101
|
Zhang J, Bai KW, He J, Niu Y, Lu Y, Zhang L, Wang T. Curcumin attenuates hepatic mitochondrial dysfunction through the maintenance of thiol pool, inhibition of mtDNA damage, and stimulation of the mitochondrial thioredoxin system in heat-stressed broilers. J Anim Sci 2018; 96:867-879. [PMID: 29566233 DOI: 10.1093/jas/sky009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 03/06/2018] [Indexed: 12/18/2022] Open
Abstract
The aim of this study was to investigate the effects of dietary curcumin supplementation on the performance, mitochondrial redox system, mitochondrial DNA (mtDNA) integrity, and antioxidant-related gene expression in the liver of broiler chickens after heat stress treatment. At day 21, a total of 400 Arbor Acres broiler chickens with similar body weight (BW) were divided into 5 groups with 8 replicates per group and then reared either at a normal temperature (22 ± 1 °C) or at a high ambient temperature (34 ± 1 °C for 8 h and 22 ± 1 °C for the remaining time) for 20 d. Broilers in the 5 groups were fed a basal diet at a normal temperature (NT group) and a basal diet with 0, 50, 100, and 200 mg/kg curcumin at a high ambient temperature (HT, CUR50, CUR100, and CUR200 groups), respectively. The serum and liver samples were analyzed for the parameters related to hepatic damage, mitochondrial function, and redox status. The results showed that the G:F was increased in the CUR50 and CUR100 groups, and the final BW was increased in CUR100 group in comparison with the HT group (P < 0.05). When compared with those in the HT group, both serum aspartate and alanine aminotransferase activities were decreased in the curcumin-supplemented groups (P < 0.05). Curcumin decreased the reactive oxygen species (ROS) production but increased the mitochondrial membrane potential in the hepatocytes of the broilers after heat stress (P < 0.05). The broilers in curcumin-supplemented groups had lower malondialdehyde and protein carbonyl concentrations as well as greater thiol concentrations (P < 0.05). The mitochondrial manganese superoxide dismutase (MnSOD) activity in the liver was increased (P < 0.05) in the CUR100 group compared with the HT group. Compared with the heat-stressed broilers, the broilers that were fed curcumin had greater (P < 0.05) mtDNA copy number and ATP concentrations than those in the HT group. Curcumin supplementation attenuated the depression of the thioredoxin 2 and peroxiredoxin-3 gene expressions (P < 0.05). The MnSOD gene expression was increased in the CUR100 and CUR200 groups, and the thioredoxin reductase 2 gene expression was increased in the CUR50 group in comparison with the HT group (P < 0.05). In conclusion, curcumin mitigated the mitochondrial dysfunction in heat-stressed broilers, as evidenced by the suppression of the ROS burst, the maintenance of the thiol pool and mtDNA content, and the enhanced mitochondrial antioxidant gene expression.
Collapse
Affiliation(s)
- Jingfei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kai Wen Bai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jintian He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yu Niu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yuan Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
102
|
Hood WR, Zhang Y, Mowry AV, Hyatt HW, Kavazis AN. Life History Trade-offs within the Context of Mitochondrial Hormesis. Integr Comp Biol 2018; 58:567-577. [PMID: 30011013 PMCID: PMC6145418 DOI: 10.1093/icb/icy073] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Evolutionary biologists have been interested in the negative interactions among life history traits for nearly a century, but the mechanisms that would create this negative interaction remain poorly understood. One variable that has emerged as a likely link between reproductive effort and longevity is oxidative stress. Specifically, it has been proposed that reproduction generates free radicals that cause oxidative stress and, in turn, oxidative stress damages cellular components and accelerates senescence. We propose that there is limited support for the hypothesis because reactive oxygen species (ROS), the free radicals implicated in oxidative damage, are not consistently harmful. With this review, we define the hormetic response of mitochondria to ROS, termed mitochondrial hormesis, and describe how to test for a mitohormetic response. We interpret existing data using our model and propose that experimental manipulations will further improve our knowledge of this response. Finally, we postulate how the mitohormetic response curve applies to variation in animal performance and longevity.
Collapse
Affiliation(s)
- W R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Y Zhang
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - A V Mowry
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
- Product Development, Stimlabs, Roswell, GA 30076, USA
| | - H W Hyatt
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | - A N Kavazis
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
103
|
Chimienti G, Picca A, Sirago G, Fracasso F, Calvani R, Bernabei R, Russo F, Carter CS, Leeuwenburgh C, Pesce V, Marzetti E, Lezza AMS. Increased TFAM binding to mtDNA damage hot spots is associated with mtDNA loss in aged rat heart. Free Radic Biol Med 2018; 124:447-453. [PMID: 29969715 PMCID: PMC6319621 DOI: 10.1016/j.freeradbiomed.2018.06.041] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 06/29/2018] [Indexed: 02/07/2023]
Abstract
The well-known age-related mitochondrial dysfunction deeply affects heart because of the tissue's large dependence on mitochondrial ATP provision. Our study revealed in aged rat heart a significant 25% decrease in mtDNA relative content, a significant 29% increase in the 4.8 Kb mtDNA deletion relative content, and a significant inverse correlation between such contents as well as a significant 38% decrease in TFAM protein amount. The TFAM-binding activity to specific mtDNA regions increased at those encompassing the mtDNA replication origins, D-loop and Ori-L. The same mtDNA regions were screened for different kinds of oxidative damage, namely Single Strand Breaks (SSBs), Double Strand Breaks (DSBs), abasic sites (AP sites) and oxidized bases as 7,8-dihydro-8-oxoguanine (8oxoG). A marked increase in the relative content of mtDNA strand damage (SSBs, DSBs and AP sites) was found in the D-loop and Ori-L regions in the aged animals, unveiling for the first time in vivo an age-related, non-stochastic accumulation of oxidative lesions in these two regions that appear as hot spots of mtDNA damage. The use of Formamidopyrimidine glycosylase (Fpg) demonstrated also a significant age-related accumulation of oxidized purines particularly in the D-loop and Ori-L regions. The detected increased binding of TFAM to the mtDNA damage hot spots in aged heart suggests a link between TFAM binding to mtDNA and loss of mitochondrial genome likely through hindrance of repair processes.
Collapse
Affiliation(s)
- Guglielmina Chimienti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Anna Picca
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Teaching Hospital "Agostino Gemelli", Rome, Italy
| | - Giuseppe Sirago
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Flavio Fracasso
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Riccardo Calvani
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Teaching Hospital "Agostino Gemelli", Rome, Italy
| | - Roberto Bernabei
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Teaching Hospital "Agostino Gemelli", Rome, Italy
| | - Francesco Russo
- Laboratory of Nutritional Pathophysiology, National Institute of Digestive Diseases - I.R.C.C.S. "Saverio de Bellis", Castellana Grotte, Italy
| | - Christy S Carter
- Department of Aging and Geriatric Research, Institute on Aging, Division of Biology of Aging, University of Florida, 2004 Mowry Rd, Gainesville, FL 32611, USA
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, Institute on Aging, Division of Biology of Aging, University of Florida, 2004 Mowry Rd, Gainesville, FL 32611, USA
| | - Vito Pesce
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Teaching Hospital "Agostino Gemelli", Rome, Italy
| | - Angela Maria Serena Lezza
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy.
| |
Collapse
|
104
|
Nissanka N, Bacman SR, Plastini MJ, Moraes CT. The mitochondrial DNA polymerase gamma degrades linear DNA fragments precluding the formation of deletions. Nat Commun 2018; 9:2491. [PMID: 29950568 PMCID: PMC6021392 DOI: 10.1038/s41467-018-04895-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 05/25/2018] [Indexed: 02/03/2023] Open
Abstract
Double-strand breaks in the mitochondrial DNA (mtDNA) result in the formation of linear fragments that are rapidly degraded. However, the identity of the nuclease(s) performing this function is not known. We found that the exonuclease function of the mtDNA polymerase gamma (POLG) is required for this rapid degradation of mtDNA fragments. POLG is recruited to linearized DNA fragments in an origin of replication-independent manner. Moreover, in the absence of POLG exonuclease activity, the prolonged existence of mtDNA linear fragments leads to increased levels of mtDNA deletions, which have been previously identified in the mutator mouse, patients with POLG mutations and normal aging. Mitochondrial DNA fragments are rapidly degraded when double strand breaks occur. Here the authors reveal that the exonuclease activity of polymerase gamma is important for efficient degradation of these fragments and to avoid formation of large deletions.
Collapse
Affiliation(s)
- Nadee Nissanka
- Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Sandra R Bacman
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Melanie J Plastini
- Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Carlos T Moraes
- Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA. .,Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
105
|
Ness KK, Kirkland JL, Gramatges MM, Wang Z, Kundu M, McCastlain K, Li-Harms X, Zhang J, Tchkonia T, Pluijm SMF, Armstrong GT. Premature Physiologic Aging as a Paradigm for Understanding Increased Risk of Adverse Health Across the Lifespan of Survivors of Childhood Cancer. J Clin Oncol 2018; 36:2206-2215. [PMID: 29874132 DOI: 10.1200/jco.2017.76.7467] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The improvement in survival of childhood cancer observed across the past 50 years has resulted in a growing acknowledgment that simply extending the lifespan of survivors is not enough. It is incumbent on both the cancer research and the clinical care communities to also improve the health span of survivors. It is well established that aging adult survivors of childhood cancer are at increased risk of chronic health conditions, relative to the general population. However, as the first generation of survivors age into their 50s and 60s, it has become increasingly evident that this population is also at risk of early onset of physiologic aging. Geriatric measures have uncovered evidence of reduced strength and speed and increased fatigue, all components of frailty, among survivors with a median age of 33 years, which is similar to adults older than 65 years of age in the general population. Furthermore, frailty in survivors independently increased the risk of morbidity and mortality. Although there has been a paucity of research investigating the underlying biologic mechanisms for advanced physiologic age in survivors, results from geriatric populations suggest five biologically plausible mechanisms that may be potentiated by exposure to cancer therapies: increased cellular senescence, reduced telomere length, epigenetic modifications, somatic mutations, and mitochondrial DNA infidelity. There is now a critical need for research to elucidate the biologic mechanisms of premature aging in survivors of childhood cancer. This research could pave the way for new frontiers in the prevention of these life-changing outcomes.
Collapse
Affiliation(s)
- Kirsten K Ness
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - James L Kirkland
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Maria Monica Gramatges
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Zhaoming Wang
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Mondira Kundu
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Kelly McCastlain
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Xiujie Li-Harms
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jinghui Zhang
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Tamar Tchkonia
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Saskia Martine Francesca Pluijm
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Gregory T Armstrong
- Kirsten K. Ness, Zhaoming Wang, Mondira Kundu, Kelly McCastlain, Xiujie Li-Harms, Jinghui Zhang, and Gregory T. Armstrong, St. Jude Children's Research Hospital, Memphis, TN; James L. Kirkland and Tamar Tchkonia, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; Maria Monica Gramatges, Texas Children's Cancer and Hematology Centers at Baylor College of Medicine, Houston, TX; and Saskia Martine Francesca Pluijm, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
106
|
Cao S, Zhang Q, Wang C, Wu H, Jiao L, Hong Q, Hu C. LPS challenge increased intestinal permeability, disrupted mitochondrial function and triggered mitophagy of piglets. Innate Immun 2018; 24:221-230. [PMID: 29642727 PMCID: PMC6830921 DOI: 10.1177/1753425918769372] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/07/2018] [Accepted: 03/15/2018] [Indexed: 12/21/2022] Open
Abstract
Here we investigated the influence of LPS-induced gut injury on antioxidant homeostasis, mitochondrial (mt) function and the level of mitophagy in piglets. The results showed that LPS-induced intestinal injury decreased the transepithelial electrical resistance, increased the paracellular permeability of F1TC dextran 4 kDa, and decreased the expression of claudin-1, occludin and zonula occludens-1 in the jejunum compared with the control group. LPS decreased the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and increased the content of malondialdehyde in the jejunum. Meanwhile, the expression of SOD-related genes ( Cu/Zn-SOD, Mn-SOD) and GSH-Px-related genes ( GPX-1, GPX-4) declined in LPS-challenged pigs compared with the control. LPS also increased TNF-α, IL-6, IL-8 and IL-1β mRNA expression. LPS induced mt dysfunction, as demonstrated by increased reactive oxygen species production and decreased membrane potential of intestinal mitochondria, intestinal content of mt DNA and activities of the intestinal mt respiratory chain. Furthermore, LPS induced an increase in expression of mitophagy related proteins, PTEN-induced putative kinase (PINK1) and Parkin in the intestinal mitochondria, as well as an enhancement of the ratio of light chain 3-II (LC3-II) to LC3-I content in the jejunal mucosa. These results suggested that LPS-induced intestinal injury accompanied by disrupted antioxidant homeostasis, caused mt dysfunction and triggered mitophagy.
Collapse
Affiliation(s)
- Shuting Cao
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Qianhui Zhang
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - ChunChun Wang
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Huan Wu
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Lefei Jiao
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Qihua Hong
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| | - Caihong Hu
- Animal Science College, Zhejiang University, The Key
Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou,
China
| |
Collapse
|
107
|
Miller VJ, Villamena FA, Volek JS. Nutritional Ketosis and Mitohormesis: Potential Implications for Mitochondrial Function and Human Health. J Nutr Metab 2018; 2018:5157645. [PMID: 29607218 PMCID: PMC5828461 DOI: 10.1155/2018/5157645] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023] Open
Abstract
Impaired mitochondrial function often results in excessive production of reactive oxygen species (ROS) and is involved in the etiology of many chronic diseases, including cardiovascular disease, diabetes, neurodegenerative disorders, and cancer. Moderate levels of mitochondrial ROS, however, can protect against chronic disease by inducing upregulation of mitochondrial capacity and endogenous antioxidant defense. This phenomenon, referred to as mitohormesis, is induced through increased reliance on mitochondrial respiration, which can occur through diet or exercise. Nutritional ketosis is a safe and physiological metabolic state induced through a ketogenic diet low in carbohydrate and moderate in protein. Such a diet increases reliance on mitochondrial respiration and may, therefore, induce mitohormesis. Furthermore, the ketone β-hydroxybutyrate (BHB), which is elevated during nutritional ketosis to levels no greater than those resulting from fasting, acts as a signaling molecule in addition to its traditionally known role as an energy substrate. BHB signaling induces adaptations similar to mitohormesis, thereby expanding the potential benefit of nutritional ketosis beyond carbohydrate restriction. This review describes the evidence supporting enhancement of mitochondrial function and endogenous antioxidant defense in response to nutritional ketosis, as well as the potential mechanisms leading to these adaptations.
Collapse
Affiliation(s)
- Vincent J. Miller
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Frederick A. Villamena
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jeff S. Volek
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
108
|
|
109
|
Mitochondrial DNA Double-Strand Breaks in Oligodendrocytes Cause Demyelination, Axonal Injury, and CNS Inflammation. J Neurosci 2017; 37:10185-10199. [PMID: 28931570 DOI: 10.1523/jneurosci.1378-17.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/21/2017] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial dysfunction has been implicated in the pathophysiology of neurodegenerative disorders, including multiple sclerosis (MS). To date, the investigation of mitochondrial dysfunction in MS has focused exclusively on neurons, with no studies exploring whether dysregulation of mitochondrial bioenergetics and/or genetics in oligodendrocytes might be associated with the etiopathogenesis of MS and other demyelinating syndromes. To address this question, we established a mouse model where mitochondrial DNA (mtDNA) double-strand breaks (DSBs) were specifically induced in myelinating oligodendrocytes (PLP:mtPstI mice) by expressing a mitochondrial-targeted endonuclease, mtPstI, starting at 3 weeks of age. In both female and male mice, DSBs of oligodendroglial mtDNA caused impairment of locomotor function, chronic demyelination, glial activation, and axonal degeneration, which became more severe with time of induction. In addition, after short transient induction of mtDNA DSBs, PLP:mtPstI mice showed an exacerbated response to experimental autoimmune encephalomyelitis. Together, our data demonstrate that mtDNA damage can cause primary oligodendropathy, which in turn triggers demyelination, proving PLP:mtPstI mice to be a useful tool to study the pathological consequences of mitochondrial dysfunction in oligodendrocytes. In addition, the demyelination and axonal loss displayed by PLP:mtPstI mice recapitulate some of the key features of chronic demyelinating syndromes, including progressive MS forms, which are not accurately reproduced in the models currently available. For this reason, the PLP:mtPstI mouse represents a unique and much needed platform for testing remyelinating therapies.SIGNIFICANCE STATEMENT In this study, we show that oligodendrocyte-specific mitochondrial DNA double-strand breaks in PLP:mtPstI mice cause oligodendrocyte death and demyelination associated with axonal damage and glial activation. Hence, PLP:mtPstI mice represent a unique tool to study the pathological consequences of mitochondrial dysfunction in oligodendrocytes, as well as an ideal platform to test remyelinating and neuroprotective agents.
Collapse
|
110
|
Melvin RG, Ballard JWO. Cellular and population level processes influence the rate, accumulation and observed frequency of inherited and somatic mtDNA mutations. Mutagenesis 2017; 32:323-334. [PMID: 28521046 DOI: 10.1093/mutage/gex004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondria are found in all animals and have the unique feature of containing multiple copies of their own small, circular DNA genome (mtDNA). The rate and pattern of mutation accumulation in the mtDNA are influenced by molecular, cellular and population level processes. We distinguish between inherited and somatic mtDNA mutations and review evidence for the often-made assumption that mutations accumulate at a higher rate in mtDNA than in nuclear DNA (nDNA). We conclude that the whole genome mutation accumulation rate is higher for mtDNA than for nDNA but include the caveat that rates overlap considerably between the individual mtDNA- and nDNA-encoded genes. Next, we discuss the postulated causal mechanisms for the high rate of mtDNA mutation accumulation in both inheritance and in somatic cells. Perhaps unexpectedly, mtDNA is resilient to many mutagens of nDNA but is prone to errors of replication. We then consider the influence of maternal inheritance, recombination and selection on the observed accumulation pattern of inherited mtDNA mutations. Finally, we discuss environmental influences of temperature and diet on the observed frequency of inherited and somatic mtDNA mutations. We conclude that it is necessary to understand the cellular processes to fully interpret the pattern of mutations and how they influence our interpretations of evolution and disease.
Collapse
Affiliation(s)
- Richard G Melvin
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - John William O Ballard
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
111
|
Nicassio L, Fracasso F, Sirago G, Musicco C, Picca A, Marzetti E, Calvani R, Cantatore P, Gadaleta MN, Pesce V. Dietary supplementation with acetyl-l-carnitine counteracts age-related alterations of mitochondrial biogenesis, dynamics and antioxidant defenses in brain of old rats. Exp Gerontol 2017; 98:99-109. [PMID: 28807823 DOI: 10.1016/j.exger.2017.08.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/31/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
We previously reported the ability of dietary supplementation with acetyl-l-carnitine (ALCAR) to prevent age-related decreases of mitochondrial biogenesis in skeletal muscle and liver of old rats. Here, we investigate the effects of ALCAR supplementation in cerebral hemispheres and cerebellum of old rats by analyzing several parameters linked to mitochondrial biogenesis, mitochondrial dynamics and antioxidant defenses. We measured the level of the coactivators PGC-1α and PGC-1β and of the factors regulating mitochondrial biogenesis, finding an age-related decrease of PGC-1β, whereas PGC-1α level was unvaried. Twenty eight-month old rats supplemented with ALCAR for one and two months showed increased levels of both factors. Accordingly, the expression of the two transcription factors NRF-1 and TFAM followed the same trend of PGC-1β. The level of mtDNA, ND1 and the activity of citrate synthase, were decreased with aging and increased following ALCAR treatment. Furthermore, ALCAR counteracted the age-related increase of deleted mtDNA. We also analyzed the content of proteins involved in mitochondrial dynamics (Drp1, Fis1, OPA1 and MNF2) and found an age-dependent increase of MFN2 and of the long form of OPA1. ALCAR treatment restored the content of the two proteins to the level of the young rats. No changes with aging and ALCAR were observed for Drp1 and Fis1. ALCAR reduced total cellular levels of oxidized PRXs and counteracted the age-related decrease of PRX3 and SOD2. Overall, our findings indicate a systemic positive effect of ALCAR dietary treatment and a tissue specific regulation of mitochondrial homeostasis in brain of old rats. Moreover, it appears that ALCAR acts as a nutrient since in most cases its effects were almost completely abolished one month after treatment suspension. Dietary supplementation of old rats with this compound seems a valuable approach to prevent age-related mitochondrial dysfunction and might ultimately represent a strategy to delay age-associated negative consequences in mitochondrial homeostasis.
Collapse
Affiliation(s)
- Luigi Nicassio
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy
| | - Flavio Fracasso
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy
| | - Giuseppe Sirago
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy
| | - Clara Musicco
- Institute of Biomembranes and Bioenergetics (IBBE), National Research Council of Italy (CNR), Bari, Italy
| | - Anna Picca
- Department of Geriatrics, Neuroscience and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Rome, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Neuroscience and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Rome, Italy
| | - Riccardo Calvani
- Department of Geriatrics, Neuroscience and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Rome, Italy
| | - Palmiro Cantatore
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy
| | - Maria Nicola Gadaleta
- Institute of Biomembranes and Bioenergetics (IBBE), National Research Council of Italy (CNR), Bari, Italy
| | - Vito Pesce
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "A. Moro", Bari, Italy.
| |
Collapse
|
112
|
Origins of mtDNA mutations in ageing. Essays Biochem 2017; 61:325-337. [PMID: 28698307 DOI: 10.1042/ebc20160090] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/24/2017] [Accepted: 05/26/2017] [Indexed: 12/21/2022]
Abstract
MtDNA mutations are one of the hallmarks of ageing and age-related diseases. It is well established that somatic point mutations accumulate in mtDNA of multiple organs and tissues with increasing age and heteroplasmy is universal in mammals. However, the origin of these mutations remains controversial. The long-lasting hypothesis stating that mtDNA mutations emanate from oxidative damage via a self-perpetuating mechanism has been extensively challenged in recent years. Contrary to this initial ascertainment, mtDNA appears to be well protected from action of reactive oxygen species (ROS) through robust protein coating and endomitochondrial microcompartmentalization. Extensive development of scrupulous high-throughput DNA sequencing methods suggests that an imperfect replication process, rather than oxidative lesions are the main sources of mtDNA point mutations, indicating that mtDNA polymerase γ (POLG) might be responsible for the majority of mtDNA mutagenic events. Here, we summarize the recent knowledge in prevention and defence of mtDNA oxidative lesions and discuss the plausible mechanisms of mtDNA point mutation generation and fixation.
Collapse
|
113
|
Abstract
: The increased prevalence of age-related comorbidities and mortality is worrisome in ageing HIV-infected patients. Here, we aim to analyse the different ageing mechanisms with regard to HIV infection. Ageing results from the time-dependent accumulation of random cellular damage. Epigenetic modifications and mitochondrial DNA haplogroups modulate ageing. In antiretroviral treatment-controlled patients, epigenetic clock appears to be advanced, and some haplogroups are associated with HIV infection severity. Telomere shortening is enhanced in HIV-infected patients because of HIV and some nucleoside analogue reverse transcriptase inhibitors. Mitochondria-related oxidative stress and mitochondrial DNA mutations are increased during ageing and also by some nucleoside analogue reverse transcriptase inhibitors. Overall, increased inflammation or 'inflammageing' is a major driver of ageing and could result from cell senescence with secreted proinflammatory mediators, altered gut microbiota, and coinfections. In HIV-infected patients, the level of inflammation and innate immunity activation is enhanced and related to most comorbidities and to mortality. This status could result, in addition to age, from the virus itself or viral protein released from reservoirs, from HIV-enhanced gut permeability and dysbiosis, from antiretroviral treatment, from frequent cytomegalovirus and hepatitis C virus coinfections, and also from personal and environmental factors, as central fat accumulation or smoking. Adaptive immune activation and immunosenescence are associated with comorbidities and mortality in the general population but are less predictive in HIV-infected patients. Biomarkers to evaluate ageing in HIV-infected patients are required. Numerous systemic or cellular inflammatory, immune activation, oxidative stress, or senescence markers can be tested in serum or peripheral blood mononuclear cells. The novel European Study to Establish Biomarkers of Human Ageing MARK-AGE algorithm, evaluating the biological age, is currently assessed in HIV-infected patients and reveals an advanced biological age. Some enhanced inflammatory or innate immune activation markers are interesting but still not validated for the patient's follow-up. To be able to assess patients' biological age is an important objective to improve their healthspan.
Collapse
|
114
|
Impact of Aging and Exercise on Mitochondrial Quality Control in Skeletal Muscle. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3165396. [PMID: 28656072 PMCID: PMC5471566 DOI: 10.1155/2017/3165396] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/03/2017] [Indexed: 12/17/2022]
Abstract
Mitochondria are characterized by its pivotal roles in managing energy production, reactive oxygen species, and calcium, whose aging-related structural and functional deteriorations are observed in aging muscle. Although it is still unclear how aging alters mitochondrial quality and quantity in skeletal muscle, dysregulation of mitochondrial biogenesis and dynamic controls has been suggested as key players for that. In this paper, we summarize current understandings on how aging regulates muscle mitochondrial biogenesis, while focusing on transcriptional regulations including PGC-1α, AMPK, p53, mtDNA, and Tfam. Further, we review current findings on the muscle mitochondrial dynamic systems in aging muscle: fusion/fission, autophagy/mitophagy, and protein import. Next, we also discuss how endurance and resistance exercises impact on the mitochondrial quality controls in aging muscle, suggesting possible effective exercise strategies to improve/maintain mitochondrial health.
Collapse
|
115
|
Li H, Shen L, Hu P, Huang R, Cao Y, Deng J, Yuan W, Liu D, Yang J, Gu H, Bai Y. Aging-associated mitochondrial DNA mutations alter oxidative phosphorylation machinery and cause mitochondrial dysfunctions. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2266-2273. [PMID: 28559044 DOI: 10.1016/j.bbadis.2017.05.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Our previous study generated a series of cybrids containing mitochondria of synaptosomes from mice at different ages. The following functional analysis on these cybrids revealed an age-dependent decline of mitochondrial function. To understand the underlying mechanisms that contribute to the age-related mitochondrial dysfunction, we focused on three cybrids carrying mitochondria derived from synaptosomes of the old mice that exhibited severe respiratory deficiencies. In particular, we started with a comprehensive analysis of mitochondrial genome by high resolution, high sensitive deep sequencing method. Compared with young control, we detected a significant accumulation of heteroplasmic mtDNA mutations. These mutations included six alterations in main control region that has been shown to regulate overall gene-expression, and four alterations in protein coding region, two of which led to significant changes in complex I subunit ND5 and complex III subunit CytB. Interestingly, a reduced mtDNA-encoded protein synthesis was associated with the changes in the main control region. Likewise, mutations in ND5 and CytB were associated with defects in assembly of respiratory complexes. Altogether, the identified age-dependent accumulation of mtDNA mutations in mouse brain likely contributes to the decline in mitochondrial function.
Collapse
Affiliation(s)
- Hongzhi Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Luxi Shen
- Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Peiqing Hu
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Rong Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ying Cao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Janice Deng
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Weihua Yuan
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Danhui Liu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jifeng Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Haihua Gu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yidong Bai
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
116
|
Mehta SR, Pérez-Santiago J, Hulgan T, Day TRC, Barnholtz-Sloan J, Gittleman H, Letendre S, Ellis R, Heaton R, Patton S, Suben JD, Franklin D, Rosario D, Clifford DB, Collier AC, Marra CM, Gelman BB, McArthur J, McCutchan A, Morgello S, Simpson D, Connor J, Grant I, Kallianpur A. Cerebrospinal fluid cell-free mitochondrial DNA is associated with HIV replication, iron transport, and mild HIV-associated neurocognitive impairment. J Neuroinflammation 2017; 14:72. [PMID: 28359324 PMCID: PMC5374652 DOI: 10.1186/s12974-017-0848-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/21/2017] [Indexed: 12/19/2022] Open
Abstract
Background Mitochondria are abundant organelles critical for energy metabolism and brain function. Mitochondrial DNA (mtDNA), released during cellular injury and as part of the innate immune response to viral pathogens, contains CpG motifs that act as TLR-9 ligands. We investigated relationships between cerebrospinal fluid (CSF) cell-free mtDNA levels and HIV viral load (VL), biomarkers of inflammation and iron transport, and neurocognitive (NC) function in the CNS HIV Antiretroviral Therapy Effects Research (CHARTER) cohort. Methods We quantified cell-free mtDNA in CSF by droplet digital PCR in 332 CHARTER participants who underwent comprehensive neuropsychiatric evaluation. NC performance was assessed using the global deficit score (GDS) as either a continuous or a binary measure (GDS ≥ 0.5, impaired vs. GDS < 0.5, unimpaired). CSF, clinical, and biomarker data from the earliest available time point were analyzed. Cell-free mtDNA associations with CSF inflammation and iron-related biomarkers [CXCL10, IL-6, IL-8, TNF-a, transferrin (TF), ceruloplasmin (CP), and vascular endothelial growth factor (VEGF)], VL, and GDS were evaluated by multivariable regression. Results CSF cell-free mtDNA levels were significantly lower in participants with undetectable (vs. detectable) VL in either plasma (p < 0.001) or CSF (p < 0.001) and in those on antiretroviral therapy (ART; p < 0.001). Participants on ART with undetectable VL in both CSF and plasma had lower mtDNA levels than those with detectable VL in both compartments (p = 0.001). Higher mtDNA levels were observed in participants in the highest vs. lowest tertile (T3 vs. T1) of CSF CXCL10 (T3 vs. T1, p < 0.001) and TNF-a (T3 vs. T1, p < 0.05) in unadjusted analyses. MtDNA levels also correlated with CSF leukocyte count. After adjusting for CSF leukocyte count and VL, mtDNA levels were also associated with other inflammation- and iron-related biomarkers in CSF, including TF (T3 vs. T1, p < 0.05) and CP (T3 vs. T1, p < 0.05). With additional correction for ART use, mtDNA was also negatively associated with CSF VEGF (p < 0.05) and IL-6 (p = 0.05). We observed no associations of CSF mtDNA levels with age or GDS-defined NC impairment. Conclusions CSF cell-free mtDNA levels were associated with HIV RNA and ART status, as well as with biomarkers of iron transport and VEGF, a growth factor with known effects on mitochondrial integrity and autophagy. CSF mtDNA may be a biomarker of iron dysregulation and/or neuroinflammation during HIV infection. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0848-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanjay R Mehta
- Department of Medicine, University of California-San Diego, San Diego, CA, USA. .,Department of Medicine, San Diego Veterans Affairs Medical Center, San Diego, CA, USA.
| | | | - Todd Hulgan
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University, Nashville, TN, USA
| | - Tyler R C Day
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Jill Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Haley Gittleman
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Scott Letendre
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Ronald Ellis
- Department of Neurology, University of California-San Diego, San Diego, CA, USA
| | - Robert Heaton
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Stephanie Patton
- Department of Neurosurgery, Pennsylvania State/Hershey College of Medicine, Hershey, PA, USA
| | - Jesse D Suben
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Donald Franklin
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Debralee Rosario
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - David B Clifford
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Ann C Collier
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Justin McArthur
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Allen McCutchan
- Department of Medicine, University of California-San Diego, San Diego, CA, USA
| | - Susan Morgello
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Simpson
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James Connor
- Department of Neurosurgery, Pennsylvania State/Hershey College of Medicine, Hershey, PA, USA
| | - Igor Grant
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Asha Kallianpur
- Genomic Medicine Institute/Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
117
|
NLRP3 Deficiency Attenuates Renal Fibrosis and Ameliorates Mitochondrial Dysfunction in a Mouse Unilateral Ureteral Obstruction Model of Chronic Kidney Disease. Mediators Inflamm 2017; 2017:8316560. [PMID: 28348462 PMCID: PMC5350413 DOI: 10.1155/2017/8316560] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/09/2017] [Indexed: 12/15/2022] Open
Abstract
Background and Aims. The nucleotide-binding domain and leucine-rich repeat containing PYD-3 (NLRP3) inflammasome has been implicated in the pathogenesis of chronic kidney disease (CKD); however, its exact role in glomerular injury and tubulointerstitial fibrosis is still undefined. The present study was performed to identify the function of NLRP3 in modulating renal injury and fibrosis and the potential involvement of mitochondrial dysfunction in the murine unilateral ureteral obstruction (UUO) model of CKD. Methods. Employing wild-type (WT) and NLRP3−/− mice with or without UUO, we evaluated renal structure, tissue injury, and mitochondrial ultrastructure, as well as expression of some vital molecules involved in the progression of fibrosis, apoptosis, inflammation, and mitochondrial dysfunction. Results. The severe glomerular injury and tubulointerstitial fibrosis induced in WT mice by UUO was markedly attenuated in NLRP3−/− mice as evidenced by blockade of extracellular matrix deposition, decreased cell apoptosis, and phenotypic alterations. Moreover, NLRP3 deletion reversed UUO-induced impairment of mitochondrial morphology and function. Conclusions. NLRP3 deletion ameliorates mitochondrial dysfunction and alleviates renal fibrosis in a murine UUO model of CKD.
Collapse
|
118
|
Kramer AC, Weber J, Zhang Y, Tolar J, Gibbens YY, Shevik M, Lund TC. TP53 Modulates Oxidative Stress in Gata1 + Erythroid Cells. Stem Cell Reports 2017; 8:360-372. [PMID: 28132886 PMCID: PMC5312256 DOI: 10.1016/j.stemcr.2016.12.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 01/28/2023] Open
Abstract
Metabolism of oxidative stress is necessary for cellular survival. We have previously utilized the zebrafish as a model of the oxidative stress response. In this study, we found that gata1-expressing erythroid cells contributed to a significant proportion of total-body oxidative stress when animals were exposed to a strong pro-oxidant. RNA-seq of zebrafish under oxidative stress revealed the induction of tp53. Zebrafish carrying tp53 with a mutation in its DNA-binding domain were acutely sensitive to pro-oxidant exposure and displayed significant reactive oxygen species (ROS) and tp53-independent erythroid cell death resulting in an edematous phenotype. We found that a major contributing factor to ROS was increased basal mitochondrial respiratory rate without reserve. These data add to the concept that tp53, while classically a tumor suppressor and cell-cycle regulator, has additional roles in controlling cellular oxidative stress. Erythroid precursors contribute significantly to total ROS after oxidative challenge Tp53 is induced after pro-oxidant challenge Mutated tp53 is associated with an increased mitochondrial oxygen consumption rate Decreased mitochondrial reserve leads to overwhelming ROS and erythroid cell death
Collapse
Affiliation(s)
- Ashley C Kramer
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jenna Weber
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jakub Tolar
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ying Y Gibbens
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Margaret Shevik
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Troy C Lund
- Division of Pediatric Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
119
|
Soysal P, Isik AT, Carvalho AF, Fernandes BS, Solmi M, Schofield P, Veronese N, Stubbs B. Oxidative stress and frailty: A systematic review and synthesis of the best evidence. Maturitas 2017; 99:66-72. [PMID: 28364871 DOI: 10.1016/j.maturitas.2017.01.006] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Oxidative stress (OS) is associated with accelerated aging. Previous studies have suggested a possible relationship between OS and frailty but this association remains unclear. We conducted a systematic review to investigate potential interactions between OS and frailty. METHODS A systematic literature search of original reports providing data on 'OS and antioxidant' parameters and frailty was carried out across major electronic databases from inception until May 2016. Cross-sectional/case control and longitudinal studies reporting data on the association between frailty and anti-oxidants-OS biomarkers were considered for inclusion. Results were summarized with a synthesis based on the best evidence. RESULTS From 1856 hits, 8 studies (cross-sectional/case control) were included (N=6349; mean age of 75±12years; 56.4% females). Overall, there were 588 (=9.3%) frail, 3036 pre-frail (=47.8%), 40 (=0.6%) pre-frail/robust, and 2685 (=42.3%) robust subjects. Six cross-sectional/case control studies demonstrated that frailty was associated with an increase in peripheral OS biomarkers, including lipoprotein phospholipase A2 (1 study), isoprostanes (2 studies), malonaldehyde (2 studies), 8-hydroxy-20-deoxyguanosine (2 studies), derivate of reactive oxygen metabolites (2 studies), oxidized glutathione/glutathione (1 study), 4-hydroxy-2,3-nonenal (1 study), and protein carbonylation levels (1 study). In addition, preliminary evidence points to lower anti-oxidant parameters (vitamin C, E, α-tocopherol, biological anti-oxidant potential, total thiol levels) in frailty. CONCLUSION Frailty and pre-frailty appear to be associated with higher OS and possibly lower anti-oxidant parameters. However, due to the cross-sectional design, it is not possible to disentangle the directionality of the relationships observed. Thus, future high-quality and in particular longitudinal research is required to confirm or refute these relationships and to further elucidate pathophysiological mechanisms.
Collapse
Affiliation(s)
- Pinar Soysal
- Kayseri Education and Research Hospital, Geriatric Center, Kayseri, Turkey; Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London Box SE5 8AF, United Kingdom
| | - Ahmet Turan Isik
- Center for Aging Brain and Dementia, Department of Geriatric Medicine, Dokuz Eylul University, Faculty of Medicine, Izmir, Turkey; Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London Box SE5 8AF, United Kingdom
| | - Andre F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil; Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London Box SE5 8AF, United Kingdom
| | - Brisa S Fernandes
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, Geelong, Australia; Laboratory of Calcium Binding Proteins in the Central Nervous System, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London Box SE5 8AF, United Kingdom
| | - Marco Solmi
- Department of Neurosciences, University of Padova, Padova, Italy; National Health Care System, Padova Local Unit ULSS 17, Italy; Institute for Clinical Research and Education in Medicine, I.R.E.M., Padua, Italy; Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London Box SE5 8AF, United Kingdom
| | - Patricia Schofield
- Health, Social Care and Education, Anglia Ruskin University, Chelmsford, United Kingdom; Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London Box SE5 8AF, United Kingdom
| | - Nicola Veronese
- Institute for Clinical Research and Education in Medicine, I.R.E.M., Padua, Italy; Geriatrics Division, Department of Medicine-DIMED, University of Padova, Italy
| | - Brendon Stubbs
- Health, Social Care and Education, Anglia Ruskin University, Chelmsford, United Kingdom; Institute of Clinical Research and Education in Medicine (IREM), Padova, Italy; Physiotherapy Department, South London and Maudsley NHS Foundation Trust, Denmark Hill, London SE5 8AZ, United Kingdom; Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London Box SE5 8AF, United Kingdom.
| |
Collapse
|
120
|
Shiloh Y, Lederman HM. Ataxia-telangiectasia (A-T): An emerging dimension of premature ageing. Ageing Res Rev 2017; 33:76-88. [PMID: 27181190 DOI: 10.1016/j.arr.2016.05.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/02/2016] [Accepted: 05/10/2016] [Indexed: 12/28/2022]
Abstract
A-T is a prototype genome instability syndrome and a multifaceted disease. A-T leads to neurodegeneration - primarily cerebellar atrophy, immunodeficiency, oculocutaneous telangiectasia (dilated blood vessels), vestigial thymus and gonads, endocrine abnormalities, cancer predisposition and varying sensitivity to DNA damaging agents, particularly those that induce DNA double-strand breaks. With the recent increase in life expectancy of A-T patients, the premature ageing component of this disease is gaining greater awareness. The complex A-T phenotype reflects the ever growing number of functions assigned to the protein encoded by the responsible gene - the homeostatic protein kinase, ATM. The quest to thoroughly understand the complex A-T phenotype may reveal yet elusive ATM functions.
Collapse
|
121
|
Barzilai A, Schumacher B, Shiloh Y. Genome instability: Linking ageing and brain degeneration. Mech Ageing Dev 2017; 161:4-18. [DOI: 10.1016/j.mad.2016.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/23/2016] [Accepted: 03/26/2016] [Indexed: 02/06/2023]
|
122
|
CEDIKOVA M, PITULE P, KRIPNEROVA M, MARKOVA M, KUNCOVA J. Multiple Roles of Mitochondria in Aging Processes. Physiol Res 2016; 65:S519-S531. [DOI: 10.33549/physiolres.933538] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aging is a multifactorial process influenced by genetic factors, nutrition, and lifestyle. According to mitochondrial theory of aging, mitochondrial dysfunction is widely considered a major contributor to age-related processes. Mitochondria are both the main source and targets of detrimental reactions initiated in association with age-dependent deterioration of the cellular functions. Reactions leading to increased reactive oxygen species generation, mtDNA mutations, and oxidation of mitochondrial proteins result in subsequent induction of apoptotic events, impaired oxidative phosphorylation capacity, mitochondrial dynamics, biogenesis and autophagy. This review summarizes the major changes of mitochondria related to aging, with emphasis on mitochondrial DNA mutations, the role of the reactive oxygen species, and structural and functional changes of mitochondria.
Collapse
Affiliation(s)
| | | | | | | | - J. KUNCOVA
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
123
|
Vaiserman AM, Lushchak OV, Koliada AK. Anti-aging pharmacology: Promises and pitfalls. Ageing Res Rev 2016; 31:9-35. [PMID: 27524412 DOI: 10.1016/j.arr.2016.08.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/06/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022]
Abstract
Life expectancy has grown dramatically in modern times. This increase, however, is not accompanied by the same increase in healthspan. Efforts to extend healthspan through pharmacological agents targeting aging-related pathological changes are now in the spotlight of geroscience, the main idea of which is that delaying of aging is far more effective than preventing the particular chronic disorders. Currently, anti-aging pharmacology is a rapidly developing discipline. It is a preventive field of health care, as opposed to conventional medicine which focuses on treating symptoms rather than root causes of illness. A number of pharmacological agents targeting basic aging pathways (i.e., calorie restriction mimetics, autophagy inducers, senolytics etc.) are now under investigation. This review summarizes the literature related to advances, perspectives and challenges in the field of anti-aging pharmacology.
Collapse
Affiliation(s)
| | - Oleh V Lushchak
- Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | | |
Collapse
|
124
|
Spotlight on the relevance of mtDNA in cancer. Clin Transl Oncol 2016; 19:409-418. [PMID: 27778302 DOI: 10.1007/s12094-016-1561-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 10/06/2016] [Indexed: 02/06/2023]
Abstract
The potential role of the mitochondrial genome has recently attracted interest because of its high mutation frequency in tumors. Different aspects of mtDNA make it relevant for cancer's biology, such as it encodes a limited but essential number of genes for OXPHOS biogenesis, it is particularly susceptible to mutations, and its copy number can vary. Moreover, most ROS in mitochondria are produced by the electron transport chain. These characteristics place the mtDNA in the center of multiple signaling pathways, known as mitochondrial retrograde signaling, which modifies numerous key processes in cancer. Cybrid studies support that mtDNA mutations are relevant and exert their effect through a modification of OXPHOS function and ROS production. However, there is still much controversy regarding the clinical relevance of mtDNA mutations. New studies should focus more on OXPHOS dysfunction associated with a specific mutational signature rather than the presence of mutations in the mtDNA.
Collapse
|
125
|
Zhang Y, Hood WR. Current versus future reproduction and longevity: a re-evaluation of predictions and mechanisms. J Exp Biol 2016; 219:3177-3189. [PMID: 27802148 PMCID: PMC5091378 DOI: 10.1242/jeb.132183] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Oxidative damage is predicted to be a mediator of trade-offs between current reproduction and future reproduction or survival, but most studies fail to support such predictions. We suggest that two factors underlie the equivocal nature of these findings: (1) investigators typically assume a negative linear relationship between current reproduction and future reproduction or survival, even though this is not consistently shown by empirical studies; and (2) studies often fail to target mechanisms that could link interactions between sequential life-history events. Here, we review common patterns of reproduction, focusing on the relationships between reproductive performance, survival and parity in females. Observations in a range of species show that performance between sequential reproductive events can decline, remain consistent or increase. We describe likely bioenergetic consequences of reproduction that could underlie these changes in fitness, including mechanisms that could be responsible for negative effects being ephemeral, persistent or delayed. Finally, we make recommendations for designing future studies. We encourage investigators to carefully consider additional or alternative measures of bioenergetic function in studies of life-history trade-offs. Such measures include reactive oxygen species production, oxidative repair, mitochondrial biogenesis, cell proliferation, mitochondrial DNA mutation and replication error and, importantly, a measure of the respiratory function to determine whether measured differences in bioenergetic state are associated with a change in the energetic capacity of tissues that could feasibly affect future reproduction or lifespan. More careful consideration of the life-history context and bioenergetic variables will improve our understanding of the mechanisms that underlie the life-history patterns of animals.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Wendy R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
126
|
Mowry AV, Kavazis AN, Sirman AE, Potts WK, Hood WR. Reproduction Does Not Adversely Affect Liver Mitochondrial Respiratory Function but Results in Lipid Peroxidation and Increased Antioxidants in House Mice. PLoS One 2016; 11:e0160883. [PMID: 27537547 PMCID: PMC4990174 DOI: 10.1371/journal.pone.0160883] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/26/2016] [Indexed: 12/31/2022] Open
Abstract
Reproduction is thought to come at a cost to longevity. Based on the assumption that increased energy expenditure during reproduction is associated with increased free-radical production by mitochondria, oxidative damage has been suggested to drive this trade-off. We examined the impact of reproduction on liver mitochondrial function by utilizing post-reproductive and non-reproductive house mice (Mus musculus) living under semi-natural conditions. The age-matched post-reproductive and non-reproductive groups were compared after the reproductive females returned to a non-reproductive state, so that both groups were in the same physiological state at the time the liver was collected. Despite increased oxidative damage (p = 0.05) and elevated CuZnSOD (p = 0.002) and catalase (p = 0.04) protein levels, reproduction had no negative impacts on the respiratory function of liver mitochondria. Specifically, in a post-reproductive, maintenance state the mitochondrial coupling (i.e., respiratory control ratio) of mouse livers show no negative impacts of reproduction. In fact, there was a trend (p = 0.059) to suggest increased maximal oxygen consumption by liver mitochondria during the ADP stimulated state (i.e., state 3) in post-reproduction. These findings suggest that oxidative damage may not impair mitochondrial respiratory function and question the role of mitochondria in the trade-off between reproduction and longevity. In addition, the findings highlight the importance of quantifying the respiratory function of mitochondria in addition to measuring oxidative damage.
Collapse
Affiliation(s)
- Annelise V. Mowry
- Department of Biological Sciences, Auburn University, Auburn, Alabama, United States of America
| | - Andreas N. Kavazis
- School of Kinesiology, Auburn University, Auburn, Alabama, United States of America
| | - Aubrey E. Sirman
- Department of Biological Sciences, Auburn University, Auburn, Alabama, United States of America
| | - Wayne K. Potts
- Department of Biology, University of Utah, Salt Lake City, Utah, United States of America
| | - Wendy R. Hood
- Department of Biological Sciences, Auburn University, Auburn, Alabama, United States of America
- * E-mail:
| |
Collapse
|
127
|
da Costa JP, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T. A synopsis on aging-Theories, mechanisms and future prospects. Ageing Res Rev 2016; 29:90-112. [PMID: 27353257 PMCID: PMC5991498 DOI: 10.1016/j.arr.2016.06.005] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/23/2016] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
Abstract
Answering the question as to why we age is tantamount to answering the question of what is life itself. There are countless theories as to why and how we age, but, until recently, the very definition of aging - senescence - was still uncertain. Here, we summarize the main views of the different models of senescence, with a special emphasis on the biochemical processes that accompany aging. Though inherently complex, aging is characterized by numerous changes that take place at different levels of the biological hierarchy. We therefore explore some of the most relevant changes that take place during aging and, finally, we overview the current status of emergent aging therapies and what the future holds for this field of research. From this multi-dimensional approach, it becomes clear that an integrative approach that couples aging research with systems biology, capable of providing novel insights into how and why we age, is necessary.
Collapse
Affiliation(s)
- João Pinto da Costa
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
| | - Rui Vitorino
- Department of Medical Sciences, Institute for Biomedicine-iBiMED, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Gustavo M Silva
- Department of Biology, Center for Genomics and Systems Biology, NY, NY 10003, USA
| | - Christine Vogel
- Department of Biology, Center for Genomics and Systems Biology, NY, NY 10003, USA
| | - Armando C Duarte
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Teresa Rocha-Santos
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
128
|
Wisnovsky S, Lei E, Jean S, Kelley S. Mitochondrial Chemical Biology: New Probes Elucidate the Secrets of the Powerhouse of the Cell. Cell Chem Biol 2016; 23:917-27. [DOI: 10.1016/j.chembiol.2016.06.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/02/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
|
129
|
Perales-Clemente E, Cook AN, Evans JM, Roellinger S, Secreto F, Emmanuele V, Oglesbee D, Mootha VK, Hirano M, Schon EA, Terzic A, Nelson TJ. Natural underlying mtDNA heteroplasmy as a potential source of intra-person hiPSC variability. EMBO J 2016; 35:1979-90. [PMID: 27436875 PMCID: PMC5282833 DOI: 10.15252/embj.201694892] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 06/24/2016] [Indexed: 01/19/2023] Open
Abstract
Functional variability among human clones of induced pluripotent stem cells (hiPSCs) remains a limitation in assembling high-quality biorepositories. Beyond inter-person variability, the root cause of intra-person variability remains unknown. Mitochondria guide the required transition from oxidative to glycolytic metabolism in nuclear reprogramming. Moreover, mitochondria have their own genome (mitochondrial DNA [mtDNA]). Herein, we performed mtDNA next-generation sequencing (NGS) on 84 hiPSC clones derived from a cohort of 19 individuals, including mitochondrial and non-mitochondrial patients. The analysis of mtDNA variants showed that low levels of potentially pathogenic mutations in the original fibroblasts are revealed through nuclear reprogramming, generating mutant hiPSCs with a detrimental effect in their differentiated progeny. Specifically, hiPSC-derived cardiomyocytes with expanded mtDNA mutations non-related with any described human disease, showed impaired mitochondrial respiration, being a potential cause of intra-person hiPSC variability. We propose mtDNA NGS as a new selection criterion to ensure hiPSC quality for drug discovery and regenerative medicine.
Collapse
Affiliation(s)
- Ester Perales-Clemente
- Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Division of Cardiovascular Diseases, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, USA
| | - Alexandra N Cook
- Departments of Cardiovascular Diseases, Molecular Pharmacology and Experimental Therapeutics, Division of General Internal Medicine, Division of Pediatric Cardiology, and Transplant Center, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, USA
| | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Samantha Roellinger
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Frank Secreto
- Departments of Cardiovascular Diseases, Molecular Pharmacology and Experimental Therapeutics, Division of General Internal Medicine, Division of Pediatric Cardiology, and Transplant Center, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, USA
| | - Valentina Emmanuele
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Vamsi K Mootha
- Department of Molecular Biology, Howard Hughes Medical Institute Massachusetts General Hospital, Boston, MA, USA
| | - Michio Hirano
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Eric A Schon
- Department of Neurology, Columbia University Medical Center, New York, NY, USA Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Andre Terzic
- Departments of Medicine, Molecular Pharmacology and Experimental Therapeutics, and Medical Genetics, Division of Cardiovascular Diseases, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, USA
| | - Timothy J Nelson
- Departments of Cardiovascular Diseases, Molecular Pharmacology and Experimental Therapeutics, Division of General Internal Medicine, Division of Pediatric Cardiology, and Transplant Center, Mayo Clinic Center for Regenerative Medicine, Rochester, MN, USA
| |
Collapse
|
130
|
DNA Damage and Repair in Schizophrenia and Autism: Implications for Cancer Comorbidity and Beyond. Int J Mol Sci 2016; 17:ijms17060856. [PMID: 27258260 PMCID: PMC4926390 DOI: 10.3390/ijms17060856] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/12/2016] [Accepted: 05/27/2016] [Indexed: 12/16/2022] Open
Abstract
Schizophrenia and autism spectrum disorder (ASD) are multi-factorial and multi-symptomatic psychiatric disorders, each affecting 0.5%-1% of the population worldwide. Both are characterized by impairments in cognitive functions, emotions and behaviour, and they undermine basic human processes of perception and judgment. Despite decades of extensive research, the aetiologies of schizophrenia and ASD are still poorly understood and remain a significant challenge to clinicians and scientists alike. Adding to this unsatisfactory situation, patients with schizophrenia or ASD often develop a variety of peripheral and systemic disturbances, one prominent example of which is cancer, which shows a direct (but sometimes inverse) comorbidity in people affected with schizophrenia and ASD. Cancer is a disease characterized by uncontrolled proliferation of cells, the molecular origin of which derives from mutations of a cell's DNA sequence. To counteract such mutations and repair damaged DNA, cells are equipped with intricate DNA repair pathways. Oxidative stress, oxidative DNA damage, and deficient repair of oxidative DNA lesions repair have been proposed to contribute to the development of schizophrenia and ASD. In this article, we summarize the current evidence of cancer comorbidity in these brain disorders and discuss the putative roles of oxidative stress, DNA damage and DNA repair in the aetiopathology of schizophrenia and ASD.
Collapse
|
131
|
Wisnovsky S, Jean SR, Liyanage S, Schimmer A, Kelley SO. Mitochondrial DNA repair and replication proteins revealed by targeted chemical probes. Nat Chem Biol 2016; 12:567-73. [DOI: 10.1038/nchembio.2102] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/24/2016] [Indexed: 01/16/2023]
|
132
|
Fetterman JL, Holbrook M, Westbrook DG, Brown JA, Feeley KP, Bretón-Romero R, Linder EA, Berk BD, Weisbrod RM, Widlansky ME, Gokce N, Ballinger SW, Hamburg NM. Mitochondrial DNA damage and vascular function in patients with diabetes mellitus and atherosclerotic cardiovascular disease. Cardiovasc Diabetol 2016; 15:53. [PMID: 27036979 PMCID: PMC4818501 DOI: 10.1186/s12933-016-0372-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 03/22/2016] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Prior studies demonstrate mitochondrial dysfunction with increased reactive oxygen species generation in peripheral blood mononuclear cells in diabetes mellitus. Oxidative stress-mediated damage to mitochondrial DNA promotes atherosclerosis in animal models. Thus, we evaluated the relation of mitochondrial DNA damage in peripheral blood mononuclear cells s with vascular function in patients with diabetes mellitus and with atherosclerotic cardiovascular disease. APPROACH AND RESULTS We assessed non-invasive vascular function and mitochondrial DNA damage in 275 patients (age 57 ± 9 years, 60 % women) with atherosclerotic cardiovascular disease alone (N = 55), diabetes mellitus alone (N = 74), combined atherosclerotic cardiovascular disease and diabetes mellitus (N = 48), and controls age >45 without diabetes mellitus or atherosclerotic cardiovascular disease (N = 98). Mitochondrial DNA damage measured by quantitative PCR in peripheral blood mononuclear cells was higher with clinical atherosclerosis alone (0.55 ± 0.65), diabetes mellitus alone (0.65 ± 1.0), and combined clinical atherosclerosis and diabetes mellitus (0.89 ± 1.32) as compared to control subjects (0.23 ± 0.64, P < 0.0001). In multivariable models adjusting for age, sex, and relevant cardiovascular risk factors, clinical atherosclerosis and diabetes mellitus remained associated with higher mitochondrial DNA damage levels (β = 0.14 ± 0.13, P = 0.04 and β = 0.21 ± 0.13, P = 0.002, respectively). Higher mitochondrial DNA damage was associated with higher baseline pulse amplitude, a measure of arterial pulsatility, but not with flow-mediated dilation or hyperemic response, measures of vasodilator function. CONCLUSIONS We found greater mitochondrial DNA damage in patients with diabetes mellitus and clinical atherosclerosis. The association of mitochondrial DNA damage and baseline pulse amplitude may suggest a link between mitochondrial dysfunction and excessive small artery pulsatility with potentially adverse microvascular impact.
Collapse
Affiliation(s)
- Jessica L Fetterman
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA.
| | - Monica Holbrook
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA
| | - David G Westbrook
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jamelle A Brown
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kyle P Feeley
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rosa Bretón-Romero
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA
| | - Erika A Linder
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA
| | - Brittany D Berk
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA
| | - Robert M Weisbrod
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA
| | - Michael E Widlansky
- Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA
| | - Scott W Ballinger
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Naomi M Hamburg
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, 72 East Concord Street, E-784, Boston, MA, 02118, USA
| |
Collapse
|
133
|
Stewart JB, Chinnery PF. The dynamics of mitochondrial DNA heteroplasmy: implications for human health and disease. Nat Rev Genet 2015; 16:530-42. [PMID: 26281784 DOI: 10.1038/nrg3966] [Citation(s) in RCA: 625] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Common genetic variants of mitochondrial DNA (mtDNA) increase the risk of developing several of the major health issues facing the western world, including neurodegenerative diseases. In this Review, we consider how these mtDNA variants arose and how they spread from their origin on one single molecule in a single cell to be present at high levels throughout a specific organ and, ultimately, to contribute to the population risk of common age-related disorders. mtDNA persists in all aerobic eukaryotes, despite a high substitution rate, clonal propagation and little evidence of recombination. Recent studies have found that de novo mtDNA mutations are suppressed in the female germ line; despite this, mtDNA heteroplasmy is remarkably common. The demonstration of a mammalian mtDNA genetic bottleneck explains how new germline variants can increase to high levels within a generation, and the ultimate fixation of less-severe mutations that escape germline selection explains how they can contribute to the risk of late-onset disorders.
Collapse
Affiliation(s)
- James B Stewart
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Patrick F Chinnery
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 1BZ, UK
| |
Collapse
|
134
|
Picca A, Lezza AMS. Regulation of mitochondrial biogenesis through TFAM-mitochondrial DNA interactions: Useful insights from aging and calorie restriction studies. Mitochondrion 2015; 25:67-75. [PMID: 26437364 DOI: 10.1016/j.mito.2015.10.001] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/24/2015] [Accepted: 10/01/2015] [Indexed: 11/24/2022]
Abstract
Mitochondrial biogenesis is regulated to adapt mitochondrial population to cell energy demands. Mitochondrial transcription factor A (TFAM) performs several functions for mtDNA and interactions between TFAM and mtDNA participate to regulation of mitochondrial biogenesis. Such interactions are modulated through different mechanisms: regulation of TFAM expression and turnover, modulation of TFAM binding activity to mtDNA through post-translational modifications and differential affinity of TFAM, occurrence of TFAM sliding on mtDNA filaments and of cooperative binding among TFAM molecules, modulation of protein-protein interactions. The tissue-specific regulation of mitochondrial biogenesis in aging and calorie restriction (CR) highlights the relevance of modulation of TFAM-mtDNA interactions.
Collapse
Affiliation(s)
- Anna Picca
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70125, Bari, Italy
| | - Angela Maria Serena Lezza
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70125, Bari, Italy.
| |
Collapse
|
135
|
Mitochondrial divergence between slow- and fast-aging garter snakes. Exp Gerontol 2015; 71:135-46. [PMID: 26403677 DOI: 10.1016/j.exger.2015.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/03/2015] [Accepted: 09/04/2015] [Indexed: 01/26/2023]
Abstract
Mitochondrial function has long been hypothesized to be intimately involved in aging processes--either directly through declining efficiency of mitochondrial respiration and ATP production with advancing age, or indirectly, e.g., through increased mitochondrial production of damaging free radicals with age. Yet we lack a comprehensive understanding of the evolution of mitochondrial genotypes and phenotypes across diverse animal models, particularly in species that have extremely labile physiology. Here, we measure mitochondrial genome-types and transcription in ecotypes of garter snakes (Thamnophis elegans) that are adapted to disparate habitats and have diverged in aging rates and lifespans despite residing in close proximity. Using two RNA-seq datasets, we (1) reconstruct the garter snake mitochondrial genome sequence and bioinformatically identify regulatory elements, (2) test for divergence of mitochondrial gene expression between the ecotypes and in response to heat stress, and (3) test for sequence divergence in mitochondrial protein-coding regions in these slow-aging (SA) and fast-aging (FA) naturally occurring ecotypes. At the nucleotide sequence level, we confirmed two (duplicated) mitochondrial control regions one of which contains a glucocorticoid response element (GRE). Gene expression of protein-coding genes was higher in FA snakes relative to SA snakes for most genes, but was neither affected by heat stress nor an interaction between heat stress and ecotype. SA and FA ecotypes had unique mitochondrial haplotypes with amino acid substitutions in both CYTB and ND5. The CYTB amino acid change (Isoleucine → Threonine) was highly segregated between ecotypes. This divergence of mitochondrial haplotypes between SA and FA snakes contrasts with nuclear gene-flow estimates, but correlates with previously reported divergence in mitochondrial function (mitochondrial oxygen consumption, ATP production, and reactive oxygen species consequences).
Collapse
|