrial genome (mtDNA). AL treatment of congenic and consomic NOD mouse stocks confirmed resistance linked to both the mtDNA and the Chr 8 locus from ALR [NOD.mtALR.ALR-(D8Mit293-D8Mit137)]. To identify possible epistatic interactions, the GWS analysis was expanded to 678 F2 mice. ALR-derived diabetes-resistance linkages on Chr 8 as well as the mt-Nd2a allele were confirmed and novel additional linkages on Chr 4, 5, 6, 7, and 13 were identified. Epistasis was observed between the linkages on Chr 8 and 2 and Chr 8 and 6. Furthermore, the mt-Nd2 genotype affected the epistatic interactions between Chr 8 and 2. These results demonstrate that a combination of nuclear-cytoplasmic genome interactions regulates β-cell sensitivity to ROS-mediated ALD.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Renhua Li
- Henry M Jackson Foundation for the Advancement of Military Medicine (HJF), Bethesda, MD, United States
| | - Sarah Knapp
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Guizhi Zhu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Robert L. Whitener
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | | | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- *Correspondence: Clayton E. Mathews,
| |
Collapse
|
7
|
Ogawa S, Darhan H, Suzuki K. Genetic and genomic analysis of oxygen consumption in mice. J Anim Breed Genet 2022; 139:596-610. [PMID: 35608337 DOI: 10.1111/jbg.12721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/07/2022] [Indexed: 12/16/2022]
Abstract
We estimated genetic parameters for oxygen consumption (OC), OC per metabolic body weight (OCMBW) and body weight at three through 8 weeks of age in divergently selected mice populations, with an animal model considering maternal genetic, common litter environmental and cytoplasmic inheritance effects. Cytoplasmic inheritance was considered based on maternal lineage information. With respect to OC, estimated direct heritability was moderate (0.32) and the estimated proportion of the variance of cytoplasmic inheritance effects to the phenotypic variance was very low (0.01), implying that causal genes for OC could be located on autosomes. To assess this hypothesis, we attempted to identify possible candidate causal genes through selective signature detection with the results of pooled whole-genome resequencing using pooled DNA samples from high and low OC mice. We made a list of possible candidate causal genes for OC, including those relating to electron transport chain and ATP-binding proteins (Ndufa12, Sdhc, Atp10b, etc.), Prr16 encoding Largen protein, Cry1 encoding a key component of the circadian core oscillator and so on. The results, although careful interpretation must be required, could contribute to elucidate the genetic mechanism of OC, an indicator for maintenance energy requirement, and therefore feed efficiency.
Collapse
Affiliation(s)
- Shinichiro Ogawa
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Hongyu Darhan
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Keiichi Suzuki
- Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
8
|
Mitochondrial Function Differences between Tumor Tissue of Human Metastatic and Premetastatic CRC. BIOLOGY 2022; 11:biology11020293. [PMID: 35205159 PMCID: PMC8869310 DOI: 10.3390/biology11020293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/28/2022] [Accepted: 02/09/2022] [Indexed: 12/25/2022]
Abstract
Simple Summary Metastasis is an important cause of death from colorectal cancer (CRC). Mitochondria, which are important organelles of cells, play a key role in the metastatic transformation of cancer cells. We aimed to evaluate the adaptations associated with mitochondrial function in tumor tissues from advanced stages of human CRC and whether they could ultimately be used as a therapeutic target in metastatic CRC. We have compared the mitochondrial functionality parameters in tumor tissue samples and the normal adjacent tissue of advanced CRC patients with no radio- or chemotherapy treatment before surgery. Notable differences in mitochondrial functionality were detected between the samples of adjacent tissue versus tumor tissue from metastatic CRC patients. These findings suggest a shift in the mitochondrial function profile occurring in tumor tissue once the metastatic stage has been reached. These changes contribute to promote and maintain the metastatic phenotype, with evidence of mitochondrial function impairment in tumor tissue in the metastatic stage samples. Abstract Most colorectal cancer (CRC) patients die as a consequence of metastasis. Mitochondrial dysfunction could enhance cancer development and metastatic progression. We aimed to evaluate the adaptations associated with mitochondrial function in tumor tissues from stages III and IV of human CRC and whether they could ultimately be used as a therapeutic target in metastatic colorectal cancer (mCRC). We analyzed the protein levels by Western blotting and the enzymatic activities of proteins involved in mitochondrial function, as well as the amount of mitochondrial DNA (mtDNA), by real-time PCR, analyzing samples of non-tumor adjacent tissue and tumor tissue from stages III and IV CRC patients without radio- or chemotherapy treatment prior to surgery. Our data indicate that the tumor tissue of pre-metastatic stage III CRC exhibited an oxidant metabolic profile very similar to the samples of non-tumor adjacent tissue of both stages. Notable differences in the protein expression levels of ATPase, IDH2, LDHA, and SIRT1, as well as mtDNA amount, were detected between the samples of non-tumor adjacent tissue and tumor tissue from metastatic CRC patients. These findings suggest a shift in the oxidative metabolic profile that takes place in the tumor tissue once the metastatic stage has been reached. Tumor tissue oxidative metabolism contributes to promote and maintain the metastatic phenotype, with evidence of mitochondrial function impairment in stage IV tumor tissue.
Collapse
|
9
|
McCarthy M, Raval AP. The peri-menopause in a woman's life: a systemic inflammatory phase that enables later neurodegenerative disease. J Neuroinflammation 2020; 17:317. [PMID: 33097048 PMCID: PMC7585188 DOI: 10.1186/s12974-020-01998-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023] Open
Abstract
The peri-menopause or menopausal transition—the time period that surrounds the final years of a woman’s reproductive life—is associated with profound reproductive and hormonal changes in a woman’s body and exponentially increases a woman’s risk of cerebral ischemia and Alzheimer’s disease. Although our understanding of the exact timeline or definition of peri-menopause is limited, it is clear that there are two stages to the peri-menopause. These are the early menopausal transition, where menstrual cycles are mostly regular, with relatively few interruptions, and the late transition, where amenorrhea becomes more prolonged and lasts for at least 60 days, up to the final menstrual period. Emerging evidence is showing that peri-menopause is pro-inflammatory and disrupts estrogen-regulated neurological systems. Estrogen is a master regulator that functions through a network of estrogen receptors subtypes alpha (ER-α) and beta (ER-β). Estrogen receptor-beta has been shown to regulate a key component of the innate immune response known as the inflammasome, and it also is involved in regulation of neuronal mitochondrial function. This review will present an overview of the menopausal transition as an inflammatory event, with associated systemic and central nervous system inflammation, plus regulation of the innate immune response by ER-β-mediated mechanisms.
Collapse
Affiliation(s)
- Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, 1420 NW 9th Avenue, Neurology Research Building, Room # 203H, Miami, FL, 33136, USA. .,Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
10
|
Sharma P, Bharat, Dogra N, Singh S. Small Regulatory Molecules Acting Big in Cancer: Potential Role of Mito-miRs in Cancer. Curr Mol Med 2020; 19:621-631. [PMID: 31340735 DOI: 10.2174/1566524019666190723165357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/08/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
MicroRNAs [miRNAs] are short, non-coding, single stranded RNA molecules regulating gene expression of their targets at the posttranscriptional level by either degrading mRNA or by inhibiting translation. Previously, miRNAs have been reported to be present inside the mitochondria and these miRNAs have been termed as mito-miRs. Origin of these mito-miRs may either be from mitochondrial genome or import from nucleus. The second class of mito-miRs makes it important to unravel the involvement of miRNAs in crosstalk between nucleus and mitochondria. Since miRNAs are involved in various physiological processes, their deregulation is often associated with disease progression, including cancer. The current review focuses on the involvement of miRNAs in different mitochondrial mediated processes. It also highlights the importance of exploring the interaction of miRNAs with mitochondrial genome, which may lead to the development of small regulatory RNA based therapeutic options.
Collapse
Affiliation(s)
- Praveen Sharma
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Mansa Road, Bathinda 151001, Punjab, India
| | - Bharat
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Mansa Road, Bathinda 151001, Punjab, India
| | - Nilambra Dogra
- Centre for Systems Biology and Bioinformatics, Panjab University, Sector-25, Chandigarh 160014, India
| | - Sandeep Singh
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Mansa Road, Bathinda 151001, Punjab, India
| |
Collapse
|
11
|
Wang W, Wang M, Ruan Y, Tan J, Wang H, Yang T, Li J, Zhou Q. Ginkgolic Acids Impair Mitochondrial Function by Decreasing Mitochondrial Biogenesis and Promoting FUNDC1-Dependent Mitophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:10097-10106. [PMID: 31418272 PMCID: PMC7192365 DOI: 10.1021/acs.jafc.9b04178] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Ginkgolic acids (GAs) are found in the leaves, nuts, and testa of Ginkgo biloba and have been reported to exhibit antitumor, antibacterial, and pro-apoptotic activities. However, their role in mitochondrial function is still unclear. Our previous study showed that genes related to the mitochondria present significant changes in GA-treated mouse bone marrow stromal cells. We hypothesize that GAs may regulate mitochondrial function. Here, we found that GA treatment induced mitochondrial fragmentation, reduced mtDNA copy numbers and mitochondrial protein levels, and impaired mitochondrial adenosine 5'-triphosphate production and oxygen consumption. The GA-induced mitochondrial mass loss may be due to decreased mitochondrial biogenesis. In addition, abolishing autophagy by Atg7 knockout or the administration of an autophagy inhibitor can restore the GA-induced decrease in mitochondrial mass. Furthermore, FUNDC1 knockdown restored the GA-induced changes in mitochondrial mass reduction and mitochondrial membrane potential loss. Together, our studies demonstrated that GAs impaired mitochondrial function by decreasing mitochondrial biogenesis and promoting FUNDC1-dependent mitophagy.
Collapse
Affiliation(s)
- Wenjun Wang
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - Miaomiao Wang
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yu Ruan
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
| | - Junyang Tan
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
| | - Hao Wang
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - Tao Yang
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan 49503, United States
| | - Jianshuang Li
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
- Corresponding Authors: (J.L.)., (Q.Z.)
| | - Qinghua Zhou
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
- Corresponding Authors: (J.L.)., (Q.Z.)
| |
Collapse
|
12
|
Identifying Pig Mitochondrial TSS: Structure and Functional Features. Mitochondrion 2019; 49:19-24. [PMID: 31279875 DOI: 10.1016/j.mito.2019.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/12/2019] [Accepted: 07/03/2019] [Indexed: 11/23/2022]
Abstract
The transcription start sites (TSSs) of porcine mitochondrial genome were firstly identified in this study, including heavy-strand promoter 1 and 2 (HSP1 and HSP2) harbored at nt 903 and nt 1369 in H strand, respectively, and light-strand promoter (LSP) located at nt 166 in L strand. HSP1 structure and expression features were investigated by analyzing mtDNA copy number, expression of 11 nucleoplasmic genes, mtDNA methylation levels, and gene expression levels of methyl-modifying enzymes, DNMT1 and TETs. The mtDNA copy number presented large differences among 15 organs/tissues, and the largest disparity, nearly 17 times, was found between pancreas (~1890 relative copy numbers) and spleen (~110 relative copy numbers, P < .01). The expression levels of HSP1 strand in these organs/tissues presented similar trends with mtDNA copy number (P < .05), and all of 11 nucleoplasmic genes (POLG, POLRMT, TERT, TFAM, TFB1M, TFB2M, NRF-1, PPARα, ESRRA, SP1 and TUFM) detected in this study displayed significantly higher expression values in pancreas than those in spleen (P < .05). Besides, bisulfite sequencing showed that all cytosine residues in the detected region (D-loop) existed methylation with different levels, and the methylation level in spleen was significantly higher than that in pancreas (P < .05). Unlike nuclear DNA, the tested region contained four types of methylation mode (CA, CC, CT, and CG). In addition, the expression of TET1 in pancreas was significantly higher than that in spleen (P < .05). Collectively, our findings indicated that mtDNA TSSs had correlation to mtDNA copy number, expression of nucleoplasmic gene, and mtDNA methylation level.
Collapse
|
13
|
Bouska M, Huang K, Kang P, Bai H. Organelle aging: Lessons from model organisms. J Genet Genomics 2019; 46:171-185. [PMID: 31080045 PMCID: PMC6553499 DOI: 10.1016/j.jgg.2019.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/22/2019] [Accepted: 03/12/2019] [Indexed: 01/07/2023]
Abstract
Most cellular processes descend into failure during aging. While a large collection of longevity pathways has been identified in the past decades, the mechanism for age-related decline of cellular homeostasis and organelle function remains largely unsolved. It is known that many organelles undergo structural and functional changes during normal aging, which significantly contributes to the decline of tissue function at old ages. Since recent studies have revealed an emerging role of organelles as regulatory hubs in maintaining cellular homeostasis, understanding of organelle aging will provide important insights into the cellular basis of organismal aging. Here we review current progress on the characterization of age-dependent structural and functional alterations in the more well-studied organelles, as well as the known mechanisms governing organelle aging in model organisms, with a special focus on the fruit fly Drosophila melanogaster.
Collapse
Affiliation(s)
- Mark Bouska
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Kerui Huang
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Ping Kang
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
14
|
Ning X, Ji X, Li G, Sang N. Ambient PM 2.5 causes lung injuries and coupled energy metabolic disorder. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 170:620-626. [PMID: 30579162 DOI: 10.1016/j.ecoenv.2018.12.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 05/05/2023]
Abstract
Ambient fine particulate matter (PM2.5) is a challenge to public health worldwide. Although increasing numbers of recent epidemiological studies have emphasized the critical role of PM2.5 in promoting respiratory diseases, the precise mechanism behind PM2.5-mediated lung obstruction remains obscure. In the present study, we analyzed lung structure and function and further investigated mitochondrial morphology and transcription-modulated energy metabolism in mice following PM2.5 aspiration. The results showed that PM2.5 exposure reduced pulmonary function and induced severe pathological alterations, including alveolar endothelial disruption and airway obstruction. Based on ultrastructural observations, we also found mitochondrial vacuolation and mitochondrial membrane rupture in alveolar type II epithelial cells. Importantly, the abnormality of mitochondrial structure was coupled with energy metabolism disorders, as evidenced by the decrease in ATP levels, the accumulation of pyruvate and lactate content, and the altered transcription of related genes. Moreover, the reduction in mitochondrial markers, including PGC-1α, NRF-1, and TFAM, were involved in mitochondrial dysfunction. These findings suggest that energy metabolic disorders and mitochondrial dysfunction may be the important contributors to pulmonary injuries in response to PM2.5 exposure, indicating possible targets for protection and therapy in polluted areas.
Collapse
Affiliation(s)
- Xia Ning
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| |
Collapse
|
15
|
Set KK, Sen K, Huq AHM, Agarwal R. Mitochondrial Disorders of the Nervous System: A Review. Clin Pediatr (Phila) 2019; 58:381-394. [PMID: 30607979 DOI: 10.1177/0009922818821890] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kallol K Set
- 1 Dayton Children's Hospital, Dayton, OH, USA.,2 Wright State University Boonshoft School of Medicine, Dayton, OH, USA
| | - Kuntal Sen
- 3 Children's Hospital of Michigan, Detroit, MI, USA.,4 Wayne State University School of Medicine, Detroit, MI, USA
| | - A H M Huq
- 3 Children's Hospital of Michigan, Detroit, MI, USA.,4 Wayne State University School of Medicine, Detroit, MI, USA
| | - Rajkumar Agarwal
- 1 Dayton Children's Hospital, Dayton, OH, USA.,2 Wright State University Boonshoft School of Medicine, Dayton, OH, USA
| |
Collapse
|
16
|
Lee CT, Wang JY, Chou KY, Hsu MI. 1,25-Dihydroxyvitamin D 3 modulates the effects of sublethal BPA on mitochondrial function via activating PI3K-Akt pathway and 17β-estradiol secretion in rat granulosa cells. J Steroid Biochem Mol Biol 2019; 185:200-211. [PMID: 30194976 DOI: 10.1016/j.jsbmb.2018.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/20/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022]
Abstract
Bisphenol A (BPA), an endocrine-disrupting chemical, is capable of producing reproductive toxicity. BPA results in mitochondrial DNA (mtDNA) deletion and mitochondrial dysfunction; however, the effect of BPA on the mitochondria of ovarian granulosa cells is not clear. Further, 1,25-dihydroxyvitamin D3 (1,25D3) may play a role in reproduction, because its receptor, VDR, contributes to the inhibition of oxidative stress and predominantly exists in the nuclei of granulosa cells. Hence, the role of 1,25D3 in BPA-mediated effects on mitochondrial function was examined in this study. Primary rat granulosa cells treated with BPA, 1,25D3, or both were subjected to molecular/biochemical assays to measure cell survival, mtDNA content, mtDNA deletion, superoxide dismutase activity, levels of proteins related to mitochondrial biogenesis, and mitochondrial function. We found that cell viability was dose-dependently reduced and reactive oxygen species (ROS) levels were increased by BPA treatment. BPA administration elevated Mn-superoxide dismutase (MnSOD) expression but negatively regulated total SOD activity. 1,25D3 treatment alone increased 17β-estradiol secretion, ATP production, and cellular oxygen consumption. In cells treated with both agents, 1,25D3 enhanced BPA-induced MnSOD protein upregulation and blocked the BPA-mediated decline in total SOD activity. Furthermore, 1,25D3 attenuated BPA-mediated mtDNA deletion but showed no effect on BPA-induced increases in mtDNA content. Although BPA had no influence on the levels of peroxisome proliferator-activated receptor-γ coactivator-1 α, nuclear respiratory factor-1, mitochondrial transcription factor A, or cytochrome c oxidase subunit IV, 1,25D3 plus BPA markedly increased mitochondrial biogenesis-related protein expression via the PI3K-Akt pathway. Moreover, BPA-mediated negative regulation of cytochrome c oxidase subunit I levels and 17β-estradiol secretion was attenuated by 1,25D3 pre-treatment. Our results suggest that 1,25D3 attenuates BPA-induced decreases in 17β-estradiol and that treatment with 1,25D3 plus BPA regulates granulosa cell mitochondria by elevating mitochondrial biogenesis-related protein levels.
Collapse
Affiliation(s)
- Ching-Tien Lee
- Department of Nursing, Hsin Sheng College of Medical Care and Management, Taoyuan, Taiwan.
| | - Jiz-Yuh Wang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Kuang-Yi Chou
- General Education Center, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
| | - Ming-I Hsu
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, and Department of Obstetrics and Gynecology, Wan Fang Hospital, Taipei Medical.
| |
Collapse
|
17
|
Roth Z. Stress-induced alterations in oocyte transcripts are further expressed in the developing blastocyst. Mol Reprod Dev 2018; 85:821-835. [DOI: 10.1002/mrd.23045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 07/18/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Zvi Roth
- Department of Animal Sciences; Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem; Rehovot Israel
| |
Collapse
|
18
|
Weikard R, Kuehn C. Different mitochondrial DNA copy number in liver and mammary gland of lactating cows with divergent genetic background for milk production. Mol Biol Rep 2018; 45:1209-1218. [PMID: 30051250 DOI: 10.1007/s11033-018-4273-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/18/2018] [Indexed: 01/05/2023]
Abstract
Adequate metabolic adaptation of key tissues playing an essential role for bioenergetic homeostasis and lactogenesis is critical in cows to adapt to changes in energy requirements and physiological processes during the lactation period. Mitochondria are recognized as central to meet energy needs and maintaining of metabolic homeostasis because mitochondrial DNA (mtDNA) is template for several polypeptides of the respiratory chain complexes essential for ATP generation. The quantity of mtDNA in a cell has been widely used as a surrogate marker for the capacity of cells for energy generation. In our study we analyzed the mtDNA copy number and the mRNA expression of important nuclear encoded genes controlling mitochondrial biogenesis in liver and mammary gland. We compared cows with a nuclear genome dairy × beef crossbred make-up to purebred German Holstein dairy cows. The study revealed tissue-specific variations of mtDNA copy number and expression levels of nuclear genes involved in mitochondrial biogenesis when comparing lactating cows with different genetic predisposition regarding milk performance. This may reflect nuclear genome-determined genetic differences between the cow groups in coping with metabolic demands and physiological changes during lactation. The results indicate that mitochondrial biogenesis processes in the liver and mammary gland appear to be impaired in high lactating dairy cows, which consequently, would point to a disturbed energy adaptation. The results provide a basis to further elucidate the adaptive and regulatory modulation of the mitochondrial biogenesis in response to lactation-associated metabolic challenges in lactating cows.
Collapse
Affiliation(s)
- Rosemarie Weikard
- Institute Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Christa Kuehn
- Institute Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany.
| |
Collapse
|
19
|
Nuclear Transcription Factor Kappa B (NF-кB) and Molecular Damage Mechanisms in Acute Cardiovascular Diseases. A Review. JOURNAL OF CARDIOVASCULAR EMERGENCIES 2018. [DOI: 10.2478/jce-2018-0008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Worldwide, cardiovascular diseases (CVDs) represent one of the main causes of morbidity and mortality, and acute coronary syndromes are responsible for a large number of sudden cardiac deaths. One of the main challenges that still exist in this area is represented by the early detection and targeted monitoring of the pathophysiology involved in CVDs. During the last couple of years, researchers have highlighted the importance of molecular and epigenetic mechanisms involved in the initiation and augmentation of CVDs, culminating in their most severe form represented by acute myocardial infarction. One of the most studied molecular factors involved in this type of pathology is represented by nuclear transcription factor kappa B (NF-κB), as well as the involvement of microRNAs (miRNAs). It has been suggested that miRNAs can also be involved in the complex process of atheromatous plaque vulnerabilization that leads to an acute cardiac event. In this review paper, we describe the most important molecular mechanisms involved in the pathogenesis of CVDs and atheromatous plaque progression and vulnerabilization, which include molecular mechanisms dependent on NF-κB. For this paper, we used international databases (PubMed and Scopus). The keywords used for the search were “miRNAs biomarkers”, “miRNAs in cardiovascular disease”, “NF-κB in cardiovascular disease”, “molecular mechanism in cardiovascular disease”, and “myocardial NF-κB mechanisms”. Numerous molecular reactions that have NF-κB as a trigger are involved in the pathogenesis of CVDs. Moreover, miRNAs play an important role in initiating and aggravating certain segments of CVDs. Therefore, miRNAs can be used as biomarkers for early evaluation of CVDs. Furthermore, in the future, miRNAs could be used as a targeted molecular therapy in order to block certain mechanisms responsible for inducing CVDs and leading to acute cardiovascular events.
Collapse
|
20
|
Meseguer S, Panadero J, Navarro-González C, Villarroya M, Boutoual R, Comi GP, Armengod ME. The MELAS mutation m.3243A>G promotes reactivation of fetal cardiac genes and an epithelial-mesenchymal transition-like program via dysregulation of miRNAs. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3022-3037. [PMID: 29928977 DOI: 10.1016/j.bbadis.2018.06.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/25/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022]
Abstract
The pathomechanisms underlying oxidative phosphorylation (OXPHOS) diseases are not well-understood, but they involve maladaptive changes in mitochondria-nucleus communication. Many studies on the mitochondria-nucleus cross-talk triggered by mitochondrial dysfunction have focused on the role played by regulatory proteins, while the participation of miRNAs remains poorly explored. MELAS (mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes) is mostly caused by mutation m.3243A>G in mitochondrial tRNALeu(UUR) gene. Adverse cardiac and neurological events are the commonest causes of early death in m.3243A>G patients. Notably, the incidence of major clinical features associated with this mutation has been correlated to the level of m.3243A>G mutant mitochondrial DNA (heteroplasmy) in skeletal muscle. In this work, we used a transmitochondrial cybrid model of MELAS (100% m.3243A>G mutant mitochondrial DNA) to investigate the participation of miRNAs in the mitochondria-nucleus cross-talk associated with OXPHOS dysfunction. High-throughput analysis of small-RNA-Seq data indicated that expression of 246 miRNAs was significantly altered in MELAS cybrids. Validation of selected miRNAs, including miR-4775 and miR-218-5p, in patient muscle samples revealed miRNAs whose expression declined with high levels of mutant heteroplasmy. We show that miR-218-5p and miR-4775 are direct regulators of fetal cardiac genes such as NODAL, RHOA, ISL1 and RXRB, which are up-regulated in MELAS cybrids and in patient muscle samples with heteroplasmy above 60%. Our data clearly indicate that TGF-β superfamily signaling and an epithelial-mesenchymal transition-like program are activated in MELAS cybrids, and suggest that down-regulation of miRNAs regulating fetal cardiac genes is a risk marker of heart failure in patients with OXPHOS diseases.
Collapse
Affiliation(s)
- Salvador Meseguer
- RNA Modification and Mitochondrial Diseases Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Carrer d'Eduardo Primo Yúfera 3, Valencia 46012, Spain.
| | - Joaquin Panadero
- Unidad de Genómica, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell, 106 Torre A 7ª planta, Valencia 46026, Spain.
| | - Carmen Navarro-González
- RNA Modification and Mitochondrial Diseases Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Carrer d'Eduardo Primo Yúfera 3, Valencia 46012, Spain.
| | - Magda Villarroya
- RNA Modification and Mitochondrial Diseases Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Carrer d'Eduardo Primo Yúfera 3, Valencia 46012, Spain.
| | - Rachid Boutoual
- RNA Modification and Mitochondrial Diseases Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Carrer d'Eduardo Primo Yúfera 3, Valencia 46012, Spain.
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, I.R.C.C.S. Foundation Ca' Granda, Ospedale Maggiore Policlinico, via F. Sforza 35, 20122 Milan, Italy.
| | - M-Eugenia Armengod
- RNA Modification and Mitochondrial Diseases Laboratory, Centro de Investigación Príncipe Felipe (CIPF), Carrer d'Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) node 721, Madrid 28029, Spain.
| |
Collapse
|
21
|
Roth Z. Symposium review: Reduction in oocyte developmental competence by stress is associated with alterations in mitochondrial function. J Dairy Sci 2018; 101:3642-3654. [DOI: 10.3168/jds.2017-13389] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 11/20/2017] [Indexed: 01/10/2023]
|
22
|
Koike N, Hatano Y, Ushimaru T. Heat shock transcriptional factor mediates mitochondrial unfolded protein response. Curr Genet 2018; 64:907-917. [PMID: 29423676 DOI: 10.1007/s00294-018-0809-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 12/22/2022]
Abstract
For maintenance of cytoplasmic protein quality control (PQC), cytoplasmic heat shock proteins (HSPs) negatively control heat shock transcriptional factor (HSF) in a negative feedback loop. However, how mitochondrial protein quality control (mtPQC) is maintained is largely unknown. Here we present evidence that HSF directly monitors mtPQC in the budding yeast Saccharomyces cerevisiae. Mitochondrial HSP70 (Ssc1) negatively regulated HSF activity. Importantly, HSF was localized not only in the nucleus but also on mitochondria. The mitochondrial localization of HSF was increased by heat shock and compromised by SSC1 overexpression. Furthermore, the mitochondrial protein translocation system downregulated HSF activity. Finally, mtPQC modulated the mtHSP genes SSC1 and MDJ1 via HSF, and SSC1 overexpression compromised mitochondrial function. These findings illustrate a model in which HSF directly monitors mtPQC.
Collapse
Affiliation(s)
- Naoki Koike
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, 422-8021, Japan
| | - Yuuki Hatano
- Faculty of Science, Shizuoka University, 836 Ohya, Shizuoka, 422-8529, Japan
| | - Takashi Ushimaru
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka, 422-8021, Japan. .,Faculty of Science, Shizuoka University, 836 Ohya, Shizuoka, 422-8529, Japan.
| |
Collapse
|
23
|
Abstract
The rising toll of chronic and debilitating diseases brought about by the exposure to an ever expanding number of environmental pollutants and socio-economic factors is calling for action. The understanding of the molecular mechanisms behind the effects of environmental exposures can lead to the development of biomarkers that can support the public health fields of both early diagnosis and intervention to limit the burden of environmental diseases. The study of mitochondrial epigenetics carries high hopes to provide important biomarkers of exposure and disease. Mitochondria are in fact on the frontline of the cellular response to the environment. Modifications of the epigenetic factors regulating the mitochondrial activity are emerging as informative tools that can effectively report on the effects of the environment on the phenotype. Here, we will discuss the emerging field of mitochondrial epigenetics. This review describes the main epigenetic phenomena that modify the activity of the mitochondrial DNA including DNA methylation, long and short non-coding RNAs. We will discuss the unique pattern of mitochondrial DNA methylation, describe the challenges of correctly measuring it, and report on the existing studies that have analysed the correlation between environmental exposures and mitochondrial DNA methylation. Finally, we provide a brief account of the therapeutic approaches targeting mitochondria currently under consideration.
Collapse
Affiliation(s)
- Luca Lambertini
- Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levi Place, Box 1057, New York, NY, 10029, USA. .,Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, One Gustave L. Levi Place, Box 1057, New York, NY, 10029, USA.
| | - Hyang-Min Byun
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne, UK.,Ageing Research Laboratory, Newcastle University, Campus for Ageing and Vitality, Edwardson Building, Newcastle upon Tyne, NE4 5PL, UK
| |
Collapse
|
24
|
Klement RJ. Fasting, Fats, and Physics: Combining Ketogenic and Radiation Therapy against Cancer. Complement Med Res 2017; 25:102-113. [DOI: 10.1159/000484045] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Radiotherapy (RT) is a mainstay in the treatment of solid tumors and works by physicochemical reactions inducing oxidative stress in cells. Because in practice the efficacy of RT is limited by its toxicity to normal tissues, any strategy that selectively increases the radiosensitivity of tumor cells or boosts the radioresistance of normal cells is a valuable adjunct to RT. In this review, I summarize preclinical and clinical data supporting the hypothesis that ketogenic therapy through fasting and/or ketogenic diets can be utilized as such an adjunct in order to improve the outcome after RT, in terms of both higher tumor control and lower normal-tissue complication probability. The first effect relates to the metabolic shift from glycolysis towards mitochondrial metabolism, which selectively increases reactive oxygen species (ROS) production and impairs adenoside triphosphate (ATP) production in tumor cells. The second effect is based on the differential stress resistance phenomenon describing the reprogramming of normal cells, but not tumor cells, from proliferation towards maintenance and stress resistance when glucose and growth factor levels are decreased and ketone body levels are elevated. Underlying both effects are metabolic differences between normal and tumor cells. Ketogenic therapy is a non-toxic and cost-effective complementary treatment option that exploits these differences and deserves further clinical investigation.
Collapse
|
25
|
Abstract
PURPOSE Radiotherapy (RT) is a mainstay in the treatment of solid tumors and works by inducing free radical stress in tumor cells, leading to loss of reproductive integrity. The optimal treatment strategy has to consider damage to both tumor and normal cells and is determined by five factors known as the 5 R's of radiobiology: Reoxygenation, DNA repair, radiosensitivity, redistribution in the cell cycle and repopulation. The aim of this review is (i) to present evidence that these 5 R's are strongly influenced by cellular and whole-body metabolism that in turn can be modified through ketogenic therapy in form of ketogenic diets and short-term fasting and (ii) to stimulate new research into this field including some research questions deserving further study. CONCLUSIONS Preclinical and some preliminary clinical data support the hypothesis that ketogenic therapy could be utilized as a complementary treatment in order to improve the outcome after RT, both in terms of higher tumor control and in terms of lower normal tissue complication probability. The first effect relates to the metabolic shift from glycolysis toward mitochondrial metabolism that selectively increases ROS production and impairs ATP production in tumor cells. The second effect is based on the differential stress resistance phenomenon, which is achieved when glucose and growth factors are reduced and ketone bodies are elevated, reprogramming normal but not tumor cells from proliferation toward maintenance and stress resistance. Underlying both effects are metabolic differences between normal and tumor cells that ketogenic therapy seeks to exploit. Specifically, the recently discovered role of the ketone body β-hydroxybutyrate as an endogenous class-I histone deacetylase inhibitor suggests a dual role as a radioprotector of normal cells and a radiosensitzer of tumor cells that opens up exciting possibilities to employ ketogenic therapy as a cost-effective adjunct to radiotherapy against cancer.
Collapse
Affiliation(s)
- Rainer J Klement
- a Department of Radiotherapy and Radiation Oncology , Leopoldina Hospital , Schweinfurt , Germany
| |
Collapse
|
26
|
De Paepe B, Van Coster R. A Critical Assessment of the Therapeutic Potential of Resveratrol Supplements for Treating Mitochondrial Disorders. Nutrients 2017; 9:E1017. [PMID: 28906460 PMCID: PMC5622777 DOI: 10.3390/nu9091017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/01/2017] [Accepted: 09/11/2017] [Indexed: 12/27/2022] Open
Abstract
In human cells, mitochondria provide the largest part of cellular energy in the form of adenosine triphosphate generated by the process of oxidative phosphorylation (OXPHOS). Impaired OXPHOS activity leads to a heterogeneous group of inherited diseases for which therapeutic options today remain very limited. Potential innovative strategies aim to ameliorate mitochondrial function by increasing the total mitochondrial load of tissues and/or to scavenge the excess of reactive oxygen species generated by OXPHOS malfunctioning. In this respect, resveratrol, a compound that conveniently combines mitogenetic with antioxidant activities and, as a bonus, possesses anti-apoptotic properties, has come forward as a promising nutraceutical. We review the scientific evidence gathered so far through experiments in both in vitro and in vivo systems, evaluating the therapeutic effect that resveratrol is expected to generate in mitochondrial patients. The obtained results are encouraging, but clearly show that achieving normalization of OXPHOS function with this strategy alone could prove to be an unattainable goal.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Centre, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Rudy Van Coster
- Department of Pediatrics-Division of Pediatric Neurology and Metabolism, Ghent University Hospital, 9000 Ghent, Belgium.
| |
Collapse
|
27
|
De Paepe B, Lefever S, Mestdagh P. How long noncoding RNAs enforce their will on mitochondrial activity: regulation of mitochondrial respiration, reactive oxygen species production, apoptosis, and metabolic reprogramming in cancer. Curr Genet 2017; 64:163-172. [DOI: 10.1007/s00294-017-0744-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/17/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022]
|
28
|
Healy TM, Bryant HJ, Schulte PM. Mitochondrial genotype and phenotypic plasticity of gene expression in response to cold acclimation in killifish. Mol Ecol 2017; 26:814-830. [DOI: 10.1111/mec.13945] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/11/2016] [Accepted: 11/16/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Timothy M. Healy
- Department of Zoology; The University of British Columbia; 6270 University Blvd Vancouver BC Canada V6T 1Z4
| | - Heather J. Bryant
- Department of Zoology; The University of British Columbia; 6270 University Blvd Vancouver BC Canada V6T 1Z4
| | - Patricia M. Schulte
- Department of Zoology; The University of British Columbia; 6270 University Blvd Vancouver BC Canada V6T 1Z4
| |
Collapse
|
29
|
Mitochondrial growth during the cell cycle of Trypanosoma brucei bloodstream forms. Sci Rep 2016; 6:36565. [PMID: 27874016 PMCID: PMC5118809 DOI: 10.1038/srep36565] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/30/2016] [Indexed: 01/11/2023] Open
Abstract
Mitochondrial organelles need to be replicated during cell division. Many aspects of this process have been studied in great detail, however the actual size increase and the position of organelle growth are less well understood. We use the protozoan parasite Trypanosoma brucei that contains a single mitochondrion to study organelle biogenesis by fluorescence microscopy. From the analysis of more than 1000 T. brucei bloodstream form cells of a nonsynchronous population we conclude that the mitochondrial network mostly grows from two areas along the main organelle axis, posterior and anterior of the nucleus. Loops and branches from these two areas eventually fuse to build a complex network. Together with the appearance of the division fold in the posterior part of the cell, pruning of the mitochondrial network and finally separation into the two daughter cells occurs. Overall organelle biogenesis is not continuous during cell growth and occurs mostly in the last part of the cell cycle. Furthermore, using 3D STED super resolution microscopy we reconstruct the volume of the organelle and characterize the region where the mitochondrial genome is positioned by serial block face scanning electron microscopy.
Collapse
|
30
|
Roth Z. Effect of Heat Stress on Reproduction in Dairy Cows: Insights into the Cellular and Molecular Responses of the Oocyte. Annu Rev Anim Biosci 2016; 5:151-170. [PMID: 27732786 DOI: 10.1146/annurev-animal-022516-022849] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Among the components of the female reproductive tract, the ovarian pool of follicles and their enclosed oocytes are highly sensitive to hyperthermia. Heat-induced alterations in small antral follicles can be expressed later as compromised maturation and developmental capacity of the ovulating oocyte. This review summarizes the most up-to-date information on the effects of heat stress on the oocyte with an emphasis on unclear points and open questions, some of which might involve new research directions, for instance, whether preantral follicles are heat resistant. The review focuses on the follicle-enclosed oocytes, provides new insights into the cellular and molecular responses of the oocyte to elevated temperature, points out the role of the follicle microenvironment, and discusses some mechanisms that might underlie oocyte impairment. Mechanisms include nuclear and cytoplasmic maturation, mitochondrial function, apoptotic pathways, and oxidative stress. Understanding the mechanism by which heat stress compromises fertility might enable development of new strategies to mitigate its effects.
Collapse
Affiliation(s)
- Zvi Roth
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, Rehovot 76100, Israel;
| |
Collapse
|
31
|
Kesner EE, Saada-Reich A, Lorberboum-Galski H. Characteristics of Mitochondrial Transformation into Human Cells. Sci Rep 2016; 6:26057. [PMID: 27184109 PMCID: PMC4868981 DOI: 10.1038/srep26057] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/27/2016] [Indexed: 12/12/2022] Open
Abstract
Mitochondria can be incorporated into mammalian cells by simple co-incubation of isolated mitochondria with cells, without the need of transfection reagents or any other type of intervention. This phenomenon was termed mitochondrial transformation, and although it was discovered in 1982, currently little is known regarding its mechanism(s). Here we demonstrate that mitochondria can be transformed into recipient cells very quickly, and co-localize with endogenous mitochondria. The isolated mitochondria interact directly with cells, which engulf the mitochondria with cellular extensions in a way, which may suggest the involvement of macropinocytosis or macropinocytosis-like mechanisms in mitochondrial transformation. Indeed, macropinocytosis inhibitors but not clathrin-mediated endocytosis inhibition-treatments, blocks mitochondria transformation. The integrity of the mitochondrial outer membrane and its proteins is essential for the transformation of the mitochondria into cells; cells can distinguish mitochondria from similar particles and transform only intact mitochondria. Mitochondrial transformation is blocked in the presence of the heparan sulfate molecules pentosan polysulfate and heparin, which indicate crucial involvement of cellular heparan sulfate proteoglycans in the mitochondrial transformation process.
Collapse
Affiliation(s)
- E. E. Kesner
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - A. Saada-Reich
- Monique and Jacques Roboh Department of Genetic Research, Department of Genetics and Metabolic Diseases, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - H. Lorberboum-Galski
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| |
Collapse
|
32
|
Medium-chain TAG improve energy metabolism and mitochondrial biogenesis in the liver of intra-uterine growth-retarded and normal-birth-weight weanling piglets. Br J Nutr 2016; 115:1521-30. [PMID: 26960981 DOI: 10.1017/s0007114516000404] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
We previously reported that medium-chain TAG (MCT) could alleviate hepatic oxidative damage in weanling piglets with intra-uterine growth retardation (IUGR). There is a relationship between oxidative status and energy metabolism, a process involved in substrate availability and glucose flux. Therefore, the aim of this study was to investigate the effects of IUGR and MCT on hepatic energy metabolism and mitochondrial function in weanling piglets. Twenty-four IUGR piglets and twenty-four normal-birth-weight (NBW) piglets were fed a diet of either soyabean oil (SO) or MCT from 21 d of postnatal age to 49 d of postnatal age. Then, the piglets' biochemical parameters and gene expressions related to energy metabolism and mitochondrial function were determined (n 4). Compared with NBW, IUGR decreased the ATP contents and succinate oxidation rates in the liver of piglets, and reduced hepatic mitochondrial citrate synthase (CS) activity (P<0·05). IUGR piglets exhibited reductions in hepatic mitochondrial DNA (mtDNA) contents and gene expressions related to mitochondrial biogenesis compared with NBW piglets (P<0·05). The MCT diet increased plasma ghrelin concentration and hepatic CS and succinate dehydrogenase activities, but decreased hepatic pyruvate kinase activity compared with the SO diet (P<0·05). The MCT-fed piglets showed improved mtDNA contents and PPARγ coactivator-1α expression in the liver (P<0·05). The MCT diet alleviated decreased mRNA abundance of the hepatic PPARα induced by IUGR (P<0·05). It can therefore be postulated that MCT may have beneficial effects in improving energy metabolism and mitochondrial function in weanling piglets.
Collapse
|
33
|
Liu X, Trakooljul N, Muráni E, Krischek C, Schellander K, Wicke M, Wimmers K, Ponsuksili S. Molecular changes in mitochondrial respiratory activity and metabolic enzyme activity in muscle of four pig breeds with distinct metabolic types. J Bioenerg Biomembr 2016; 48:55-65. [PMID: 26759028 DOI: 10.1007/s10863-015-9639-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/21/2015] [Indexed: 02/05/2023]
Abstract
Skeletal muscles are metabolically active and have market value in meat-producing farm animals. A better understanding of biological pathways affecting energy metabolism in skeletal muscle could advance the science of skeletal muscle. In this study, comparative pathway-focused gene expression profiling in conjunction with muscle fiber typing were analyzed in skeletal muscles from Duroc, Pietrain, and Duroc-Pietrain crossbred pigs. Each breed type displayed a distinct muscle fiber-type composition. Mitochondrial respiratory activity and glycolytic and oxidative enzyme activities were comparable among genotypes, except for significantly lower complex I activity in Pietrain pigs homozygous-positive for malignant hyperthermia syndrome. At the transcriptional level, lactate dehydrogenase B showed breed specificity, with significantly lower expression in Pietrain pigs homozygous-positive for malignant hyperthermia syndrome. A similar mRNA expression pattern was shown for several subunits of oxidative phosphorylation complexes, including complex I, complex II, complex IV, and ATP synthase. Significant correlations were observed between mRNA expression of genes in focused pathways and enzyme activities in a breed-dependent manner. Moreover, expression patterns of pathway-focused genes were well correlated with muscle fiber-type composition. These results stress the importance of regulation of transcriptional rate of genes related to oxidative and glycolytic pathways in the metabolic capacity of muscle fibers. Overall, the results further the breed-specific understanding of the molecular basis of metabolic enzyme activities, which directly impact meat quality.
Collapse
Affiliation(s)
- Xuan Liu
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Eduard Muráni
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Carsten Krischek
- Institute of Food Quality and Food Safety, University of Veterinary Medicine Hannover, D-30173, Hannover, Germany
| | - Karl Schellander
- Insititute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, Bonn, Germany
| | - Michael Wicke
- Department of Animal Sciences, Quality of Food of Animal Origin, Georg-August-University Goettingen, Goettingen, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
34
|
Ghaffari Novin M, Noruzinia M, Allahveisi A, Saremi A, Fadaei Fathabadi F, Mastery Farahani R, Dehghani Fard A, Pooladi A, Mazaherinezhad Fard R, Yousefian E. Comparison of mitochondrial-related transcriptional levels of TFAM, NRF1 and MT-CO1 genes in single human oocytes at various stages of the oocyte maturation. IRANIAN BIOMEDICAL JOURNAL 2015; 19:23-8. [PMID: 25605486 PMCID: PMC4322229 DOI: 10.6091/ibj.1400.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background: The aim of the current study was to assess the mRNA levels of two mitochondria-related genes, including nuclear-encoded NRF1 (nuclear respiratory factor 1), mitochondrial transcription factor A (TFAM), and mitochondrial-encoded cytochrome c oxidase subunit 1 (MT-CO1) genes in various stages of the human oocyte maturation. Methods: Oocytes were obtained from nine infertile women with male factor undergoing in vitro fertilization (IVF)/intra-cytoplasmic sperm injection protocol. Mitochondrial-related mRNA levels were performed by single-cell TaqMan real-time PCR. Results: the expression level of the target genes was low at the germinal vesicle stage (P>0.05). Although the mRNA level of NRF1gene remained stable in metaphase I, the mRNA level of TFAM and MT-CO1 increased significantly (P<0.05).In metaphase II, the expression level of all genes increased compared to metaphase I (P<0.05).Conclusion: The overexpression levels of NRF1, TFAM, and MT-CO1 genes are related to the oocyte maturation. Therefore, the current study could be used clinically to improve the success rate of IVF.
Collapse
Affiliation(s)
- Marefat Ghaffari Novin
- Dept. of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Velenjak, Tehran, Iran
| | - Mehrdad Noruzinia
- Dept. of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Azra Allahveisi
- Dept. of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Velenjak, Tehran, Iran
| | - Aboutaleb Saremi
- Sarem Cell Research Center (SCRC), Sarem Women’s Hospital, Tehran, Iran
| | - Fateme Fadaei Fathabadi
- Dept. of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Velenjak, Tehran, Iran
| | - Reza Mastery Farahani
- Dept. of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Velenjak, Tehran, Iran
| | - Ali Dehghani Fard
- Sarem Cell Research Center (SCRC), Sarem Women’s Hospital, Tehran, Iran
| | - Arash Pooladi
- Sarem Cell Research Center (SCRC), Sarem Women’s Hospital, Tehran, Iran
| | | | - Elham Yousefian
- Dept. of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Velenjak, Tehran, Iran
| |
Collapse
|
35
|
Collett JA, Paulose JK, Cassone VM, Osborn JL. Kidney-Specific Reduction of Oxidative Phosphorylation Genes Derived from Spontaneously Hypertensive Rat. PLoS One 2015; 10:e0136441. [PMID: 26308211 PMCID: PMC4550288 DOI: 10.1371/journal.pone.0136441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 08/03/2015] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial (Mt) dysfunction contributes to the pathophysiology of renal function and promotes cardiovascular disease such as hypertension. We hypothesize that renal Mt-genes derived from female spontaneously hypertensive rats (SHR) that exhibit hypertension have reduced expression specific to kidney cortex. After breeding a female Okamoto-Aoki SHR (SAP = 188mmHg) with Brown Norway (BN) males (SAP = 100 and 104 mmHg), hypertensive female progeny were backcrossed with founder BN for 5 consecutive generations in order to maintain the SHR mitochondrial genome in offspring that contain over increasing BN nuclear genome. Mt-protein coding genes (13 total) and nuclear transcription factors mediating Mt-gene transcription were evaluated in kidney, heart and liver of normotensive (NT: n = 20) vs. hypertensive (HT: n = 20) BN/SHR-mtSHR using quantitative real-time PCR. Kidney cortex, but not liver or heart Mt-gene expression was decreased ~2–5 fold in 12 of 13 protein encoding genes of HT BN/SHR-mtSHR. Kidney cortex but not liver mRNA expression of the nuclear transcription factors Tfam, NRF1, NRF2 and Pgc1α were also decreased in HT BN/SHR-mtSHR. Kidney cortical tissue of HT BN/SHR-mtSHR exhibited lower cytochrome oxidase histochemical staining, indicating a reduction in renal oxidative phosphorylation but not in liver or heart. These results support the hypothesis that renal cortex of rats with SHR mitochondrial genome has specifically altered renal expression of genes encoding mitochondrial proteins. This kidney-specific coordinated reduction of mitochondrial and nuclear oxidative metabolism genes may be associated with heritable hypertension in SHR.
Collapse
Affiliation(s)
- Jason A. Collett
- Department of Biology, University of Kentucky, Lexington, KY, the United States of America
- * E-mail:
| | - Jiffin K. Paulose
- Department of Biology, University of Kentucky, Lexington, KY, the United States of America
| | - Vincent M. Cassone
- Department of Biology, University of Kentucky, Lexington, KY, the United States of America
| | - Jeffrey L. Osborn
- Department of Biology, University of Kentucky, Lexington, KY, the United States of America
| |
Collapse
|
36
|
Kalifa L, Gewandter JS, Staversky RJ, Sia EA, Brookes PS, O'Reilly MA. DNA double-strand breaks activate ATM independent of mitochondrial dysfunction in A549 cells. Free Radic Biol Med 2014; 75:30-9. [PMID: 25048973 PMCID: PMC4171189 DOI: 10.1016/j.freeradbiomed.2014.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 10/25/2022]
Abstract
Excessive nuclear or mitochondrial DNA damage can lead to mitochondrial dysfunction, decreased energy production, and increased generation of reactive oxygen species (ROS). Although numerous cell signaling pathways are activated when cells are injured, the ataxia telangiectasia mutant (ATM) protein has emerged as a major regulator of the response to both mitochondrial dysfunction and nuclear DNA double-strand breaks (DSBs). Because mitochondrial dysfunction is often a response to excessive DNA damage, it has been difficult to determine whether nuclear and/or mitochondrial DNA DSBs activate ATM independent of mitochondrial dysfunction. In this study, mitochondrial and nuclear DNA DSBs were generated in the A549 human lung adenocarcinoma cell line by infecting with retroviruses expressing the restriction endonuclease PstI fused to a mitochondrial targeting sequence (MTS) or nuclear localization sequence (NLS) and a hemagglutinin antigen epitope tag (HA). Expression of MTS-PstI-HA or NLS-PstI-HA activated the DNA damage response defined by phosphorylation of ATM, the tumor suppressor protein p53 (TP53), KRAB-associated protein (KAP)-1, and structural maintenance of chromosomes (SMC)-1. Phosphorylated ATM and SMC1 were detected in nuclear fractions, whereas phosphorylated TP53 and KAP1 were detected in both mitochondrial and nuclear fractions. PstI also enhanced expression of the cyclin-dependent kinase inhibitor p21 and inhibited cell growth. This response to DNA damage occurred in the absence of detectable mitochondrial dysfunction and excess production of ROS. These findings reveal that DNA DSBs are sufficient to activate ATM independent of mitochondrial dysfunction and suggest that the activated form of ATM and some of its substrates are restricted to the nuclear compartment, regardless of the site of DNA damage.
Collapse
Affiliation(s)
- Lidza Kalifa
- Department of Environmental Medicine, The University of Rochester, Rochester, NY 14642, USA
| | - Jennifer S Gewandter
- Department of Anesthesiology, The University of Rochester, Rochester, NY 14642, USA
| | - Rhonda J Staversky
- Department of Pediatrics, The University of Rochester, Rochester, NY 14642, USA
| | - Elaine A Sia
- Department of Biology, The University of Rochester, Rochester, NY 14642, USA
| | - Paul S Brookes
- Department of Anesthesiology, The University of Rochester, Rochester, NY 14642, USA
| | - Michael A O'Reilly
- Department of Pediatrics, The University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
37
|
Wijnen WJ, van der Made I, van den Oever S, Hiller M, de Boer BA, Picavet DI, Chatzispyrou IA, Houtkooper RH, Tijsen AJ, Hagoort J, van Veen H, Everts V, Ruijter JM, Pinto YM, Creemers EE. Cardiomyocyte-specific miRNA-30c over-expression causes dilated cardiomyopathy. PLoS One 2014; 9:e96290. [PMID: 24789369 PMCID: PMC4008570 DOI: 10.1371/journal.pone.0096290] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 04/07/2014] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs (miRNAs) regulate many aspects of cellular function and their deregulation has been implicated in heart disease. MiRNA-30c is differentially expressed in the heart during the progression towards heart failure and in vitro studies hint to its importance in cellular physiology. As little is known about the in vivo function of miRNA-30c in the heart, we generated transgenic mice that specifically overexpress miRNA-30c in cardiomyocytes. We show that these mice display no abnormalities until about 6 weeks of age, but subsequently develop a severely dilated cardiomyopathy. Gene expression analysis of the miRNA-30c transgenic hearts before onset of the phenotype indicated disturbed mitochondrial function. This was further evident by the downregulation of mitochondrial oxidative phosphorylation (OXPHOS) complexes III and IV at the protein level. Taken together these data indicate impaired mitochondrial function due to OXPHOS protein depletion as a potential cause for the observed dilated cardiomyopathic phenotype in miRNA-30c transgenic mice. We thus establish an in vivo role for miRNA-30c in cardiac physiology, particularly in mitochondrial function.
Collapse
Affiliation(s)
- Wino J. Wijnen
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
- Interuniversitair Cardiologisch Instituut Nederland (ICIN-NHI), Utrecht, The Netherlands
| | | | | | - Monika Hiller
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Bouke A. de Boer
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Daisy I. Picavet
- Department of Cell Biology and Histology, Leeuwenhoek Center of Advanced Microscopy (LCAM), Academic Medical Center, Amsterdam, The Netherlands
| | - Iliana A. Chatzispyrou
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Anke J. Tijsen
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Jaco Hagoort
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Henk van Veen
- Department of Cell Biology and Histology, Leeuwenhoek Center of Advanced Microscopy (LCAM), Academic Medical Center, Amsterdam, The Netherlands
| | - Vincent Everts
- Department of Cell Biology and Histology, Leeuwenhoek Center of Advanced Microscopy (LCAM), Academic Medical Center, Amsterdam, The Netherlands
| | - Jan M. Ruijter
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Yigal M. Pinto
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther E. Creemers
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Melo-Ferreira J, Vilela J, Fonseca MM, da Fonseca RR, Boursot P, Alves PC. The elusive nature of adaptive mitochondrial DNA evolution of an arctic lineage prone to frequent introgression. Genome Biol Evol 2014; 6:886-96. [PMID: 24696399 PMCID: PMC4007550 DOI: 10.1093/gbe/evu059] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2014] [Indexed: 12/21/2022] Open
Abstract
Mitochondria play a fundamental role in cellular metabolism, being responsible for most of the energy production of the cell in the oxidative phosphorylation (OXPHOS) pathway. Mitochondrial DNA (mtDNA) encodes for key components of this process, but its direct role in adaptation remains far from understood. Hares (Lepus spp.) are privileged models to study the impact of natural selection on mitogenomic evolution because 1) species are adapted to contrasting environments, including arctic, with different metabolic pressures, and 2) mtDNA introgression from arctic into temperate species is widespread. Here, we analyzed the sequences of 11 complete mitogenomes (ten newly obtained) of hares of temperate and arctic origins (including two of arctic origin introgressed into temperate species). The analysis of patterns of codon substitutions along the reconstructed phylogeny showed evidence for positive selection in several codons in genes of the OXPHOS complexes, most notably affecting the arctic lineage. However, using theoretical models, no predictable effect of these differences was found on the structure and physicochemical properties of the encoded proteins, suggesting that the focus of selection may lie on complex interactions with nuclear encoded peptides. Also, a cloverleaf structure was detected in the control region only from the arctic mtDNA lineage, which may influence mtDNA replication and transcription. These results suggest that adaptation impacted the evolution of hare mtDNA and may have influenced the occurrence and consequences of the many reported cases of massive mtDNA introgression. However, the origin of adaptation remains elusive.
Collapse
Affiliation(s)
- José Melo-Ferreira
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, InBIO Laboratório Associado, Portugal
| | - Joana Vilela
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, InBIO Laboratório Associado, Portugal
- Departamento de Biologia da Faculdade de Ciências da Universidade do Porto, Portugal
| | - Miguel M. Fonseca
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, InBIO Laboratório Associado, Portugal
- Departamento de Bioquímica, Genética e Inmunología, Facultad de Biología, Universidad de Vigo, Spain
| | - Rute R. da Fonseca
- Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, Denmark
- CIMAR/CIIMAR, Centro Interdisciplinar de Investigação Marinha e Ambiental, Universidade do Porto, Portugal
| | - Pierre Boursot
- Institut des Sciences de l’Evolution, Université Montpellier 2, CNRS, IRD, France
| | - Paulo C. Alves
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, InBIO Laboratório Associado, Portugal
- Departamento de Biologia da Faculdade de Ciências da Universidade do Porto, Portugal
- Wildlife Biology Program, College of Forestry and Conservation, University of Montana, Missoula
| |
Collapse
|
39
|
Xie Y, Li J, Fan G, Qi S, Li B. Reperfusion promotes mitochondrial biogenesis following focal cerebral ischemia in rats. PLoS One 2014; 9:e92443. [PMID: 24667167 PMCID: PMC3965405 DOI: 10.1371/journal.pone.0092443] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 02/21/2014] [Indexed: 12/21/2022] Open
Abstract
Background and Purpose Reperfusion after transient cerebral ischemia causes severe damage to mitochondria; however, little is known regarding the continuous change in mitochondrial biogenesis during reperfusion. Mitochondrial biogenesis causes an increase in the individual mitochondrial mass of neurons and maintains their aerobic set-point in the face of declining function. The aim of this study was to examine mitochondrial biogenesis in the cortex during reperfusion following focal cerebral ischemia. Methods Male Wistar rats were subjected to transient focal cerebral ischemia. The relative amount of cortical mitochondrial DNA was analyzed using quantitative real-time PCR at 0 h, 24 h, 72 h, and 7 d after reperfusion. Three critical transcriptional regulators of mitochondrial biogenesis were measured by semi-quantitative reverse-transcription PCR. The protein expression of cytochrome C oxidase subunits I and IV was detected by Western blotting. Results Evidence of increased mitochondrial biogenesis was observed after reperfusion. The cortical mitochondrial DNA content increased after 24 h, peaked after 72 h, and maintained a high level for 7 d. The cortical expression of three critical genes for the transcriptional regulation of mitochondrial biogenesis, namely, peroxisome proliferator-activated receptor coactivator-1α, nuclear respiratory factor-1, and mitochondrial transcription factor A, also increased at 24 h and 72 h. The expression of peroxisome proliferator-activated receptor coactivator-1α returned to the baseline level at 7 d, but two other factors maintained higher levels compared with the controls. Moreover, the expression of cytochrome C oxidase subunits I and IV was increased in the cortex. Conclusions These results indicate that reperfusion increased mitochondrial biogenesis following focal cerebral ischemia, and this tendency was exacerbated as the reperfusion time was extended. Reperfusion-induced mitochondrial biogenesis was mediated through up-regulation of critical transcriptional regulators of mitochondrial biogenesis.
Collapse
Affiliation(s)
- Yuying Xie
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jun Li
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guibo Fan
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Sihua Qi
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
- * E-mail: (SQ); (BL)
| | - Bing Li
- Department of Nephrology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
- * E-mail: (SQ); (BL)
| |
Collapse
|
40
|
Mitochondrial respiration in blood platelets of depressive patients. Mitochondrion 2013; 13:795-800. [DOI: 10.1016/j.mito.2013.05.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/26/2013] [Accepted: 05/07/2013] [Indexed: 12/17/2022]
|
41
|
Baldelli S, Aquilano K, Ciriolo MR. Punctum on two different transcription factors regulated by PGC-1α: Nuclear factor erythroid-derived 2-like 2 and nuclear respiratory factor 2. Biochim Biophys Acta Gen Subj 2013; 1830:4137-46. [DOI: 10.1016/j.bbagen.2013.04.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/21/2013] [Accepted: 04/02/2013] [Indexed: 12/30/2022]
|
42
|
Kawada I, Hasina R, Lennon FE, Bindokas VP, Usatyuk P, Tan YHC, Krishnaswamy S, Arif Q, Carey G, Hseu RD, Robinson M, Tretiakova M, Brand TM, Iida M, Ferguson MK, Wheeler DL, Husain AN, Natarajan V, Vokes EE, Singleton PA, Salgia R. Paxillin mutations affect focal adhesions and lead to altered mitochondrial dynamics: relevance to lung cancer. Cancer Biol Ther 2013; 14:679-91. [PMID: 23792636 PMCID: PMC3742497 DOI: 10.4161/cbt.25091] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cytoskeletal and focal adhesion abnormalities are observed in several types of cancer, including lung cancer. We have previously reported that paxillin (PXN) was mutated, amplified, and overexpressed in a significant number of lung cancer patient samples, that PXN protein was upregulated in more advanced stages of lung cancer compared with lower stages, and that the PXN gene was also amplified in some pre-neoplastic lung lesions. Among the mutations investigated, we previously found that PXN variant A127T in lung cancer cells enhanced cell proliferation and focal adhesion formation and colocalized with the anti-apoptotic protein B Cell Lymphoma 2 (BCL-2), which is known to localize to the mitochondria, among other sites. To further explore the effects of activating mutations of PXN on mitochondrial function, we cloned and expressed wild-type PXN and variants containing the most commonly occurring PXN mutations (P46S, P52L, G105D, A127T, P233L, T255I, D399N, E423K, P487L, and K506R) in a GFP-tagged vector using HEK-293 human embryonic kidney cells. Utilizing live-cell imaging to systematically study the effects of wild-type PXN vs. mutants, we created a model that recapitulates the salient features of the measured dynamics and conclude that compared with wild-type, some mutant clones confer enhanced focal adhesion and lamellipodia formation (A127T, P233L, and P487L) and some confer increased association with BCL-2, Dynamin-related Protein-1 (DRP-1), and Mitofusion-2 (MFN-2) proteins (P233L and D399N). Further, PXN mutants, through their interactions with BCL-2 and DRP-1, could regulate cisplatin drug resistance in human lung cancer cells. The data reported herein suggest that mutant PXN variants play a prominent role in mitochondrial dynamics with direct implications on lung cancer progression and hence, deserve further exploration as therapeutic targets.
Collapse
Affiliation(s)
- Ichiro Kawada
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pienaar IS, Chinnery PF. Existing and emerging mitochondrial-targeting therapies for altering Parkinson's disease severity and progression. Pharmacol Ther 2013; 137:1-21. [DOI: 10.1016/j.pharmthera.2012.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 08/07/2012] [Indexed: 02/07/2023]
|
44
|
Zhao XY, Sun JL, Hu YJ, Yang Y, Zhang WJ, Hu Y, Li J, Sun Y, Zhong Y, Peng W, Zhang HL, Kong WJ. The effect of overexpression of PGC-1α on the mtDNA4834 common deletion in a rat cochlear marginal cell senescence model. Hear Res 2012; 296:13-24. [PMID: 23159434 DOI: 10.1016/j.heares.2012.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 10/17/2012] [Accepted: 11/07/2012] [Indexed: 10/27/2022]
Abstract
Aging is a natural process usually defined as a progressive loss of function with an accumulation of senescent cells. The clinical manifestations of this process include age-related hearing loss (AHL)/presbycusis. Several investigations indicated the association between a mitochondrial common deletion (CD) (mtDNA 4977-bp deletion in humans, corresponding to 4834-bp deletion in rats) and presbycusis. Previous researches have shown that peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) is a key regulator of mitochondrial biogenesis and energy metabolism. However, the expression of PGC-1α in the inner ear and the possible effect of PGC-1α on presbycusis are not clear. Our data demonstrated the distribution of PGC-1α and its downstream transcription factors nuclear respiratory factor-1 (NRF-1), mitochondrial transcription factor A (Tfam) and nuclear factor κB (NF-κB) in marginal cells (MCs) for the first time. To explore the role of PGC-1α in cellular senescence, we established a model of marginal cell senescence harboring the mtDNA4834 common deletion induced by d-galactose. We also found that PGC-1α and its downstream transcription factors compensatorily increased in our cell senescence model. Furthermore, the overexpression of PGC-1α induced by transfection largely increased the expression levels of NRF-1 and TFAM and significantly decreased the expression level of NF-κB in the cell senescence model. And the levels of CD, senescent cells and apoptotic cells in the cell model decreased after PGC-1α overexpression. These results suggested that PGC-1α might protect MCs in this cell model from senescence through a nuclear-mitochondrial interaction and against apoptosis. Our study may shed light on the pathogenesis of presbycusis and provide a new therapeutic target for presbycusis.
Collapse
Affiliation(s)
- Xue-Yan Zhao
- Department of Otolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Li P, Jiao J, Gao G, Prabhakar BS. Control of mitochondrial activity by miRNAs. J Cell Biochem 2012; 113:1104-10. [PMID: 22135235 DOI: 10.1002/jcb.24004] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondria supply energy for physiological function and they participate in the regulation of other cellular events including apoptosis, calcium homeostasis, and production of reactive oxygen species. Thus, mitochondria play a critical role in the cells. However, dysfunction of mitochondria is related to a variety of pathological processes and diseases. MicroRNAs (miRNAs) are a class of small noncoding RNAs about 22 nucleotides long, and they can bind to the 3'-untranslated region (3'-UTR) of mRNAs, thereby inhibiting mRNA translation or promoting mRNA degradation. We summarize the molecular regulation of mitochondrial metabolism, structure, and function by miRNAs. Modulation of miRNAs levels may provide a new therapeutic approach for the treatment of mitochondria-related diseases.
Collapse
Affiliation(s)
- Peifeng Li
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | |
Collapse
|
46
|
Parr RL, Martin LH. Mitochondrial and nuclear genomics and the emergence of personalized medicine. Hum Genomics 2012; 6:3. [PMID: 23244780 PMCID: PMC3437567 DOI: 10.1186/1479-7364-6-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 07/05/2012] [Indexed: 11/29/2022] Open
Abstract
Developing early detection biosensors for disease has been the long‒held goal of the Human Genome Project, but with little success. Conversely, the biological properties of the mitochondrion coupled with the relative simplicity of the mitochondrial genome give this organelle extraordinary functionality as a biosensor and places the field of mitochondrial genomics in a position of strategic advantage to launch significant advances in personalized medicine. Numerous factors make the mitochondrion organelle uniquely suited to be an early detection biosensor with applications in oncology as well as many other aspects of human health and disease. Early detection of disease translates into more effective, less expensive treatments for disease and overall better prognoses for those at greater risk for developing diseases.
Collapse
|
47
|
Bellizzi D, D'Aquila P, Giordano M, Montesanto A, Passarino G. Global DNA methylation levels are modulated by mitochondrial DNA variants. Epigenomics 2012; 4:17-27. [PMID: 22332655 DOI: 10.2217/epi.11.109] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
AIM In the present study, we investigated whether global DNA methylation levels are affected by mitochondrial DNA (mtDNA) variants, which are known to modulate mitochondrial functions. MATERIALS & METHODS Global DNA methylation levels were evaluated in peripheral blood DNA collected from adult subjects and in vitro using the DNA of cybrid cells harboring mtDNAs of different haplogroups. In these cells, mRNA expression of genes involved in DNA methylation processes, and ATP and reactive oxygen species levels were also analyzed. RESULTS The analysis revealed that methylation levels were higher in the subjects carrying the J haplogroup than in non-J carriers. Consistently, cybrids with J haplogroup mtDNA showed higher methylation levels than other cybrids. Interestingly, we observed overexpression of the MAT1A gene and low ATP and ROS levels in J cybrids. CONCLUSION Our findings indicate that mtDNA-specific interactions between mitochondria and the nucleus regulate epigenetic changes, possibly by affecting oxidative phosphorylation efficiency.
Collapse
Affiliation(s)
- Dina Bellizzi
- Department of Cell Biology, University of Calabria, 87036 Rende, Italy
| | | | | | | | | |
Collapse
|
48
|
Mercer TR, Neph S, Dinger ME, Crawford J, Smith MA, Shearwood AMJ, Haugen E, Bracken CP, Rackham O, Stamatoyannopoulos JA, Filipovska A, Mattick JS. The human mitochondrial transcriptome. Cell 2011; 146:645-58. [PMID: 21854988 DOI: 10.1016/j.cell.2011.06.051] [Citation(s) in RCA: 629] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 06/15/2011] [Accepted: 06/27/2011] [Indexed: 11/27/2022]
Abstract
The human mitochondrial genome comprises a distinct genetic system transcribed as precursor polycistronic transcripts that are subsequently cleaved to generate individual mRNAs, tRNAs, and rRNAs. Here, we provide a comprehensive analysis of the human mitochondrial transcriptome across multiple cell lines and tissues. Using directional deep sequencing and parallel analysis of RNA ends, we demonstrate wide variation in mitochondrial transcript abundance and precisely resolve transcript processing and maturation events. We identify previously undescribed transcripts, including small RNAs, and observe the enrichment of several nuclear RNAs in mitochondria. Using high-throughput in vivo DNaseI footprinting, we establish the global profile of DNA-binding protein occupancy across the mitochondrial genome at single-nucleotide resolution, revealing regulatory features at mitochondrial transcription initiation sites and functional insights into disease-associated variants. This integrated analysis of the mitochondrial transcriptome reveals unexpected complexity in the regulation, expression, and processing of mitochondrial RNA and provides a resource for future studies of mitochondrial function (accessed at http://mitochondria.matticklab.com).
Collapse
Affiliation(s)
- Tim R Mercer
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sripathi SR, He W, Atkinson CL, Smith JJ, Liu Z, Elledge BM, Jahng WJ. Mitochondrial-nuclear communication by prohibitin shuttling under oxidative stress. Biochemistry 2011; 50:8342-51. [PMID: 21879722 DOI: 10.1021/bi2008933] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mitochondrial-nuclear communication is critical for maintaining mitochondrial activity under stress conditions. Adaptation of the mitochondrial-nuclear network to changes in the intracellular oxidation and reduction milieu is critical for the survival of retinal and retinal pigment epithelial (RPE) cells, in relation to their high oxygen demand and rapid metabolism. However, the generation and transmission of the mitochondrial signal to the nucleus remain elusive. Previously, our in vivo study revealed that prohibitin is upregulated in the retina, but downregulated in RPE cells in the aging and diabetic model. In this study, the functional role of prohibitin in the retina and RPE cells was examined using biochemical methods, including a lipid binding assay, two-dimensional gel electrophoresis, immunocytochemistry, Western blotting, and a knockdown approach. Protein depletion by siRNA characterized prohibitin as an anti-apoptotic molecule in mitochondria, while the lipid binding assay demonstrated subcellular communication between mitochondria and the nucleus under oxidative stress. The changes in the expression and localization of mitochondrial prohibitin triggered by reactive oxygen species are crucial for mitochondrial integrity. We propose that prohibitin shuttles between mitochondria and the nucleus as an anti-apoptotic molecule and a transcriptional regulator in a stress environment in the retina and RPE cells.
Collapse
Affiliation(s)
- Srinivas R Sripathi
- Biological Sciences, Michigan Technological University, Houghton, Michigan 49931, United States
| | | | | | | | | | | | | |
Collapse
|
50
|
Wang S, Kamat A, Pergola P, Swamy A, Tio F, Cusi K. Metabolic factors in the development of hepatic steatosis and altered mitochondrial gene expression in vivo. Metabolism 2011; 60:1090-9. [PMID: 21310443 DOI: 10.1016/j.metabol.2010.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 12/08/2010] [Accepted: 12/08/2010] [Indexed: 12/19/2022]
Abstract
The objective of the study was to understand the role in vivo of elevated plasma free fatty acids (FFA), insulin, and glucose levels in the development of steatosis and altered mitochondrial gene/protein expression. We studied 4 groups of Sprague-Dawley rats: (1) high-fat diet (HFD), (2) high-dose streptozotocin-induced diabetes (T1DM), (3) low-dose streptozotocin-induced diabetic rats on an HFD (T2DM), and (4) controls. Liver histology and expression of genes/proteins related to mitochondrial fatty acid oxidation and biogenesis were analyzed. Despite an attempt to compensate by increasing expression of genes of fatty acid oxidation (carnitine palmitoyl transferase-1/medium chain acyl-CoA dehydrogenase), the HFD and diabetic groups developed marked steatosis and suffered a significant reduction in mitochondrial biogenesis gene expression (nuclear respiratory factor 1/transcriptional factor A, mitochondrial). In T2DM rats, the combination of high glucose and FFA unexpectedly did not lead to greater fat accumulation than HFD alone. Greater steatosis in HFD vs T2DM (P < .001) correlated with impairment in the gene expression of PPAR-α (ie, fatty acid oxidation) and PGC1α, a major coactivator for mitochondrial biogenesis. Steatosis was not severe in insulin-deficient T1DM rats despite very elevated FFA and glucose levels. Increased carnitine palmitoyl transferase-1/medium chain acyl-CoA dehydrogenase/PPAR-α gene expression suggested inadequate adaptation to high FFA in both T1DM/T2DM rats. Hyperinsulinemia combined with elevated FFA is the key metabolic factor driving hepatic lipogenesis in vivo (HFD rats). Mitochondrial biogenesis (nuclear respiratory factor 1; transcriptional factor A, mitochondrial) is highly susceptible to FFA-induced steatosis. In contrast, hyperglycemia does not have an additive effect (T2DM) and leads to only a modest degree of steatosis in the absence of hyperinsulinemia, even when FFA are extremely elevated as in T1DM rats.
Collapse
Affiliation(s)
- Shaoyun Wang
- The University of Texas Health Science Center at San Antonio, TX 78229-3900, USA
| | | | | | | | | | | |
Collapse
|