1
|
De la Cruz-Góngora V, Palazuelos-González R, Domínguez-Flores O. Micronutrient Deficiencies in Older Adults in Latin America: A Narrative Review. Food Nutr Bull 2024; 45:S26-S38. [PMID: 38146136 DOI: 10.1177/03795721231214587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
BACKGROUND The population in Latin America and Caribbean (LAC) has experienced a major demographic transition with increased numbers of older adults (OA). This change brings opportunities in the public health sector to implement health prevention interventions and delay the onset of geriatric syndromes. Micronutrients play an important role in the maintenance of biological function which contributes to longevity. Micronutrient deficiencies (MD) in OA increase the risk for onset of chronic comorbidities and geriatric syndromes. AIM To review and summarize the existing data on micronutrient status in OA in the LAC region and discuss the gaps and challenges in public health approaches to address deficiencies. METHODS Literature review in Medline for records describing nutritional biomarkers in older adults (≥ 60y) from community dwelling and population-based studies in LAC. RESULTS Few countries (including Chile, Ecuador, Costa Rica, Brazil, and Mexico) have documented one or more nutritional deficiencies for OA in national health surveys, however across the entire region, evidence of micronutrient levels is scarce. Some surveys have documented a high prevalence and large heterogeneity in the prevalence of vitamin D followed by B12 deficiency, being the 2 MDs most studied due their effects on cognition, frailty, and bone mineral density in the OA population. Other MD including C, E, A, copper, zinc, iron, and selenium have also been reported. CONCLUSION Information on the micronutrient status in OA from LAC is poorly documented. Research and capacity building initiatives in the region are crucial to develop tailored strategies that address the specific nutritional needs and challenges faced by the ageing population in Latin America.
Collapse
Affiliation(s)
- Vanessa De la Cruz-Góngora
- Center for Evaluation and Survey Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
- Global Brain Health Institute, Trinity College, Dublin, Ireland
| | | | - Omar Domínguez-Flores
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| |
Collapse
|
2
|
Zheng Q, Wang F, Nie C, Zhang K, Sun Y, Al-Ansi W, Wu Q, Wang L, Du J, Li Y. Elevating the significance of legume intake: A novel strategy to counter aging-related mitochondrial dysfunction and physical decline. Compr Rev Food Sci Food Saf 2024; 23:e13342. [PMID: 38634173 DOI: 10.1111/1541-4337.13342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024]
Abstract
Mitochondrial dysfunction increasingly becomes a target for promoting healthy aging and longevity. The dysfunction of mitochondria with age ultimately leads to a decline in physical functions. Among them, biogenesis dysfunction and the imbalances in the metabolism of reactive oxygen species and mitochondria as signaling organelles in the aging process have aroused our attention. Dietary intervention in mitochondrial dysfunction and physical decline during aging processes is essential, and greater attention should be directed toward healthful legume intake. Legumes are constantly under investigation for their nutritional and bioactive properties, and their consumption may yield antiaging and mitochondria-protecting benefits. This review summarizes mitochondrial dysfunction with age, discusses the benefits of legumes on mitochondrial function, and introduces the potential role of legumes in managing aging-related physical decline. Additionally, it reveals the benefits of legume intake for the elderly and offers a viable approach to developing legume-based functional food.
Collapse
Affiliation(s)
- Qingwei Zheng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Feijie Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chenzhipeng Nie
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Kuiliang Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yujie Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Waleed Al-Ansi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qiming Wu
- Nutrilite Health Institute, Shanghai, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
3
|
Toljić B, Milašin J, De Luka SR, Dragović G, Jevtović D, Maslać A, Ristić-Djurović JL, Trbovich AM. HIV-Infected Patients as a Model of Aging. Microbiol Spectr 2023; 11:e0053223. [PMID: 37093018 PMCID: PMC10269491 DOI: 10.1128/spectrum.00532-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/01/2023] [Indexed: 04/25/2023] Open
Abstract
We appraised the relationship between the biological and the chronological age and estimated the rate of biological aging in HIV-infected patients. Two independent biomarkers, the relative telomere length and iron metabolism parameters, were analyzed in younger (<35) and older (>50) HIV-infected and uninfected patients (control group). In our control group, telomeres of younger patients were significantly longer than telomeres of older ones. However, in HIV-infected participants, the difference in the length of telomeres was lost. By combining the length of telomeres with serum iron, ferritin, and transferrin iron-binding capacity, a new formula for determination of the aging process was developed. The life expectancy of the healthy population was related to their biological age, and HIV-infected patients were biologically older. The effect of antiretroviral HIV drug therapies varied with respect to the biological aging process. IMPORTANCE This article is focused on the dynamics of human aging. Moreover, its interdisciplinary approach is applicable to various systems that are aging.
Collapse
Affiliation(s)
- Boško Toljić
- School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Milašin
- School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | | | | | | | | | | | | |
Collapse
|
4
|
Lin PH, Su WP, Li CJ, Lin LT, Sheu JJC, Wen ZH, Cheng JT, Tsui KH. Investigating the Role of Ferroptosis-Related Genes in Ovarian Aging and the Potential for Nutritional Intervention. Nutrients 2023; 15:2461. [PMID: 37299424 PMCID: PMC10255416 DOI: 10.3390/nu15112461] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
With advancing age, women experience irreversible deterioration in the quality of their oocytes, resulting in reduced fertility. To gain a deeper understanding of the influence of ferroptosis-related genes on ovarian aging, we employed a comprehensive approach encompassing spatial transcriptomics, single-cell RNA sequencing, human ovarian pathology, and clinical biopsy. This investigation revealed the intricate interactions between ferroptosis and cellular energy metabolism in aging germ cells, shedding light on the underlying mechanisms. Our study involved 75 patients with ovarian senescence insufficiency, and we utilized multi-histological predictions of ferroptosis-related genes. Following a two-month supplementation period with DHEA, Ubiquinol CoQ10, and Cleo-20 T3, we examined the changes in hub genes. Our results showed that TFRC, NCOA4, and SLC3A2 were significantly reduced and GPX4 was increased in the supplement group, confirming our prediction based on multi-omic analysis. Our hypothesis is that supplementation would enhance the mitochondrial tricarboxylic acid cycle (TCA) or electron transport chain (ETC), resulting in increased levels of the antioxidant enzyme GPX4, reduced lipid peroxide accumulation, and reduced ferroptosis. Overall, our results suggest that supplementation interventions have a notable positive impact on in vitro fertilization (IVF) outcomes in aging cells by improving metal ion and energy metabolism, thereby enhancing oocyte quality in older women.
Collapse
Affiliation(s)
- Pei-Hsuan Lin
- Institute of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
- Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Wan-Ping Su
- Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Li-Te Lin
- Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Jiin-Tsuey Cheng
- Institute of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Kuan-Hao Tsui
- Institute of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
- Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Obstetrics and Gynaecology, National Yang-Ming University School of Medicine, Taipei 112, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
5
|
Wang Y, Wu W, Gong J. Live or death in cells: from micronutrition metabolism to cell fate. Front Cell Dev Biol 2023; 11:1185989. [PMID: 37250891 PMCID: PMC10213646 DOI: 10.3389/fcell.2023.1185989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Micronutrients and cell death have a strong relationship and both are essential for human to maintain good body health. Dysregulation of any micronutrients causes metabolic or chronic diseases, including obesity, cardiometabolic condition, neurodegeneration, and cancer. The nematode Caenorhabditis elegans is an ideal genetic organism for researching the mechanisms of micronutrients in metabolism, healthspan, and lifespan. For example, C. elegans is a haem auxotroph, and the research of this special haem trafficking pathway contributes important reference to mammal study. Also, C. elegans characteristics including anatomy simply, clear cell lineage, well-defined genetics, and easily differentiated cell forms make it a powerful tool for studying the mechanisms of cell death including apoptosis, necrosis, autophagy, and ferroptosis. Here, we describe the understanding of micronutrient metabolism currently and also sort out the fundamental mechanisms of different kinds of cell death. A thorough understanding of these physiological processes not only builds a foundation for developing better treatments for various micronutrient disorders but also provides key insights into human health and aging.
Collapse
Affiliation(s)
- Yuting Wang
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianke Gong
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
6
|
Costa I, Barbosa DJ, Benfeito S, Silva V, Chavarria D, Borges F, Remião F, Silva R. Molecular mechanisms of ferroptosis and their involvement in brain diseases. Pharmacol Ther 2023; 244:108373. [PMID: 36894028 DOI: 10.1016/j.pharmthera.2023.108373] [Citation(s) in RCA: 77] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Ferroptosis is a type of regulated cell death characterized by intracellular accumulation of iron and reactive oxygen species, inhibition of system Xc-, glutathione depletion, nicotinamide adenine dinucleotide phosphate oxidation and lipid peroxidation. Since its discovery and characterization in 2012, many efforts have been made to reveal the underlying mechanisms, modulating compounds, and its involvement in disease pathways. Ferroptosis inducers include erastin, sorafenib, sulfasalazine and glutamate, which, by inhibiting system Xc-, prevent the import of cysteine into the cells. RSL3, statins, Ml162 and Ml210 induce ferroptosis by inhibiting glutathione peroxidase 4 (GPX4), which is responsible for preventing the formation of lipid peroxides, and FIN56 and withaferin trigger GPX4 degradation. On the other side, ferroptosis inhibitors include ferrostatin-1, liproxstatin-1, α-tocopherol, zileuton, FSP1, CoQ10 and BH4, which interrupt the lipid peroxidation cascade. Additionally, deferoxamine, deferiprone and N-acetylcysteine, by targeting other cellular pathways, have also been classified as ferroptosis inhibitors. Increased evidence has established the involvement of ferroptosis in distinct brain diseases, including Alzheimer's, Parkinson's and Huntington's diseases, amyotrophic lateral sclerosis, multiple sclerosis, and Friedreich's ataxia. Thus, a deep understanding of how ferroptosis contributes to these diseases, and how it can be modulated, can open a new window of opportunities for novel therapeutic strategies and targets. Other studies have shown a sensitivity of cancer cells with mutated RAS to ferroptosis induction and that chemotherapeutic agents and ferroptosis inducers synergize in tumor treatment. Thus, it is tempting to consider that ferroptosis may arise as a target mechanistic pathway for the treatment of brain tumors. Therefore, this work provides an up-to-date review on the molecular and cellular mechanisms of ferroptosis and their involvement in brain diseases. In addition, information on the main ferroptosis inducers and inhibitors and their molecular targets is also provided.
Collapse
Affiliation(s)
- Inês Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- TOXRUN - Toxicology Research Unit, Department of Sciences, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal; Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal.
| | - Vera Silva
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernanda Borges
- CIQUP-IMS - Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Renata Silva
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
7
|
Radakovich LB, Burton LH, Culver LA, Afzali MF, Marolf AJ, Olver CS, Santangelo KS. Systemic iron reduction via an iron deficient diet decreases the severity of knee cartilage lesions in the Dunkin-Hartley guinea pig model of osteoarthritis. Osteoarthritis Cartilage 2022; 30:1482-1494. [PMID: 36030059 PMCID: PMC9588731 DOI: 10.1016/j.joca.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Iron accumulation is emerging as a player in aging-related disorders due to its propensity for generating reactive oxygen species (ROS). Studies investigating the role of iron in the pathogenesis of primary osteoarthritis (OA) are limited. We designed a proof-of-principle study to determine the effect of systemic iron deficiency, via an iron deficient diet, on knee OA in an animal model. METHODS Twelve-week-old male Hartley guinea pigs received the standard diet (n = 6) or a diet devoid of iron (n = 6) for 19-weeks. Iron levels were determined in the serum, liver, and articular cartilage. Knees were collected to assess structural changes related to OA (microcomputed tomography, histopathology). Immunohistochemistry was performed to evaluate the presence and distribution of a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) and ROS-driven 4-hydroxynonenal (4-HNE)-induced protein adducts. Transcript expression was also assessed. RESULTS Relative to control animals, an iron deficient diet reduced the concentration of this mineral in serum, liver, and articular cartilage. Iron deficient animals had lower histologic OA scores; decreased subchondral bone mineral density was also noted. This reduction in knee joint pathology was accompanied by a decrease in: ADAMTS4 in synovium; and 4-HNE protein adducts from lipid peroxidation in both the menisci and articular cartilage of iron deficient animals. Expression of iron-related genes in these tissues was also altered in treated animals. CONCLUSIONS Results from this study suggest that systemic iron levels may play a role in knee OA pathogenesis, with a short-term deficit in dietary iron reducing the severity of knee cartilage lesions.
Collapse
Affiliation(s)
- L B Radakovich
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA.
| | - L H Burton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.
| | - L A Culver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - M F Afzali
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - A J Marolf
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - C S Olver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - K S Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
8
|
Sze SCW, Zhang L, Zhang S, Lin K, Ng TB, Ng ML, Lee KF, Lam JKW, Zhang Z, Yung KKL. Aberrant Transferrin and Ferritin Upregulation Elicits Iron Accumulation and Oxidative Inflammaging Causing Ferroptosis and Undermines Estradiol Biosynthesis in Aging Rat Ovaries by Upregulating NF-Κb-Activated Inducible Nitric Oxide Synthase: First Demonstration of an Intricate Mechanism. Int J Mol Sci 2022; 23:ijms232012689. [PMID: 36293552 PMCID: PMC9604315 DOI: 10.3390/ijms232012689] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022] Open
Abstract
We report herein a novel mechanism, unraveled by proteomics and validated by in vitro and in vivo studies, of the aberrant aging-associated upregulation of ovarian transferrin and ferritin in rat ovaries. The ovarian mass and serum estradiol titer plummeted while the ovarian labile ferrous iron and total iron levels escalated with age in rats. Oxidative stress markers, such as nitrite/nitrate, 3-nitrotyrosine, and 4-hydroxy-2-nonenal, accumulated in the aging ovaries due to an aberrant upregulation of the ovarian transferrin, ferritin light/heavy chains, and iron regulatory protein 2(IRP2)-mediated transferrin receptor 1 (TfR1). Ferritin inhibited estradiol biosynthesis in ovarian granulosa cells in vitro via the upregulation of a nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and p65/p50-induced oxidative and inflammatory factor inducible nitric oxide synthase (iNOS). An in vivo study demonstrated how the age-associated activation of NF-κB induced the upregulation of iNOS and the tumor necrosis factor α (TNFα). The downregulation of the keap1-mediated nuclear factor erythroid 2-related factor 2 (Nrf2), that induced a decrease in glutathione peroxidase 4 (GPX4), was observed. The aberrant transferrin and ferritin upregulation triggered an iron accumulation via the upregulation of an IRP2-induced TfR1. This culminates in NF-κB-iNOS-mediated ovarian oxi-inflamm-aging and serum estradiol decrement in naturally aging rats. The iron accumulation and the effect on ferroptosis-related proteins including the GPX4, TfR1, Nrf2, Keap1, and ferritin heavy chain, as in testicular ferroptosis, indicated the triggering of ferroptosis. In young rats, an intraovarian injection of an adenovirus, which expressed iron regulatory proteins, upregulated the ovarian NF-κB/iNOS and downregulated the GPX4. These novel findings have contributed to a prompt translational research on the ovarian aging-associated iron metabolism and aging-associated ovarian diseases.
Collapse
Affiliation(s)
- Stephen Cho Wing Sze
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Correspondence: (S.C.W.S.); (K.K.L.Y.); Tel.: +852-34112318 (S.C.W.S.); Tel.: +852-34117060 (K.K.L.Y.)
| | - Liang Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China
| | - Shiqing Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou 999077, China
| | - Kaili Lin
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- School of Public Health, Guangzhou Medical University, Guangzhou 999077, China
| | - Tzi Bun Ng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR 999077, China
| | - Man Ling Ng
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, HKU, Pokfulam, Hong Kong SAR 999077, China
| | - Jenny Ka Wing Lam
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
- Department of Pharmacology & Pharmacy, LKS Faculty of Medicine, HKU, Pokfulam, Hong Kong SAR 999077, China
| | - Zhang Zhang
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
| | - Ken Kin Lam Yung
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon, Hong Kong SAR 999077, China
- Correspondence: (S.C.W.S.); (K.K.L.Y.); Tel.: +852-34112318 (S.C.W.S.); Tel.: +852-34117060 (K.K.L.Y.)
| |
Collapse
|
9
|
Trigo D, Vitória JJ, da Cruz e Silva OAB. Novel therapeutic strategies targeting mitochondria as a gateway in neurodegeneration. Neural Regen Res 2022; 18:991-995. [PMID: 36254979 PMCID: PMC9827793 DOI: 10.4103/1673-5374.355750] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In recent years, multiple disciplines have focused on mitochondrial biology and contributed to understanding its relevance towards adult-onset neurodegenerative disorders. These are complex dynamic organelles that have a variety of functions in ensuring cellular health and homeostasis. The plethora of mitochondrial functionalities confers them an intrinsic susceptibility to internal and external stressors (such as mutation accumulation or environmental toxins), particularly so in long-lived postmitotic cells such as neurons. Thus, it is reasonable to postulate an involvement of mitochondria in aging-associated neurological disorders, notably neurodegenerative pathologies including Alzheimer's disease and Parkinson's disease. On the other hand, biological effects resulting from neurodegeneration can in turn affect mitochondrial health and function, promoting a feedback loop further contributing to the progression of neuronal dysfunction and cellular death. This review examines state-of-the-art knowledge, focus on current research exploring mitochondrial health as a contributing factor to neuroregeneration, and the development of therapeutic approaches aimed at restoring mitochondrial homeostasis in a pathological setting.
Collapse
Affiliation(s)
- Diogo Trigo
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal,Correspondence to: Diogo Trigo, .
| | - José João Vitória
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Odete A. B. da Cruz e Silva
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
10
|
Role of Iron-Related Oxidative Stress and Mitochondrial Dysfunction in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5124553. [PMID: 36120592 PMCID: PMC9473912 DOI: 10.1155/2022/5124553] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022]
Abstract
Iron is indispensable in numerous biologic processes, but abnormal iron regulation and accumulation is related to pathological processes in cardiovascular diseases. However, the underlying mechanisms still need to be further explored. Iron plays a key role in metal-catalyzed oxidative reactions that generate reactive oxygen species (ROS), which can cause oxidative stress. As the center for oxygen and iron utilization, mitochondria are vulnerable to damage from iron-induced oxidative stress and participate in processes involved in iron-related damage in cardiovascular disease, although the mechanism remains unclear. In this review, the pathological roles of iron-related oxidative stress in cardiovascular diseases are summarized, and the potential effects and mechanisms of mitochondrial iron homeostasis and dysfunction in these diseases are especially highlighted.
Collapse
|
11
|
Role of Iron in Aging Related Diseases. Antioxidants (Basel) 2022; 11:antiox11050865. [PMID: 35624729 PMCID: PMC9137504 DOI: 10.3390/antiox11050865] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/17/2022] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
Iron progressively accumulates with age and can be further exacerbated by dietary iron intake, genetic factors, and repeated blood transfusions. While iron plays a vital role in various physiological processes within the human body, its accumulation contributes to cellular aging in several species. In its free form, iron can initiate the formation of free radicals at a cellular level and contribute to systemic disorders. This is most evident in high iron conditions such as hereditary hemochromatosis, when accumulation of iron contributes to the development of arthritis, cirrhosis, or cardiomyopathy. A growing body of research has further identified iron’s contributory effects in neurodegenerative diseases, ocular disorders, cancer, diabetes, endocrine dysfunction, and cardiovascular diseases. Reducing iron levels by repeated phlebotomy, iron chelation, and dietary restriction are the common therapeutic considerations to prevent iron toxicity. Chelators such as deferoxamine, deferiprone, and deferasirox have become the standard of care in managing iron overload conditions with other potential applications in cancer and cardiotoxicity. In certain animal models, drugs with iron chelating ability have been found to promote health and even extend lifespan. As we further explore the role of iron in the aging process, iron chelators will likely play an increasingly important role in our health.
Collapse
|
12
|
Burton LH, Afzali MF, Radakovich LB, Campbell MA, Culver LA, Olver CS, Santangelo KS. Systemic administration of a pharmacologic iron chelator reduces cartilage lesion development in the Dunkin-Hartley model of primary osteoarthritis. Free Radic Biol Med 2022; 179:47-58. [PMID: 34923104 PMCID: PMC8760171 DOI: 10.1016/j.freeradbiomed.2021.12.257] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Iron has been emerging as a key contributor to aging-associated, chronic disorders due to the propensity for generating reactive oxygen species. To date, there are a limited number of publications exploring the role of iron in the pathogenesis of primary/age-related osteoarthritis (OA). The objective of this study was to determine whether reduced iron via pharmacologic iron chelation with deferoxamine (DFO) affected the development and/or severity of cartilage lesions in a primary OA model. At 12-weeks-of-age, 15 male Dunkin-Hartley guinea pigs received either 46 mg/kg DFO (n = 8) or vehicle control (n = 7) injected subcutaneously twice daily for five days each week. Movement changes, captured via overhead enclosure monitoring, were also determined. Termination occurred at 30-weeks-of-age. Iron was quantified in serum, urine, liver, and femoral head articular cartilage. Left knees were evaluated for: structural changes using histopathology guidelines; and immunohistochemistry. Gene expression analysis was conducted on right knee articular cartilage. DFO reduced iron levels in femoral head articular cartilage (p = 0.0006) and liver (p = 0.02), and increased iron within urine (p = 0.04) and serum (p = 0.0009). Mobility of control animals declined, while the DFO group maintained activity levels similar to the first month of treatment (p = 0.05). OA-associated cartilage lesions were reduced in knees of DFO animals (p = 0.0001), with chondrocyte hypocellularity a key histologic difference between groups (p < 0.0001). DFO-receiving animals had increased immunostaining for phosphorylated adenosine monophosphate activated protein kinase alpha within knee articular cartilage; lower transcript counts of several proapoptotic genes (p = 0.04-0.0004) and matrix-degrading enzymes (p = 0.02-<0.0001), and increased expression of the anti-apoptotic gene Bcl-2 (p < 0.0001) and a tissue inhibitor of matrix-metalloproteinases (p = 0.03) were also observed. These results suggest that iron chelation delayed the progression of primary OA in an animal model and could hold potential as a translational intervention. These findings provide expanded insight into factors that may contribute to the pathogenesis of primary OA.
Collapse
Affiliation(s)
- Lindsey H Burton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Maryam F Afzali
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Lauren B Radakovich
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Margaret A Campbell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Lauren A Culver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Christine S Olver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Kelly S Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
13
|
Jayakumar D, S Narasimhan KK, Periandavan K. Triad role of hepcidin, ferroportin, and Nrf2 in cardiac iron metabolism: From health to disease. J Trace Elem Med Biol 2022; 69:126882. [PMID: 34710708 DOI: 10.1016/j.jtemb.2021.126882] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 11/28/2022]
Abstract
Iron is an essential trace element required for several vital physiological and developmental processes, including erythropoiesis, bone, and neuronal development. Iron metabolism and oxygen homeostasis are interlinked to perform a vital role in the functionality of the heart. The metabolic machinery of the heart utilizes almost 90 % of oxygen through the electron transport chain. To handle this tremendous level of oxygen, the iron metabolism in the heart is utmost crucial. Iron availability to the heart is therefore tightly regulated by (i) the hepcidin/ferroportin axis, which controls dietary iron absorption, storage, and recycling, and (ii) iron regulatory proteins 1 and 2 (IRP1/2) via hypoxia inducible factor 1 (HIF1) pathway. Despite iron being vital to the heart, recent investigations have demonstrated that iron imbalance is a common manifestation in conditions of heart failure (HF), since free iron readily transforms between Fe2+ and Fe3+via the Fenton reaction, leading to reactive oxygen species (ROS) production and oxidative damage. Therefore, to combat iron-mediated oxidative stress, targeting Nrf2/ARE antioxidant signaling is rational. The involvement of Nrf2 in regulating several genes engaged in heme synthesis, iron storage, and iron export is beginning to be uncovered. Consequently, it is possible that Nrf2/hepcidin/ferroportin might act as an epicenter connecting iron metabolism to redox alterations. However, the mechanism bridging the two remains obscure. In this review, we tried to summarize the contemporary insight of how cardiomyocytes regulate intracellular iron levels and discussed the mechanisms linking cardiac dysfunction with iron imbalance. Further, we emphasized the impact of Nrf2 on the interplay between systemic/cardiac iron control in the context of heart disease, particularly in myocardial ischemia and HF.
Collapse
Affiliation(s)
- Deepthy Jayakumar
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India
| | - Kishore Kumar S Narasimhan
- Department of Pharmacology and Neurosciences, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Kalaiselvi Periandavan
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India.
| |
Collapse
|
14
|
Lee YN, Wang HH, Su CH, Lee HI, Chou YH, Hsieh CL, Liu WT, Shu KT, Chang KT, Yeh HI, Wu YJ. Deferoxamine accelerates endothelial progenitor cell senescence and compromises angiogenesis. Aging (Albany NY) 2021; 13:21364-21384. [PMID: 34508614 PMCID: PMC8457614 DOI: 10.18632/aging.203469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 08/02/2021] [Indexed: 11/25/2022]
Abstract
Senescence reduces the circulating number and angiogenic activity of endothelial progenitor cells (EPCs), and is associated with aging-related vascular diseases. However, it is very time-consuming to obtain aged cells (~1 month of repeated replication) or animals (~2 years) for senescence studies. Here, we established an accelerated senescence model by treating EPCs with deferoxamine (DFO), an FDA-approved iron chelator. Four days of low-dose (3 μM) DFO induced senescent phenotypes in EPCs, including a senescent pattern of protein expression, impaired mitochondrial bioenergetics, altered mitochondrial protein levels and compromised angiogenic activity. DFO-treated early EPCs from young and old donors (< 35 vs. > 70 years old) displayed similar senescent phenotypes, including elevated senescence-associated β-galactosidase activity and reduced relative telomere lengths, colony-forming units and adenosine triphosphate levels. To validate this accelerated senescence model in vivo, we intraperitoneally injected Sprague-Dawley rats with DFO for 4 weeks. Early EPCs from DFO-treated rats displayed profoundly senescent phenotypes compared to those from control rats. Additionally, in hind-limb ischemic mice, DFO pretreatment compromised EPC angiogenesis by reducing both blood perfusion and capillary density. DFO thus accelerates EPC senescence and appears to hasten model development for cellular senescence studies.
Collapse
Affiliation(s)
- Yi-Nan Lee
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Hsueh-Hsiao Wang
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
| | - Cheng-Huang Su
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
| | - Hsin-I Lee
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
| | - Yen-Hung Chou
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan.,Institute of Biomedical Sciences, MacKay Medical College, New Taipei 25245, Taiwan
| | - Chin-Ling Hsieh
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Wen-Ting Liu
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Kuo-Tung Shu
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Kai-Ting Chang
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Hung-I Yeh
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
| | - Yih-Jer Wu
- Cardiovascular Center, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan.,Institute of Biomedical Sciences, MacKay Medical College, New Taipei 25245, Taiwan
| |
Collapse
|
15
|
Impact of Tuning the Surface Charge Distribution on Colloidal Iron Oxide Nanoparticle Toxicity Investigated in Caenorhabditis elegans. NANOMATERIALS 2021; 11:nano11061551. [PMID: 34208275 PMCID: PMC8230852 DOI: 10.3390/nano11061551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/09/2021] [Indexed: 01/31/2023]
Abstract
Assessing the toxic effect in living organisms remains a major issue for the development of safe nanomedicines and exposure of researchers involved in the synthesis, handling and manipulation of nanoparticles. In this study, we demonstrate that Caenorhabditis elegans could represent an in vivo model alternative to superior mammalians for the collection of several physiological functionality parameters associated to both short-term and long-term effects of colloidally stable nanoparticles even in absence of microbial feeding, usually reported to be necessary to ensure appropriate intake. Contextually, we investigated the impact of surface charge on toxicity of superparamagnetic iron oxide coated with a wrapping polymeric envelop that confers them optimal colloidal stability. By finely tuning the functional group composition of this shallow polymer–obtaining totally anionic, partially pegylated, partially anionic and partially cationic, respectively–we showed that the ideal surface charge organization to optimize safety of colloidal nanoparticles is the one containing both cationic and anionic groups. Our results are in accordance with previous evidence that zwitterionic nanoparticles allow long circulation, favorable distribution in the tumor area and optimal tumor penetration and thus support the hypothesis that zwitterionic iron oxide nanoparticles could be an excellent solution for diagnostic imaging and therapeutic applications in nanooncology.
Collapse
|
16
|
Picca A, Calvani R, Sirago G, Coelho-Junior HJ, Marzetti E. Molecular routes to sarcopenia and biomarker development: per aspera ad astra. Curr Opin Pharmacol 2021; 57:140-147. [PMID: 33721617 DOI: 10.1016/j.coph.2021.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/31/2020] [Accepted: 02/09/2021] [Indexed: 12/17/2022]
Abstract
Sarcopenia, the age-related decline in muscle mass and strength/function, is a prototypical geroscience condition. The dissection of muscle-specific molecular pathways through analyses of tissue biopsies has provided valuable insights into the pathophysiology of sarcopenia. However, such an approach is unsuitable for capturing the dynamic nature of the condition. Furthermore, the muscle sampling procedure may be perceived as burdensome especially by multimorbid, frail older adults. To overcome these limitations, sophisticated statistical methods have been devised for the simultaneous analysis of circulating factors related to the multiple domains of sarcopenia. This approach has shown potential for achieving a more comprehensive appraisal of the condition, unveiling new therapeutic targets, and identifying meaningful biomarkers. Here, we discuss the main pathogenetic pathways of sarcopenia, with a focus on mediators that are currently in the spotlight as biomarkers and potential treatment targets.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Giuseppe Sirago
- Department of Biomedical Sciences DBS, Università degli Studi di Padova, Padua, Italy
| | - Hélio José Coelho-Junior
- Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, Rome, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, Rome, Italy.
| |
Collapse
|
17
|
Picca A, Saini SK, Mankowski RT, Kamenov G, Anton SD, Manini TM, Buford TW, Wohlgemuth SE, Xiao R, Calvani R, Coelho-Júnior HJ, Landi F, Bernabei R, Hood DA, Marzetti E, Leeuwenburgh C. Altered Expression of Mitoferrin and Frataxin, Larger Labile Iron Pool and Greater Mitochondrial DNA Damage in the Skeletal Muscle of Older Adults. Cells 2020; 9:E2579. [PMID: 33276460 PMCID: PMC7760001 DOI: 10.3390/cells9122579] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial dysfunction and iron (Fe) dyshomeostasis are invoked among the mechanisms contributing to muscle aging, possibly via a detrimental mitochondrial-iron feed-forward loop. We quantified the labile Fe pool, Fe isotopes, and the expression of mitochondrial Fe handling proteins in muscle biopsies obtained from young and older adults. The expression of key proteins of mitochondrial quality control (MQC) and the abundance of the mitochondrial DNA common deletion (mtDNA4977) were also assessed. An inverse association was found between total Fe and the heavier Fe isotope (56Fe), indicating an increase in labile Fe abundance in cells with greater Fe content. The highest levels of labile Fe were detected in old participants with a Short Physical Performance Battery (SPPB) score ≤ 7 (low-functioning, LF). Protein levels of mitoferrin and frataxin were, respectively, higher and lower in the LF group relative to young participants and older adults with SPPB scores ≥ 11 (high-functioning, HF). The mtDNA4977 relative abundance was greater in old than in young participants, regardless of SPPB category. Higher protein levels of Pink1 were detected in LF participants compared with young and HF groups. Finally, the ratio between lipidated and non-lipidated microtubule-associated protein 1A/1B-light chain 3 (i.e., LC3B II/I), as well as p62 protein expression was lower in old participants regardless of SPPB scores. Our findings indicate that cellular and mitochondrial Fe homeostasis is perturbed in the aged muscle (especially in LF older adults), as reflected by altered levels of mitoferrin and frataxin, which, together with MQC derangements, might contribute to loss of mtDNA stability.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (R.C.); (F.L.); (R.B.)
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, SE-171 77 Stockholm, Sweden
| | - Sunil K. Saini
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32611, USA; (S.K.S.); (R.T.M.); (S.D.A.); (T.M.M.); (S.E.W.); (R.X.); (C.L.)
| | - Robert T. Mankowski
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32611, USA; (S.K.S.); (R.T.M.); (S.D.A.); (T.M.M.); (S.E.W.); (R.X.); (C.L.)
| | - George Kamenov
- Department of Geological Sciences, University of Florida, Gainesville, FL 32605, USA;
| | - Stephen D. Anton
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32611, USA; (S.K.S.); (R.T.M.); (S.D.A.); (T.M.M.); (S.E.W.); (R.X.); (C.L.)
| | - Todd M. Manini
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32611, USA; (S.K.S.); (R.T.M.); (S.D.A.); (T.M.M.); (S.E.W.); (R.X.); (C.L.)
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35205, USA;
| | - Stephanie E. Wohlgemuth
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32611, USA; (S.K.S.); (R.T.M.); (S.D.A.); (T.M.M.); (S.E.W.); (R.X.); (C.L.)
| | - Rui Xiao
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32611, USA; (S.K.S.); (R.T.M.); (S.D.A.); (T.M.M.); (S.E.W.); (R.X.); (C.L.)
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (R.C.); (F.L.); (R.B.)
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, SE-171 77 Stockholm, Sweden
| | - Hélio José Coelho-Júnior
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (R.C.); (F.L.); (R.B.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Roberto Bernabei
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (R.C.); (F.L.); (R.B.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - David A. Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada;
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (A.P.); (R.C.); (F.L.); (R.B.)
- Institute of Internal Medicine and Geriatrics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, Institute on Aging, University of Florida, Gainesville, FL 32611, USA; (S.K.S.); (R.T.M.); (S.D.A.); (T.M.M.); (S.E.W.); (R.X.); (C.L.)
| |
Collapse
|
18
|
Ijomone OM, Ifenatuoha CW, Aluko OM, Ijomone OK, Aschner M. The aging brain: impact of heavy metal neurotoxicity. Crit Rev Toxicol 2020; 50:801-814. [PMID: 33210961 DOI: 10.1080/10408444.2020.1838441] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aging process is accompanied by critical changes in cellular and molecular functions, which upset the homeostatic balance in the central nervous system. Accumulation of metals renders the brain susceptible to neurotoxic insults by mechanisms such as mitochondrial dysfunction, neuronal calcium-ion dyshomeostasis, buildup of damaged molecules, compromised DNA repair, reduction in neurogenesis, and impaired energy metabolism. These hallmarks have been identified to be responsible for neuronal injuries, resulting in several neurological disorders. Various studies have shown solid associations between metal accumulation, abnormal protein expressions, and pathogenesis of neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and Amyotrophic lateral sclerosis. This review highlights metals (such as manganese, zinc, iron, copper, and nickel) for their accumulation, and consequences in the development of neurological disorders, in relation to the aging brain.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.,Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Chibuzor W Ifenatuoha
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Oritoke M Aluko
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.,Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Olayemi K Ijomone
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.,Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology, Pediatrics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
19
|
Leukocyte telomere length is associated with iron overload in male adults with hereditary hemochromatosis. Biosci Rep 2020; 40:226596. [PMID: 33026063 PMCID: PMC7584811 DOI: 10.1042/bsr20201916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/12/2020] [Accepted: 09/11/2020] [Indexed: 11/17/2022] Open
Abstract
Background: Hereditary hemochromatosis (HH) is a primary iron overload (IO) condition. Absolute telomere length (ATL) is a marker of cellular aging and DNA damage associated with chronic diseases and mortality. Aim: To evaluate the relationship between ATL and IO in patients with HH. Methods: Cross-sectional study including 25 patients with HH: 8 with IO and 17 without IO (ferritin < 300 ng/ml) and 25 healthy controls. Inclusion criteria were: age > 18 years, male sex and HH diagnosis. Patients with diabetes or other endocrine and autoimmune diseases were excluded. ATL was measured by real-time PCR. Results: HH patients with IO were older (P<0.001) and showed higher ferritin concentration (P<0.001). Patients with HH, disregarding the iron status, showed higher glucose and body mass index (BMI) than controls (both P<0.01). ATL was shorter in patients with IO than controls [with IO: 8 (6–14), without IO: 13 (9–20), and controls: 19 (15–25) kilobase pairs, P<0.01]; with a linear trend within groups (P for trend <0.01). Differences in ATL remained statistically significant after adjusting by age, BMI and glucose (P<0.05). Discussion: Patients with IO featured shorter ATL while patients without IO showed only mild alterations vs. controls. Screening for IO is encouraged to prevent iron-associated cellular damage and early telomere attrition.
Collapse
|
20
|
Inoue H, Hanawa N, Katsumata SI, Aizawa Y, Katsumata-Tsuboi R, Tanaka M, Takahashi N, Uehara M. Iron deficiency negatively regulates protein methylation via the downregulation of protein arginine methyltransferase. Heliyon 2020; 6:e05059. [PMID: 33033759 PMCID: PMC7533365 DOI: 10.1016/j.heliyon.2020.e05059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/25/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022] Open
Abstract
Iron is an essential trace metal for all biological processes and plays a role in almost every aspect of body growth. Previously, we found that iron-depletion downregulated the expression of proteins, arginine methyltransferase-1 and 3 (PRMT1 and PRMT3), by an iron-specific chelator, deferoxamine (DFO), in rat liver FAO cell line using DNA microarray analysis (unpublished data). However, regulatory mechanisms underlying the association between iron deficiency and PRMT expression are unclear in vitro and in vivo. In the present study, we revealed that the treatment of cells with two iron-specific chelators, DFO and deferasirox (DFX), downregulated the gene and protein expression of PRMT1 and 3 as compared with the untreated cells. Subsequently, DFO and DFX treatments decreased protein methylation. Importantly, these effects were attenuated by a holo-transferrin treatment. Furthermore, weanling Wistar-strain rats were fed a control diet or an iron-deficient diet for 4 weeks. Dietary iron deficiency was found to decrease the concentration of hemoglobin and liver iron while increasing the heart weight. PRMT and protein methylation levels were also significantly reduced in the iron-deficient group as compared to the control group. To our knowledge, this is the first study to demonstrate that PRMT levels and protein methylation are reduced in iron-deficient models, in vitro and in vivo.
Collapse
Affiliation(s)
- Hirofumi Inoue
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Japan
| | - Nobuaki Hanawa
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, Japan
| | - Shin-Ichi Katsumata
- Department of Nutritional Science, Faculty of Applied Bioscience, Tokyo University of Agriculture, Japan
| | - Yumi Aizawa
- Department of Agricultural Chemistry, Faculty of Applied Biosciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Rie Katsumata-Tsuboi
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Japan
| | - Miori Tanaka
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Japan
| | - Nobuyuki Takahashi
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Japan
| | - Mariko Uehara
- Department of Nutritional Science and Food Safety, Faculty of Applied Bioscience, Tokyo University of Agriculture, Japan
| |
Collapse
|
21
|
Espósito BP, Martins AC, de Carvalho RRV, Aschner M. High throughput fluorimetric assessment of iron traffic and chelation in iron-overloaded Caenorhabditis elegans. Biometals 2020; 33:255-267. [PMID: 32979113 DOI: 10.1007/s10534-020-00250-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/16/2020] [Indexed: 01/31/2023]
Abstract
The nematode Caenorhabditis elegans (C. elegans) is a convenient tool to evaluate iron metabolism as it shares great orthology with human proteins involved in iron transport, in addition to being transparent and readily available. In this work, we describe how wild-type (N2) C. elegans nematodes in the first larval stage can be loaded with acetomethoxycalcein (CAL-AM) and study it as a whole-organism model for both iron speciation and chelator permeability of the labile iron pool (LIP). This model may be relevant for high throughput assessment of molecules intended for chelation therapy of iron overload diseases.
Collapse
Affiliation(s)
- Breno Pannia Espósito
- Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.
| | - Airton Cunha Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
22
|
Burton LH, Radakovich LB, Marolf AJ, Santangelo KS. Systemic iron overload exacerbates osteoarthritis in the strain 13 guinea pig. Osteoarthritis Cartilage 2020; 28:1265-1275. [PMID: 32629162 PMCID: PMC7484276 DOI: 10.1016/j.joca.2020.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Iron is emerging as a key player in aging-associated diseases due to its propensity for driving free radical formation. Studies examining the role of iron in the pathogenesis of primary osteoarthritis (OA) are limited. Our objective was to establish a direct relationship between excess iron and OA by administering iron dextran to a guinea pig strain with decreased propensity for developing this disease. DESIGN Twenty, 12-week-old Strain 13 guinea pigs received either iron dextran or dextran control intraperitoneally once weekly for 4 weeks; termination occurred at 16 weeks of age. Iron levels were determined systemically (serum and liver) and within diarthrodial joints [femoral head articular cartilage and infrapatellar fat pads (IFPs) of knee joints]. One knee was collected to score structural changes associated with OA via microcomputed tomography (microCT) and histology using published grading schemes. Articular cartilage and IFPs were harvested from contralateral knees for gene expression analyses. RESULTS Iron overload was confirmed systemically via increased serum iron and liver iron concentration. Articular cartilage and IFPs in the iron dextran group also had higher levels of iron. Excess iron worsened knee OA using both microCT and histologic scoring systems. Gene analyses revealed that exogenous iron altered the expression of iron trafficking proteins, select cytokines, and structural components of cartilage. CONCLUSION These results demonstrate that systemic iron overload caused cellular iron accumulation in the knee joint. This excess iron is associated with increased expression of local inflammatory mediators and early onset and progression of knee joint OA in Strain 13 animals.
Collapse
Affiliation(s)
- Lindsey H. Burton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Lauren B. Radakovich
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Angela J. Marolf
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Kelly S. Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
23
|
Zhou J, Jin Y, Lei Y, Liu T, Wan Z, Meng H, Wang H. Ferroptosis Is Regulated by Mitochondria in Neurodegenerative Diseases. NEURODEGENER DIS 2020; 20:20-34. [PMID: 32814328 DOI: 10.1159/000510083] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Neurodegenerative diseases are characterized by a gradual decline in motor and/or cognitive function caused by the selective degeneration and loss of neurons in the central nervous system, but their pathological mechanism is still unclear. Previous research has revealed that many forms of cell death, such as apoptosis and necrosis, occur in neurodegenerative diseases. Research in recent years has noticed that there is a new type of cell death in neurodegenerative diseases: ferroptosis. An increasing body of literature provides evidence for an involvement of ferroptosis in neurodegenerative diseases. SUMMARY In this article, we review a new form of cell death in neurodegenerative diseases: ferroptosis. Ferroptosis is defined as an iron-dependent form of regulated cell death, which occurs through the lethal accumulation of lipid-based reactive oxygen species when glutathione-dependent lipid peroxide repair systems are compromised. Several salient and established features of neurodegenerative diseases (including lipid peroxidation and iron dyshomeostasis) are consistent with ferroptosis, which means that ferroptosis may be involved in the progression of neurodegenerative diseases. In addition, as the center of energy metabolism in cells, mitochondria are also closely related to the regulation of iron homeostasis in the nervous system. At the same time, neurodegenerative diseases are often accompanied by degeneration of mitochondrial activity. Mitochondrial damage has been found to be involved in lipid peroxidation and iron dyshomeostasis in neurodegenerative diseases. Key Messages: Based on the summary of the related mechanisms of ferroptosis, we conclude that mitochondrial damage may affect neurodegenerative diseases by regulating many aspects of ferroptosis, including cell metabolism, iron dyshomeostasis, and lipid peroxidation.
Collapse
Affiliation(s)
- Juepu Zhou
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yao Jin
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yuhong Lei
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Tianyi Liu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Zheng Wan
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Hao Meng
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China,
| | - Honglei Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
24
|
Jenkins NL, James SA, Salim A, Sumardy F, Speed TP, Conrad M, Richardson DR, Bush AI, McColl G. Changes in ferrous iron and glutathione promote ferroptosis and frailty in aging Caenorhabditis elegans. eLife 2020; 9:e56580. [PMID: 32690135 PMCID: PMC7373428 DOI: 10.7554/elife.56580] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
All eukaryotes require iron. Replication, detoxification, and a cancer-protective form of regulated cell death termed ferroptosis, all depend on iron metabolism. Ferrous iron accumulates over adult lifetime in Caenorhabditis elegans. Here, we show that glutathione depletion is coupled to ferrous iron elevation in these animals, and that both occur in late life to prime cells for ferroptosis. We demonstrate that blocking ferroptosis, either by inhibition of lipid peroxidation or by limiting iron retention, mitigates age-related cell death and markedly increases lifespan and healthspan. Temporal scaling of lifespan is not evident when ferroptosis is inhibited, consistent with this cell death process acting at specific life phases to induce organismal frailty, rather than contributing to a constant aging rate. Because excess age-related iron elevation in somatic tissue, particularly in brain, is thought to contribute to degenerative disease, post-developmental interventions to limit ferroptosis may promote healthy aging.
Collapse
Affiliation(s)
- Nicole L Jenkins
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health and University of MelbourneParkvilleAustralia
| | | | - Agus Salim
- Department of Mathematics and Statistics, La Trobe UniversityBundooraAustralia
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Melbourne School of Population and Global Health, and School of Mathematics and Statistics, University of MelbourneMelbourneAustralia
| | - Fransisca Sumardy
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health and University of MelbourneParkvilleAustralia
| | - Terence P Speed
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Mathematics and Statistics, University of MelbourneMelbourneAustralia
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell DeathNeuherbergGermany
| | - Des R Richardson
- Department of Pathology and Bosch Institute, University of SydneySydneyAustralia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health and University of MelbourneParkvilleAustralia
| | - Gawain McColl
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health and University of MelbourneParkvilleAustralia
| |
Collapse
|
25
|
Mitochondrial ROS-Modulated mtDNA: A Potential Target for Cardiac Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9423593. [PMID: 32308810 PMCID: PMC7139858 DOI: 10.1155/2020/9423593] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/05/2020] [Accepted: 03/16/2020] [Indexed: 02/05/2023]
Abstract
Mitochondrial DNA (mtDNA) damage is associated with the development of cardiovascular diseases. Cardiac aging plays a central role in cardiovascular diseases. There is accumulating evidence linking cardiac aging to mtDNA damage, including mtDNA mutation and decreased mtDNA copy number. Current wisdom indicates that mtDNA is susceptible to damage by mitochondrial reactive oxygen species (mtROS). This review presents the cellular and molecular mechanisms of cardiac aging, including autophagy, chronic inflammation, mtROS, and mtDNA damage, and the effects of mitochondrial biogenesis and oxidative stress on mtDNA. The importance of nucleoid-associated proteins (Pol γ), nuclear respiratory factors (NRF1 and NRF2), the cGAS-STING pathway, and the mitochondrial biogenesis pathway concerning the development of mtDNA damage during cardiac aging is discussed. Thus, the repair of damaged mtDNA provides a potential clinical target for preventing cardiac aging.
Collapse
|
26
|
Advanced Age Is Associated with Iron Dyshomeostasis and Mitochondrial DNA Damage in Human Skeletal Muscle. Cells 2019; 8:cells8121525. [PMID: 31783583 PMCID: PMC6953082 DOI: 10.3390/cells8121525] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Whether disruption of iron metabolism is implicated in human muscle aging is presently unclear. We explored the relationship among iron metabolism, muscle mitochondrial homeostasis, inflammation, and physical function in older adults and young controls. Eleven young and 23 older men and women were included. Older adults were classified into high-functioning (HF) and low-functioning (LF) groups according to their Short Physical Performance Battery score. Vastus lateralis muscle biopsies were assayed for total iron content, expression of 8-oxoguanine and DNA glycosylase (OGG1), 3-nitrotyrosine (3-NT) levels, and mitochondrial DNA (mtDNA) content and damage. Circulating ferritin and hepcidin levels were also quantified. Muscle iron levels were greater in the old group. Protein expression of transferrin receptor 1, Zrt-Irt-like protein (ZIP) 8, and ZIP14 were lower in old participants. Circulating levels of ferritin, hepcidin, interleukin 6 (IL6), and C-reactive protein were higher in the old group. Old participants showed lower mtDNA content and greater mtDNA damage. OGG1 protein expression declined with age, whereas 3-NT levels were greater in old participants. Finally, a negative correlation was determined between ZIP14 expression and circulating IL6 levels in LF older adults. None of assayed parameters differed between HF and LF participants. Our findings suggest that muscle iron homeostasis is altered in old age, which might contribute to loss of mtDNA stability. Muscle iron metabolism may therefore represent a target for interventions against muscle aging.
Collapse
|
27
|
Gouda W, Mageed L, Abd El Dayem SM, Ashour E, Afify M. Evaluation of pro-inflammatory and anti-inflammatory cytokines in type 1 diabetes mellitus. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2018; 42:14. [DOI: 10.1186/s42269-018-0016-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/08/2018] [Indexed: 09/02/2023]
|
28
|
Inoue H, Hanawa N, Katsumata-Tsuboi R, Katsumata SI, Takahashi N, Uehara M. Down-regulation of senescence marker protein 30 by iron-specific chelator deferoxamine drives cell senescence. Biosci Biotechnol Biochem 2018; 82:900-903. [DOI: 10.1080/09168451.2018.1440190] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract
To our knowledge, this is the first study to report down-regulation of senescence marker protein 30 (SMP30) by iron-specific chelator deferoxamine (DFO) on FAO cell senescence, using a DNA microarray. Furthermore, DFO treatment increased senescence marker β-galactosidase activity, whereas this activity was attenuated by overexpression of SMP30. Our data suggested that down-regulation of SMP30 drives cell senescence in iron-chelated condition.
Collapse
Affiliation(s)
- Hirofumi Inoue
- Faculty of Applied Bioscience, Department of Nutritional Science and Food Safety, Tokyo University of Agriculture, Tokyo, Japan
| | - Nobuaki Hanawa
- Faculty of Applied Bioscience, Department of Food and Nutritional Science, Tokyo University of Agriculture, Tokyo, Japan
| | - Rie Katsumata-Tsuboi
- Faculty of Applied Bioscience, Department of Nutritional Science and Food Safety, Tokyo University of Agriculture, Tokyo, Japan
| | - Shi-Ichi Katsumata
- Faculty of Applied Bioscience, Department of Food and Nutritional Science, Tokyo University of Agriculture, Tokyo, Japan
| | - Nobuyuki Takahashi
- Faculty of Applied Bioscience, Department of Nutritional Science and Food Safety, Tokyo University of Agriculture, Tokyo, Japan
| | - Mariko Uehara
- Faculty of Applied Bioscience, Department of Nutritional Science and Food Safety, Tokyo University of Agriculture, Tokyo, Japan
| |
Collapse
|
29
|
Martín-Fernández B, Gredilla R. Mitochondrial oxidative stress and cardiac ageing. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2018; 30:74-83. [PMID: 29398015 DOI: 10.1016/j.arteri.2017.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 10/18/2022]
Abstract
According with different international organizations, cardiovascular diseases are becoming the first cause of death in western countries. Although exposure to different risk factors, particularly those related to lifestyle, contribute to the etiopathogenesis of cardiac disorders, the increase in average lifespan and aging are considered major determinants of cardiac diseases events. Mitochondria and oxidative stress have been pointed out as relevant factors both in heart aging and in the development of cardiac diseases such as heart failure, cardiac hypertrophy and diabetic cardiomyopathy. During aging, cellular processes related with mitochondrial function, such as bioenergetics, apoptosis and inflammation are altered leading to cardiac dysfunction. Increasing our knowledge about the mitochondrial mechanisms related with the aging process, will provide new strategies in order to improve this process, particularly the cardiovascular ones.
Collapse
Affiliation(s)
- Beatriz Martín-Fernández
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España.
| | - Ricardo Gredilla
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| |
Collapse
|
30
|
Carraro U. Exciting perspectives for Translational Myology in the Abstracts of the 2018Spring PaduaMuscleDays: Giovanni Salviati Memorial - Chapter II - Abstracts of March 15, 2018. Eur J Transl Myol 2018; 28:7364. [PMID: 30057726 PMCID: PMC6047880 DOI: 10.4081/ejtm.2018.7364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/20/2018] [Indexed: 12/03/2022] Open
Abstract
Myologists working in Padua (Italy) were able to continue a half-century tradition of studies of skeletal muscles, that started with a research on fever, specifically if and how skeletal muscle contribute to it by burning bacterial toxin. Beside main publications in high-impact-factor journals by Padua myologists, I hope to convince readers (and myself) of the relevance of the editing Basic and Applied Myology (BAM), retitled from 2010 European Journal of Translational Myology (EJTM), of the institution of the Interdepartmental Research Center of Myology of the University of Padova (CIR-Myo), and of a long series of International Conferences organized in Euganei Hills and Padova, that is, the PaduaMuscleDays. The 2018Spring PaduaMuscleDays (2018SpPMD), were held in Euganei Hills and Padua (Italy), in March 14-17, and were dedicated to Giovanni Salviati. The main event of the "Giovanni Salviati Memorial", was held in the Aula Guariento, Accademia Galileiana di Scienze, Lettere ed Arti of Padua to honor a beloved friend and excellent scientist 20 years after his premature passing. Using the words of Prof. Nicola Rizzuto, we all share his believe that Giovanni "will be remembered not only for his talent and originality as a biochemist, but also for his unassuming and humanistic personality, a rare quality in highly successful people like Giovanni. The best way to remember such a person is to gather pupils and colleagues, who shared with him the same scientific interests and ask them to discuss recent advances in their own fields, just as Giovanni have liked to do". Since Giovanni's friends sent many abstracts still influenced by their previous collaboration with him, all the Sessions of the 2018SpPMD reflect both to the research aims of Giovanni Salviati and the traditional topics of the PaduaMuscleDays, that is, basics and applications of physical, molecular and cellular strategies to maintain or recover functions of skeletal muscles. The translational researches summarized in the 2018SpPMD Abstracts are at the appropriate high level to attract approval of Ethical Committees, the interest of International Granting Agencies and approval for publication in top quality, international journals. In this chapter II are listed the abstracts of the March 15, 2018 Padua Muscle Day. All 2018SpPMD Abstracts are indexed at the end of the Chapter IV.
Collapse
Affiliation(s)
- Ugo Carraro
- Laboratory of Translational Myology, Department of Biomedical Sciences, University of Padova
- A&C M-C Foundation for Translational Myology, Padova
- IRCCS Fondazione Ospedale San Camillo, Venezia-Lido, Italy
| |
Collapse
|
31
|
Teixeira J, Cagide F, Benfeito S, Soares P, Garrido J, Baldeiras I, Ribeiro JA, Pereira CM, Silva AF, Andrade PB, Oliveira PJ, Borges F. Development of a Mitochondriotropic Antioxidant Based on Caffeic Acid: Proof of Concept on Cellular and Mitochondrial Oxidative Stress Models. J Med Chem 2017; 60:7084-7098. [DOI: 10.1021/acs.jmedchem.7b00741] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- José Teixeira
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
- CNC—Center
for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech
Building, Biocant Park, Cantanhede 3060-197, Portugal
| | - Fernando Cagide
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Sofia Benfeito
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Pedro Soares
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Jorge Garrido
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
- Department
of Chemical Engineering, School of Engineering (ISEP), Polytechnic Institute of Porto, Porto 4200-072, Portugal
| | - Inês Baldeiras
- Faculty
of Medicine, University of Coimbra, Coimbra 3004-504, Portugal
- Laboratory
of Neurochemistry, Coimbra University Hospital (CHUC), Coimbra 3000-075, Portugal
| | - José A. Ribeiro
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Carlos M. Pereira
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - António F. Silva
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Paula B. Andrade
- REQUIMTE/LAQV-Laboratory
of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Paulo J. Oliveira
- CNC—Center
for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech
Building, Biocant Park, Cantanhede 3060-197, Portugal
| | - Fernanda Borges
- CIQUP/Department
of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| |
Collapse
|
32
|
Pette D. What Can be Learned from the Time Course of Changes in Low-Frequency Stimulated Muscle? Eur J Transl Myol 2017; 27:6723. [PMID: 28713537 PMCID: PMC5505094 DOI: 10.4081/ejtm.2017.6723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Dirk Pette
- Department of Biology, University of Konstanz, Germany
| |
Collapse
|
33
|
Lopez TE, Pham HM, Nguyen BV, Tahmasian Y, Ramsden S, Coskun V, Schriner SE, Jafari M. Green tea polyphenols require the mitochondrial iron transporter, mitoferrin, for lifespan extension in Drosophila melanogaster. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2016; 93:210-221. [PMID: 27696504 PMCID: PMC5121014 DOI: 10.1002/arch.21353] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Green tea has been found to increase the lifespan of various experimental animal models including the fruit fly, Drosophila melanogaster. High in polyphenolic content, green tea has been shown to reduce oxidative stress in part by its ability to bind free iron, a micronutrient that is both essential for and toxic to all living organisms. Due to green tea's iron-binding properties, we questioned whether green tea acts to increase the lifespan of the fruit fly by modulating iron regulators, specifically, mitoferrin, a mitochondrial iron transporter, and transferrin, found in the hemolymph of flies. Publicly available hypomorph mutants for these iron regulators were utilized to investigate the effect of green tea on lifespan and fertility. We identified that green tea could not increase the lifespan of mitoferrin mutants but did rescue the reduced male fertility phenotype. The effect of green tea on transferrin mutant lifespan and fertility were comparable to w1118 flies, as observed in our previous studies, in which green tea increased male fly lifespan and reduced male fertility. Expression levels in both w1118 flies and mutant flies, supplemented with green tea, showed an upregulation of mitoferrin but not transferrin. Total body and mitochondrial iron levels were significantly reduced by green tea supplementation in w1118 and mitoferrin mutants but not transferrin mutant flies. Our results demonstrate that green tea may act to increase the lifespan of Drosophila in part by the regulation of mitoferrin and reduction of mitochondrial iron.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mahtab Jafari
- Corresponding Author: Dr. Mahtab Jafari, Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697,
| |
Collapse
|
34
|
Walker T, Michaelides C, Ekonomou A, Geraki K, Parkes HG, Suessmilch M, Herlihy AH, Crum WR, So PW. Dissociation between iron accumulation and ferritin upregulation in the aged substantia nigra: attenuation by dietary restriction. Aging (Albany NY) 2016; 8:2488-2508. [PMID: 27743512 PMCID: PMC5115902 DOI: 10.18632/aging.101069] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/26/2016] [Indexed: 01/13/2023]
Abstract
Despite regulation, brain iron increases with aging and may enhance aging processes including neuroinflammation. Increases in magnetic resonance imaging transverse relaxation rates, R2 and R2*, in the brain have been observed during aging. We show R2 and R2* correlate well with iron content via direct correlation to semi-quantitative synchrotron-based X-ray fluorescence iron mapping, with age-associated R2 and R2* increases reflecting iron accumulation. Iron accumulation was concomitant with increased ferritin immunoreactivity in basal ganglia regions except in the substantia nigra (SN). The unexpected dissociation of iron accumulation from ferritin-upregulation in the SN suggests iron dyshomeostasis in the SN. Occurring alongside microgliosis and astrogliosis, iron dyshomeotasis may contribute to the particular vulnerability of the SN. Dietary restriction (DR) has long been touted to ameliorate brain aging and we show DR attenuated age-related in vivo R2 increases in the SN over ages 7 - 19 months, concomitant with normal iron-induction of ferritin expression and decreased microgliosis. Iron is known to induce microgliosis and conversely, microgliosis can induce iron accumulation, which of these may be the initial pathological aging event warrants further investigation. We suggest iron chelation therapies and anti-inflammatory treatments may be putative 'anti-brain aging' therapies and combining these strategies may be synergistic.
Collapse
Affiliation(s)
- Thomas Walker
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Christos Michaelides
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Antigoni Ekonomou
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Kalotina Geraki
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, Oxfordshire, United Kingdom
| | - Harold G Parkes
- CR-UK Clinical MR Research Group, Institute of Cancer Research, London, United Kingdom
| | - Maria Suessmilch
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | | | - William R Crum
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
35
|
Aydemir TB, Troche C, Kim J, Kim MH, Teran OY, Leeuwenburgh C, Cousins RJ. Aging amplifies multiple phenotypic defects in mice with zinc transporter Zip14 (Slc39a14) deletion. Exp Gerontol 2016; 85:88-94. [PMID: 27647172 DOI: 10.1016/j.exger.2016.09.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/15/2016] [Accepted: 09/14/2016] [Indexed: 12/17/2022]
Abstract
Inflammation and zinc dyshomeostasis are two common hallmarks of aging. A major zinc transporter ZIP14 (slc39a14) is upregulated by proinflammatory stimuli, e.g. interleukin-6. We have evaluated the influence of age on the Zip14 KO phenotype using wild-type (WT) and Zip14 knockout (KO) mice. Aging produced a major increase in serum IL-6 concentrations that was dramatically augmented in the Zip14 KO mice. In keeping with enhanced serum IL-6 concentrations, aging produced tissue-specific increases in zinc concentration of skeletal muscle and white adipose tissue. Metabolic endotoxemia produced by Zip14 ablation is maintained in aged KO mice. Muscle non-heme iron (NHI) was increased in aged WT mice but not in aged Zip14 KO mice demonstrating NHI uptake by muscle is ZIP14-dependent and increases with age. NF-κB and STAT3 activation was greater in aged mice, but was tissue specific and inversely related to tissue zinc. Micro-CT analysis revealed that Zip14 KO mice had markedly reduced trabecular bone that was greatly amplified with aging. These results demonstrate that the inflammation-responsive zinc transporter ZIP14 has phenotypic effects that are amplified with aging.
Collapse
Affiliation(s)
- Tolunay Beker Aydemir
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agriculture and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Catalina Troche
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agriculture and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Jinhee Kim
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agriculture and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Min-Hyun Kim
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agriculture and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Oriana Y Teran
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agriculture and Life Sciences, University of Florida, Gainesville, FL, United States
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, College of Medicine, University of Florida, Gainesville, FL, United States; Institute on Aging, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Robert J Cousins
- Food Science and Human Nutrition Department, Center for Nutritional Sciences, College of Agriculture and Life Sciences, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
36
|
Martín-Fernández B, Gredilla R. Mitochondria and oxidative stress in heart aging. AGE (DORDRECHT, NETHERLANDS) 2016; 38:225-238. [PMID: 27449187 PMCID: PMC5061683 DOI: 10.1007/s11357-016-9933-y] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 07/12/2016] [Indexed: 05/06/2023]
Abstract
As average lifespan of humans increases in western countries, cardiac diseases become the first cause of death. Aging is among the most important risk factors that increase susceptibility for developing cardiovascular diseases. The heart has very aerobic metabolism, and is highly dependent on mitochondrial function, since mitochondria generate more than 90 % of the intracellular ATP consumed by cardiomyocytes. In the last few decades, several investigations have supported the relevance of mitochondria and oxidative stress both in heart aging and in the development of cardiac diseases such as heart failure, cardiac hypertrophy, and diabetic cardiomyopathy. In the current review, we compile different studies corroborating this role. Increased mitochondria DNA instability, impaired bioenergetic efficiency, enhanced apoptosis, and inflammation processes are some of the events related to mitochondria that occur in aging heart, leading to reduced cellular survival and cardiac dysfunction. Knowing the mitochondrial mechanisms involved in the aging process will provide a better understanding of them and allow finding approaches to more efficiently improve this process.
Collapse
Affiliation(s)
- Beatriz Martín-Fernández
- Department of Physiology, Faculty of Medicine, Complutense University, Plaza Ramon y Cajal s/n, 28040, Madrid, Spain.
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Plaza Ramon y Cajal s/n, 28040, Madrid, Spain.
| |
Collapse
|
37
|
Shawn, the Drosophila Homolog of SLC25A39/40, Is a Mitochondrial Carrier That Promotes Neuronal Survival. J Neurosci 2016; 36:1914-29. [PMID: 26865615 DOI: 10.1523/jneurosci.3432-15.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Mitochondria play an important role in the regulation of neurotransmission, and mitochondrial impairment is a key event in neurodegeneration. Cells rely on mitochondrial carrier proteins of the SLC25 family to shuttle ions, cofactors, and metabolites necessary for enzymatic reactions. Mutations in these carriers often result in rare but severe pathologies in the brain, and some of the genes, including SLC25A39 and SLC25A40, reside in susceptibility loci of severe forms of epilepsy. However, the role of most of these carriers has not been investigated in neurons in vivo. We identified shawn, the Drosophila homolog of SLC25A39 and SLC25A40, in a genetic screen to identify genes involved in neuronal function. Shawn localizes to mitochondria, and missense mutations result in an accumulation of reactive oxygen species, mitochondrial dysfunction, and neurodegeneration. Shawn regulates metal homeostasis, and we found in shawn mutants increased levels of manganese, calcium, and mitochondrial free iron. Mitochondrial mutants often cannot maintain synaptic transmission under demanding conditions, but shawn mutants do, and they also do not display endocytic defects. In contrast, shawn mutants harbor a significant increase in neurotransmitter release. Our work provides the first functional annotation of these essential mitochondrial carriers in the nervous system, and the results suggest that metal imbalances and mitochondrial dysfunction may contribute to defects in synaptic transmission and neuronal survival. SIGNIFICANCE STATEMENT We describe for the first time the role of the mitochondrial carrier Shawn/SLC25A39/SLC25A40 in the nervous system. In humans, these genes reside in susceptibility loci for epilepsy, and, in flies, we observe neuronal defects related to mitochondrial dysfunction and metal homeostasis defects. Interestingly, shawn mutants also harbor increased neurotransmitter release and neurodegeneration. Our data suggest a connection between maintaining a correct metal balance and mitochondrial function to regulate neuronal survival and neurotransmitter release.
Collapse
|
38
|
Boero M, Pagliaro P, Tullio F, Pellegrino RM, Palmieri A, Ferbo L, Saglio G, De Gobbi M, Penna C, Roetto A. A comparative study of myocardial molecular phenotypes of two tfr2β null mice: role in ischemia/reperfusion. Biofactors 2015; 41:360-71. [PMID: 26458496 DOI: 10.1002/biof.1237] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/05/2015] [Indexed: 01/01/2023]
Abstract
Transferrin receptor 2 (Tfr2) is an iron-modulator transcribed in two isoforms, Tfr2α and Tfr2β. The latter is expressed in the heart. We obtained two mouse models with silencing of Tfr2β: one with a normal systemic iron amount (SIA), i.e., Tfr2-KI, and the other, i.e., LCKO-KI, with high SIA due to hepatic Tfr2α silencing. We aimed to assess whether Tfr2β might play a role in myocardial injury and whether Tfr2β silencing might modify proteins of iron metabolism, antioxidant, apoptotic, and survival enzyme activities in the heart undergoing ischemia/reperfusion (I/R). Isolated hearts of wild-type (WT) and Tfr2-null mice were studied before or after an I/R protocol, and proteins/RNA analyzed by Western blot and/or quantitative PCR. Tfr2β increased in WT hearts subject to I/R, and both Tfr2β null mice hearts were protected against I/R injury (about 40% smaller infarct-size compared to WT hearts). RISK kinases (ERK1/2-AKT-PKCε) were found up-regulated after I/R in Tfr2-KI, whereas SAFE enzyme (Stat3) and GSK3β resulted phosphorylated during I/R in LCKO-KI hearts. While HO-1 and HIF-2a were high in both Tfr2β-null mice, Catalase, and proapoptotic factors were upregulated only in LCKO-KI. Finally, Tfr2-KI hearts presented an increased Ferritin-H and a decreased Ferroportin1, whereas LCKO-KI hearts displayed an upregulation of Ferritin-L chain and DMT1/Hamp-RNA. In conclusion, Tfr2β isoform is involved in cardiac iron metabolism and its silencing leads to a protected phenotype (antioxidants, RISK, and/or SAFE upregulation) against I/R challenging. Iron-dependent signals involved in cardioprotection seem to be positively affected by Tfr2β downregulation and subsequent Ferritins upregulation.
Collapse
Affiliation(s)
- Martina Boero
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
- Cardiovascular Physiology Lab (Torino), National Institute for Cardiovascular Researches (INRC), Bologna, Italy
| | - Francesca Tullio
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
- Cardiovascular Physiology Lab (Torino), National Institute for Cardiovascular Researches (INRC), Bologna, Italy
| | - Rosa M Pellegrino
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
| | - Antonietta Palmieri
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
| | - Ludovica Ferbo
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
| | - Marco De Gobbi
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
- Cardiovascular Physiology Lab (Torino), National Institute for Cardiovascular Researches (INRC), Bologna, Italy
| | - Antonella Roetto
- Department of Clinical and Biological Sciences, University of Torino, AOU San Luigi Gonzaga, Orbassano, Torino, Italy
| |
Collapse
|
39
|
Iron-induced damage in cardiomyopathy: oxidative-dependent and independent mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:230182. [PMID: 25878762 PMCID: PMC4387903 DOI: 10.1155/2015/230182] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/06/2015] [Accepted: 03/15/2015] [Indexed: 02/08/2023]
Abstract
The high incidence of cardiomyopathy in patients with hemosiderosis, particularly in transfusional iron overload, strongly indicates that iron accumulation in the heart plays a major role in the process leading to heart failure. In this context, iron-mediated generation of noxious reactive oxygen species is believed to be the most important pathogenetic mechanism determining cardiomyocyte damage, the initiating event of a pathologic progression involving apoptosis, fibrosis, and ultimately cardiac dysfunction. However, recent findings suggest that additional mechanisms involving subcellular organelles and inflammatory mediators are important factors in the development of this disease. Moreover, excess iron can amplify the cardiotoxic effect of other agents or events. Finally, subcellular misdistribution of iron within cardiomyocytes may represent an additional pathway leading to cardiac injury. Recent advances in imaging techniques and chelators development remarkably improved cardiac iron overload detection and treatment, respectively. However, increased understanding of the pathogenic mechanisms of iron overload cardiomyopathy is needed to pave the way for the development of improved therapeutic strategies.
Collapse
|
40
|
Chaturvedi R, Banerjee S, Chattopadhyay P, Bhattacharjee CR, Raul P, Borah K. High iron accumulation in hair and nail of people living in iron affected areas of Assam, India. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2014; 110:216-20. [PMID: 25261608 DOI: 10.1016/j.ecoenv.2014.08.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 08/21/2014] [Accepted: 08/23/2014] [Indexed: 05/12/2023]
Abstract
Human populace of Assam, India repeatedly exposed to high concentration of iron in groundwater results in adverse health effects like hemochromatosis, liver cirrhosis and siderosis. In the present study, human hair and nail analysis were carried out to establish a possible relationship between iron toxicity and its deposition among the residents. Nail and hair iron concentrations ranged from 28.2 to 1046μgg(-1) (n=114) and 26.5-838 (n=108)μgg(-1) levels, respectively, among all the study participants. The iron content of the groundwater (421-5340μgL(-1)) (n=64) used for drinking purposes was positively correlated with both nail (r=0.788, p<0.0001) and hair (r=0.709, p<0.0001) iron concentrations. Age- and sex-matched controls corresponding to each group were selected from population residing in other parts of the country where groundwater does not have excess iron. All the study groups included population drinking iron-contaminated water above the WHO/BIS limit (>300µgL(-1)) for 5 years (Group 1), for more than 5-10 years (Group 2) and for more than 10 years (Group 3). Results suggested that the participants consuming groundwater exceeding the WHO limit of iron had significantly more iron accumulation than those using groundwater containing ≤300μgL(-1) iron (p<0.01). There was statistically higher concentration of iron in the nail samples than the hair samples in all the study groups (p<0.01). There was a positive correlation in iron concentration and the residence time of the participants (p<0.01). Iron levels in the male participants were significantly higher than the female participants in the present study (p<0.01). The current findings are sufficiently compelling to warrant more extensive study of iron exposure through drinking water and adverse effects to the human in the areas where iron concentration is high.
Collapse
Affiliation(s)
- Richa Chaturvedi
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam; Department of Chemistry, Assam University, Silchar-788001, Assam
| | - Saumen Banerjee
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam.
| | | | | | - Prasanta Raul
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam
| | - Kusum Borah
- Department of Chemistry, Defence Research Laboratory, Tezpur-784001, Assam
| |
Collapse
|
41
|
Nkuipou-Kenfack E, Koeck T, Mischak H, Pich A, Schanstra JP, Zürbig P, Schumacher B. Proteome analysis in the assessment of ageing. Ageing Res Rev 2014; 18:74-85. [PMID: 25257180 DOI: 10.1016/j.arr.2014.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/05/2014] [Accepted: 09/15/2014] [Indexed: 12/14/2022]
Abstract
Based on demographic trends, the societies in many developed countries are facing an increasing number and proportion of people over the age of 65. The raise in elderly populations along with improved health-care will be concomitant with an increased prevalence of ageing-associated chronic conditions like cardiovascular, renal, and respiratory diseases, arthritis, dementia, and diabetes mellitus. This is expected to pose unprecedented challenges both for individuals and societies and their health care systems. An ultimate goal of ageing research is therefore the understanding of physiological ageing and the achievement of 'healthy' ageing by decreasing age-related pathologies. However, on a molecular level, ageing is a complex multi-mechanistic process whose contributing factors may vary individually, partly overlap with pathological alterations, and are often poorly understood. Proteome analysis potentially allows modelling of these multifactorial processes. This review summarises recent proteomic research on age-related changes identified in animal models and human studies. We combined this information with pathway analysis to identify molecular mechanisms associated with ageing. We identified some molecular pathways that are affected in most or even all organs and others that are organ-specific. However, appropriately powered studies are needed to confirm these findings based in in silico evaluation.
Collapse
Affiliation(s)
- Esther Nkuipou-Kenfack
- Mosaiques Diagnostics GmbH, Hannover, Germany; Hannover Medical School, Core Facility Proteomics, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | | | - Harald Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany; BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Andreas Pich
- Hannover Medical School, Core Facility Proteomics, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier, Toulouse, France
| | | | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease and Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD) Research Center, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| |
Collapse
|
42
|
DiTommaso T, Cottle DL, Pearson HB, Schlüter H, Kaur P, Humbert PO, Smyth IM. Keratin 76 is required for tight junction function and maintenance of the skin barrier. PLoS Genet 2014; 10:e1004706. [PMID: 25340345 PMCID: PMC4207637 DOI: 10.1371/journal.pgen.1004706] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 08/26/2014] [Indexed: 11/18/2022] Open
Abstract
Keratins are cytoskeletal intermediate filament proteins that are increasingly being recognised for their diverse cellular functions. Here we report the consequences of germ line inactivation of Keratin 76 (Krt76) in mice. Homozygous disruption of this epidermally expressed gene causes neonatal skin flaking, hyperpigmentation, inflammation, impaired wound healing, and death prior to 12 weeks of age. We show that this phenotype is associated with functionally defective tight junctions that are characterised by mislocalization of the integral protein CLDN1. We further demonstrate that KRT76 interacts with CLDN1 and propose that this interaction is necessary to correctly position CLDN1 in tight junctions. The mislocalization of CLDN1 has been associated in various dermopathies, including the inflammatory disease, psoriasis. These observations establish a previously unknown connection between the intermediate filament cytoskeleton network and tight junctions and showcase Krt76 null mice as a possible model to study aberrant tight junction driven skin diseases.
Collapse
Affiliation(s)
- Tia DiTommaso
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Denny L. Cottle
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Helen B. Pearson
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Holger Schlüter
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Pritinder Kaur
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Anatomy & Neuroscience, University of Melbourne, Melbourne, Australia
| | - Patrick O. Humbert
- Research Division, The Sir Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Australia
- Department of Pathology, University of Melbourne, Melbourne, Australia
| | - Ian M. Smyth
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
- * E-mail:
| |
Collapse
|
43
|
Yang X, Xu Y. Mutations in the ATP13A2 gene and Parkinsonism: a preliminary review. BIOMED RESEARCH INTERNATIONAL 2014; 2014:371256. [PMID: 25197640 PMCID: PMC4147200 DOI: 10.1155/2014/371256] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/14/2014] [Accepted: 07/28/2014] [Indexed: 02/05/2023]
Abstract
Parkinson's disease (PD) is a major neurodegenerative disorder for which the etiology and pathogenesis remain as elusive as for Alzheimer's disease. PD appears to be caused by genetic and environmental factors, and pedigree and cohort studies have identified numerous susceptibility genes and loci related to PD. Autosomal recessive mutations in the genes Parkin, Pink1, DJ-1, ATP13A2, PLA2G6, and FBXO7 have been linked to PD susceptibility. Such mutations in ATP13A2, also named PARK9, were first identified in 2006 in a Chilean family and are associated with a juvenile-onset, levodopa-responsive type of Parkinsonism called Kufor-Rakeb syndrome (KRS). KRS involves pyramidal degeneration, supranuclear palsy, and cognitive impairment. Here we review current knowledge about the ATP13A2 gene, clinical characteristics of patients with PD-associated ATP13A2 mutations, and models of how the ATP13A2 protein may help prevent neurodegeneration by inhibiting α-synuclein aggregation and supporting normal lysosomal and mitochondrial function. We also discuss another ATP13A2 mutation that is associated with the family of neurodegenerative disorders called neuronal ceroid lipofuscinoses (NCLs), and we propose a single pathway whereby ATP13A2 mutations may contribute to NCLs and Parkinsonism. Finally, we highlight how studies of mutations in this gene may provide new insights into PD pathogenesis and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Xinglong Yang
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| |
Collapse
|
44
|
Obanda V, Omondi GP, Chiyo PI. The influence of body mass index, age and sex on inflammatory disease risk in semi-captive Chimpanzees. PLoS One 2014; 9:e104602. [PMID: 25121995 PMCID: PMC4133249 DOI: 10.1371/journal.pone.0104602] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/15/2014] [Indexed: 01/25/2023] Open
Abstract
Obesity and ageing are emerging issues in the management of captive primates, including Chimpanzees, Pan troglodytes. Studies on humans show that obesity and old age can independently increase the risk of inflammatory-associated diseases indicated by elevated levels of pro-inflammatory cells and proteins in the blood of older or obese compared to levels in younger or non-obese individuals. In humans, sex can influence the outcomes of these risks. Health management of these problems in chimpanzee populations requires an understanding of similarities and differences of factors influencing inflammatory disease risks in humans and in chimpanzees. We examined the relationship between age, sex and Body Mass Index (BMI) with hematological biomarkers of inflammatory disease risk established for humans which include the neutrophil to lymphocyte ratio (NLR), and neutrophil, white blood cell (WBC), platelet microparticle and platelet counts. We found that higher values of NLR, neutrophil count and platelet microparticle count were associated with higher BMI values and older age indicating increased inflammation risk in these groups; a similar pattern to humans. There was a strong sex by age interaction on inflammation risk, with older males more at risk than older females. In contrast to human studies, total WBC count was not influenced by BMI, but like humans, WBC and platelet counts were lower in older individuals compared to younger individuals. Our findings are similar to those of humans and suggest that further insight on managing chimpanzees can be gained from extensive studies of ageing and obesity in humans. We suggest that managing BMI should be an integral part of health management in captive chimpanzee populations in order to partially reduce the risk of diseases associated with inflammation. These results also highlight parallels in inflammation risk between humans and chimpanzees and have implications for understanding the evolution of inflammation related diseases in apes.
Collapse
Affiliation(s)
- Vincent Obanda
- Veterinary Services Department, Kenya Wildlife Service, Nairobi, Kenya
- Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - George Paul Omondi
- Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
- Ol Pejeta Conservancy, Private Bag, Nanyuki, Kenya
| | - Patrick Ilukol Chiyo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
45
|
Moeller M, Hirose M, Mueller S, Roolf C, Baltrusch S, Ibrahim S, Junghanss C, Wolkenhauer O, Jaster R, Köhling R, Kunz M, Tiedge M, Schofield PN, Fuellen G. Inbred mouse strains reveal biomarkers that are pro-longevity, antilongevity or role switching. Aging Cell 2014; 13:729-38. [PMID: 24862908 PMCID: PMC4326954 DOI: 10.1111/acel.12226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2014] [Indexed: 01/23/2023] Open
Abstract
Traditionally, biomarkers of aging are classified as either pro-longevity or antilongevity. Using longitudinal data sets from the large-scale inbred mouse strain study at the Jackson Laboratory Nathan Shock Center, we describe a protocol to identify two kinds of biomarkers: those with prognostic implication for lifespan and those with longitudinal evidence. Our protocol also identifies biomarkers for which, at first sight, there is conflicting evidence. Conflict resolution is possible by postulating a role switch. In these cases, high biomarker values are, for example, antilongevity in early life and pro-longevity in later life. Role-switching biomarkers correspond to features that must, for example, be minimized early, but maximized later, for optimal longevity. The clear-cut pro-longevity biomarkers we found reflect anti-inflammatory, anti-immunosenescent or anti-anaemic mechanisms, whereas clear-cut antilongevity biomarkers reflect inflammatory mechanisms. Many highly significant blood biomarkers relate to immune system features, indicating a shift from adaptive to innate processes, whereas most role-switching biomarkers relate to blood serum features and whole-body phenotypes. Our biomarker classification approach is applicable to any combination of longitudinal studies with life expectancy data, and it provides insights beyond a simplified scheme of biomarkers for long or short lifespan.
Collapse
Affiliation(s)
- Mark Moeller
- Institute for Biostatistics and Informatics in Medicine und Ageing Research Rostock University Medical Center Rostock Germany
| | - Misa Hirose
- Department of Dermatology University of Lübeck Lübeck Germany
| | - Sarah Mueller
- Division of Gastroenterology Department of Medicine II Rostock University Medical Center Rostock Germany
| | - Catrin Roolf
- Department of Hematology/Oncology/Palliative Medicine Rostock University Medical Center Rostock Germany
| | - Simone Baltrusch
- Institute of Medical Biochemistry and Molecular Biology Rostock University Medical Center Rostock Germany
| | - Saleh Ibrahim
- Department of Dermatology University of Lübeck Lübeck Germany
| | - Christian Junghanss
- Department of Hematology/Oncology/Palliative Medicine Rostock University Medical Center Rostock Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics University of Rostock Rostock Germany
| | - Robert Jaster
- Division of Gastroenterology Department of Medicine II Rostock University Medical Center Rostock Germany
| | - Rüdiger Köhling
- Institute of Physiology Rostock University Medical Center Rostock Germany
| | - Manfred Kunz
- Department of Dermatology, Venereology and Allergology University of Leipzig Leipzig Germany
| | - Markus Tiedge
- Institute of Medical Biochemistry and Molecular Biology Rostock University Medical Center Rostock Germany
| | - Paul N. Schofield
- Department of Physiology, Development and Neuroscience University of Cambridge Cambridge UK
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine und Ageing Research Rostock University Medical Center Rostock Germany
| |
Collapse
|
46
|
Age modulates Fe3O4 nanoparticles liver toxicity: dose-dependent decrease in mitochondrial respiratory chain complexes activities and coupling in middle-aged as compared to young rats. BIOMED RESEARCH INTERNATIONAL 2014; 2014:474081. [PMID: 24949453 PMCID: PMC4032731 DOI: 10.1155/2014/474081] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/03/2014] [Accepted: 04/04/2014] [Indexed: 12/17/2022]
Abstract
We examined the effects of iron oxide nanoparticles (IONPs) on mitochondrial respiratory chain complexes activities and mitochondrial coupling in young (3 months) and middle-aged (18 months) rat liver, organ largely involved in body iron detoxification. Isolated liver mitochondria were extracted using differential centrifugations. Maximal oxidative capacities (Vmax, complexes I, III, and IV activities), Vsucc (complexes II, III, and IV activities), and Vtmpd, (complex IV activity), together with mitochondrial coupling (Vmax/V0) were determined in controls conditions and after exposure to 250, 300, and 350 μg/ml Fe3O4 in young and middle-aged rats. In young liver mitochondria, exposure to IONPs did not alter mitochondrial function. In contrast, IONPs dose-dependently impaired all complexes of the mitochondrial respiratory chain in middle-aged rat liver: Vmax (from 30 ± 1.6 to 17.9 ± 1.5; P < 0.001), Vsucc (from 33.9 ± 1.7 to 24.3 ± 1.0; P < 0.01), Vtmpd (from 43.0 ± 1.6 to 26.3 ± 2.2 µmol O2/min/g protein; P < 0.001) using Fe3O4 350 µg/ml. Mitochondrial coupling also decreased. Interestingly, 350 μg/ml Fe3O4 in the form of Fe3+ solution did not impair liver mitochondrial function in middle-aged rats. Thus, IONPs showed a specific toxicity in middle-aged rats suggesting caution when using it in old age.
Collapse
|
47
|
Isaya G. Mitochondrial iron-sulfur cluster dysfunction in neurodegenerative disease. Front Pharmacol 2014; 5:29. [PMID: 24624085 PMCID: PMC3939683 DOI: 10.3389/fphar.2014.00029] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/15/2014] [Indexed: 12/21/2022] Open
Abstract
Growing evidence supports a role for mitochondrial iron metabolism in the pathophysiology of neurodegenerative disorders such as Friedreich ataxia (FRDA) and Parkinson disease (PD) as well as in the motor and cognitive decline associated with the aging process. Iron-sulfur enzyme deficits and regional iron accumulation have been observed in each of these conditions. In spite of significant etiological, clinical and pathological differences that exist between FRDA and PD, it is possible that defects in mitochondrial iron-sulfur clusters (ISCs) biogenesis represent a common underlying mechanism leading to abnormal intracellular iron distribution with mitochondrial iron accumulation, oxidative phosphorylation deficits and oxidative stress in susceptible cells and specific regions of the nervous system. Moreover, a similar mechanism may contribute to the age-dependent iron accumulation that occurs in certain brain regions such as the globus pallidus and the substantia nigra. Targeting chelatable iron and reactive oxygen species appear as possible therapeutic options for FRDA and PD, and possibly other age-related neurodegenerative conditions. However, new technology to interrogate ISC synthesis in humans is needed to (i) assess how defects in this pathway contribute to the natural history of neurodegenerative disorders and (ii) develop treatments to correct those defects early in the disease process, before they cause irreversible neuronal cell damage.
Collapse
Affiliation(s)
- Grazia Isaya
- Department of Pediatric & Adolescent Medicine and Mayo Clinic Children's Center Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
48
|
Mallikarjun V, Sriram A, Scialo F, Sanz A. The interplay between mitochondrial protein and iron homeostasis and its possible role in ageing. Exp Gerontol 2014; 56:123-34. [PMID: 24394155 DOI: 10.1016/j.exger.2013.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/23/2013] [Accepted: 12/27/2013] [Indexed: 01/18/2023]
Abstract
Free (labile or chelatable) iron is extremely redox-active and only represents a small fraction of the total mitochondrial iron population. Several studies have shown that the proportion of free iron increases with age, leading to increased Fenton chemistry in later life. It is not clear why free iron accumulates in mitochondria, but it does so in parallel with an inability to degrade and recycle damaged proteins that causes loss of mitochondrial protein homeostasis (proteostasis). The increase in oxidative damage that has been shown to occur with age might be explained by these two processes. While this accumulation of oxidative damage has often been cited as causative to ageing there are examples of model organisms that possess high levels of oxidative damage throughout their lives with no effect on lifespan. Interestingly, these same animals are characterised by an outstanding ability to maintain correct proteostasis during their entire life. ROS can damage critical components of the iron homeostasis machinery, while the efficacy of mitochondrial quality control mechanisms will determine how detrimental that damage is. Here we review the interplay between iron and organellar quality control in mitochondrial dysfunction and we suggest that a decline in mitochondrial proteostasis with age leaves iron homeostasis (where several key stages are thought to be dependent on proteostasis machinery) vulnerable to oxidative damage and other age-related stress factors. This will have severe consequences for the electron transport chain and TCA cycle (among other processes) where several components are acutely dependent on correct assembly, insertion and maintenance of iron-sulphur clusters, leading to energetic crisis and death.
Collapse
Affiliation(s)
- Venkatesh Mallikarjun
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland.
| | - Ashwin Sriram
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland
| | - Filippo Scialo
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland
| | - Alberto Sanz
- Institute of Biomedical Technology and BioMediTech, University of Tampere, FI-33014 Tampere, Finland.
| |
Collapse
|
49
|
Lin SC, Karoly ED, Taatjes DJ. The human ΔNp53 isoform triggers metabolic and gene expression changes that activate mTOR and alter mitochondrial function. Aging Cell 2013; 12:863-72. [PMID: 23734707 DOI: 10.1111/acel.12108] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2013] [Indexed: 12/20/2022] Open
Abstract
A naturally occurring p53 isoform that lacks 39 residues at the N-terminus (denoted ΔNp53), when expressed with wild-type p53 (WTp53), forms mixed ΔNp53:WTp53 tetramers and causes accelerated aging in mice. Cellular alterations specific to ΔNp53:WTp53 have been difficult to assess because ΔNp53 and WTp53 coexpression results in tetramer heterogeneity, including formation of contaminating WTp53 tetramers. Based on the p53 tetramer structure, we expressed ΔNp53 and WTp53 as a single transcript that maintained tetramer architecture, ensuring a 2:2 ΔNp53:WTp53 stoichiometry. As expected, ΔNp53:WTp53 tetramers were stable and transcriptionally active in vitro and in cells, largely mimicking the function of WTp53 tetramers. Microarray analyses, however, revealed about 80 genes whose expression was altered twofold or more in ΔNp53:WTp53 cells. Moreover, global metabolomic profiling quantitated hundreds of biochemicals across different experiments (WTp53, ΔNp53:WTp53, plus controls). When evaluated collectively, these data suggested altered mTOR signaling and mitochondrial function-each canonical regulators of longevity-in cells expressing ΔNp53:WTp53 vs. WTp53. Increased levels of free amino acids, increased expression of IRS-1, and decreased expression of INPP5D/SHIP-1 suggested activated mTOR signaling in ΔNp53:WTp53 cells; this was confirmed upon comparative analyses of several mTOR pathway intermediates. We also observed changes in mitochondrial function in ΔNp53:WTp53 cells, which correlated with increased MARS2 expression and increased levels of carnitine, acetyl CoA, ATP, and Krebs cycle intermediates. Finally, increased levels of succinate and 2-hydroxyglutarate indicate potential epigenetic means to propagate ΔNp53:WTp53-induced gene expression changes to cell progeny. This may be especially important for aging, as biological effects manifest over time.
Collapse
Affiliation(s)
- Shih-Chieh Lin
- Department of Chemistry and Biochemistry; University of Colorado; Boulder; CO 80303; USA
| | | | - Dylan J. Taatjes
- Department of Chemistry and Biochemistry; University of Colorado; Boulder; CO 80303; USA
| |
Collapse
|
50
|
Calvani R, Joseph AM, Adhihetty PJ, Miccheli A, Bossola M, Leeuwenburgh C, Bernabei R, Marzetti E. Mitochondrial pathways in sarcopenia of aging and disuse muscle atrophy. Biol Chem 2013; 394:393-414. [PMID: 23154422 DOI: 10.1515/hsz-2012-0247] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 11/13/2012] [Indexed: 12/18/2022]
Abstract
Muscle loss during aging and disuse is a highly prevalent and disabling condition, but knowledge about cellular pathways mediating muscle atrophy is still limited. Given the postmitotic nature of skeletal myocytes, the maintenance of cellular homeostasis relies on the efficiency of cellular quality control mechanisms. In this scenario, alterations in mitochondrial function are considered a major factor underlying sarcopenia and muscle atrophy. Damaged mitochondria are not only less bioenergetically efficient, but also generate increased amounts of reactive oxygen species, interfere with cellular quality control mechanisms, and display a greater propensity to trigger apoptosis. Thus, mitochondria stand at the crossroad of signaling pathways that regulate skeletal myocyte function and viability. Studies on these pathways have sometimes provided unexpected and counterintuitive results, which suggests that they are organized into a complex, heterarchical network that is currently insufficiently understood. Untangling the complexity of such a network will likely provide clinicians with novel and highly effective therapeutics to counter the muscle loss associated with aging and disuse. In this review, we summarize the current knowledge on the mechanisms whereby mitochondrial dysfunction intervenes in the pathogenesis of sarcopenia and disuse atrophy, and highlight the prospect of targeting specific processes to treat these conditions.
Collapse
Affiliation(s)
- Riccardo Calvani
- Institute of Crystallography, Italian National Research Council (CNR), Bari 70126, Italy
| | | | | | | | | | | | | | | |
Collapse
|