1
|
Lan B, Liu C, Wang S, Jin Y, Yadav AK, Srivastava P, Yuan S, Hu C, Zhu G. Enhanced electron transfer for the improvement of nitrogen removal efficiency and N 2O reduction at low temperatures. WATER RESEARCH 2024; 272:122993. [PMID: 39708380 DOI: 10.1016/j.watres.2024.122993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Low temperature generally restricts biological activity, slowing down electron transfer in biogeochemical cycles and causing a series of environmental problems such as nitrogen pollution. We present a strategy to boost electron transfer in microbial cell at low temperatures via stimulation with low current. It is demonstrated by establishing a constructed wetland system coupled with solar powered microbial electrolysis cell, which enhances microbial activity through external micro currents (18.9 ± 5.5 μA) for removing nitrogen pollution in winter (average temperature from -6.6 to 4.5 °C). We investigated the efficiency of pollutants removal, microbial activity, N2O production and its mechanisms using complexes activity detection, RT-qPCR, incubation, and 15N-18O dual-isotope labeling techniques. The activity of complexes I, II, III, and IV collectively represent the microbial electron transfer rate. Results indicated that the microcurrents increased the activity of complexes II, III and IV by 96 %, 172 %, and 313 %, respectively. The transcription abundance of amoA genes in ammonia oxidation and nirS/K genes in denitrification by 263 % and 51 %, respectively. Consequently, NH4+-N removal efficiency improved from 23 % to 35 %, and NO3--N removal efficiency from 21 % to 31 %. Moreover, microcurrents reduced N2O emission by 44 %. However, external microcurrent stimulation did not alter the microbial production pathway of N2O as determined by the 15N-18O dual isotope labeling technique. The relative abundance of the nitrifying bacteria Nitrosomonas, Nitrosospira, and Nitrospira, as well as the denitrifying bacteria Methylotenera, significantly increased due to microcurrent stimulation. Specifically, Nitrospira exhibited the highest increase of 156 %. Our findings provide a novel way to enhance N removal efficiency and simultaneously reduce N2O emission of biological system like constructed wetlands in winter conditions.
Collapse
Affiliation(s)
- Bangrui Lan
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Chunlei Liu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shanyun Wang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yucheng Jin
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; School of Municipal and Environmental Engineering, Jilin Jianzhu University, Changchun, 130118, PR China
| | - Asheesh Kumar Yadav
- CSIR-Institute of Minerals and Materials Technology, Bhubaneswar, 751013, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pratiksha Srivastava
- Department of Chemical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, 3010, VIC, Australia
| | - Shengguang Yuan
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Chengzhi Hu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guibing Zhu
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
2
|
Hu Z, Yang L, Zhang M, Tang H, Huang Y, Su Y, Ding Y, Li C, Wang M, Zhou Y, Zhang Q, Guo L, Wu Y, Wang Q, Liu N, Kang H, Wu Y, Yao D, Li Y, Ruan Z, Wang H, Bao F, Liu G, Wang J, Wang Y, Wang W, Lu G, Qin D, Pei D, Chan WY, Liu X. A novel protein CYTB-187AA encoded by the mitochondrial gene CYTB modulates mammalian early development. Cell Metab 2024; 36:1586-1597.e7. [PMID: 38703762 DOI: 10.1016/j.cmet.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/10/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The mitochondrial genome transcribes 13 mRNAs coding for well-known proteins essential for oxidative phosphorylation. We demonstrate here that cytochrome b (CYTB), the only mitochondrial-DNA-encoded transcript among complex III, also encodes an unrecognized 187-amino-acid-long protein, CYTB-187AA, using the standard genetic code of cytosolic ribosomes rather than the mitochondrial genetic code. After validating the existence of this mtDNA-encoded protein arising from cytosolic translation (mPACT) using mass spectrometry and antibodies, we show that CYTB-187AA is mainly localized in the mitochondrial matrix and promotes the pluripotent state in primed-to-naive transition by interacting with solute carrier family 25 member 3 (SLC25A3) to modulate ATP production. We further generated a transgenic knockin mouse model of CYTB-187AA silencing and found that reduction of CYTB-187AA impairs females' fertility by decreasing the number of ovarian follicles. For the first time, we uncovered the novel mPACT pattern of a mitochondrial mRNA and demonstrated the physiological function of this 14th protein encoded by mtDNA.
Collapse
Affiliation(s)
- Zhijuan Hu
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Liang Yang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Maolei Zhang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haite Tang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yile Huang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China
| | - Yujie Su
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yingzhe Ding
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China
| | - Chong Li
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Mengfei Wang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yunhao Zhou
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Qing Zhang
- Proteomics and Metabolomics Core Facility, Guangzhou National Laboratory, Guangzhou, China
| | - Liman Guo
- Proteomics and Metabolomics Core Facility, Guangzhou National Laboratory, Guangzhou, China
| | - Yue Wu
- Proteomics and Metabolomics Core Facility, Guangzhou National Laboratory, Guangzhou, China
| | - Qianqian Wang
- State Key Laboratory of Medicinal Chemistry Biology, Nankai University, Tianjin, China
| | - Ning Liu
- State Key Laboratory of Medicinal Chemistry Biology, Nankai University, Tianjin, China
| | - Haoran Kang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yi Wu
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Deyang Yao
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yukun Li
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zifeng Ruan
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China; University of Chinese Academy of Sciences, Beijing, China
| | - Hao Wang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Feixiang Bao
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Guopan Liu
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China
| | - Junwei Wang
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yaofeng Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China
| | - Wuming Wang
- CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, CUHK-Jinan University Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Gang Lu
- CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, CUHK-Jinan University Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Duanqing Pei
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China; Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Wai-Yee Chan
- CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, CUHK-Jinan University Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Xingguo Liu
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, SAR, China; Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
3
|
Kaw A, Wu T, Starosolski Z, Zhou Z, Pedroza AJ, Majumder S, Duan X, Kaw K, Pinelo JEE, Fischbein MP, Lorenzi PL, Tan L, Martinez SA, Mahmud I, Devkota L, Taegtmeyer H, Ghaghada KB, Marrelli SP, Kwartler CS, Milewicz DM. Augmenting Mitochondrial Respiration in Immature Smooth Muscle Cells with an ACTA2 Pathogenic Variant Mitigates Moyamoya-like Cerebrovascular Disease. RESEARCH SQUARE 2023:rs.3.rs-3304679. [PMID: 37886459 PMCID: PMC10602100 DOI: 10.21203/rs.3.rs-3304679/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
ACTA2 pathogenic variants altering arginine 179 cause childhood-onset strokes due to moyamoya disease (MMD)-like occlusion of the distal internal carotid arteries. A smooth muscle cell (SMC)-specific knock-in mouse model (Acta2SMC-R179C/+) inserted the mutation into 67% of aortic SMCs, whereas explanted SMCs were uniformly heterozygous. Acta2R179C/+ SMCs fail to fully differentiate and maintain stem cell-like features, including high glycolytic flux, and increasing oxidative respiration (OXPHOS) with nicotinamide riboside (NR) drives the mutant SMCs to differentiate and decreases migration. Acta2SMC-R179C/+ mice have intraluminal MMD-like occlusive lesions and strokes after carotid artery injury, whereas the similarly treated WT mice have no strokes and patent lumens. Treatment with NR prior to the carotid artery injury attenuates the strokes, MMD-like lumen occlusions, and aberrant vascular remodeling in the Acta2SMC-R179C/+ mice. These data highlight the role of immature SMCs in MMD-associated occlusive disease and demonstrate that altering SMC metabolism to drive quiescence of Acta2R179C/+ SMCs attenuates strokes and aberrant vascular remodeling in the Acta2SMC-R179C/+ mice.
Collapse
Affiliation(s)
- Anita Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ting Wu
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Zbigniew Starosolski
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suravi Majumder
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Xueyan Duan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Kaveeta Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Jose E. E. Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sara A. Martinez
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Iqbal Mahmud
- Metabolomics Core Facility, Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laxman Devkota
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Heinrich Taegtmeyer
- Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ketan B. Ghaghada
- Department of Radiology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Sean P. Marrelli
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, McGovern Medical School, 6431 Fannin Street, Houston, TX 77030, USA
| | - Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| |
Collapse
|
4
|
Amato I, Meurant S, Renard P. The Key Role of Mitochondria in Somatic Stem Cell Differentiation: From Mitochondrial Asymmetric Apportioning to Cell Fate. Int J Mol Sci 2023; 24:12181. [PMID: 37569553 PMCID: PMC10418455 DOI: 10.3390/ijms241512181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
The study of the mechanisms underlying stem cell differentiation is under intensive research and includes the contribution of a metabolic switch from glycolytic to oxidative metabolism. While mitochondrial biogenesis has been previously demonstrated in number of differentiation models, it is only recently that the role of mitochondrial dynamics has started to be explored. The discovery of asymmetric distribution of mitochondria in stem cell progeny has strengthened the interest in the field. This review attempts to summarize the regulation of mitochondrial asymmetric apportioning by the mitochondrial fusion, fission, and mitophagy processes as well as emphasize how asymmetric mitochondrial apportioning in stem cells affects their metabolism, and thus epigenetics, and determines cell fate.
Collapse
Affiliation(s)
- Ilario Amato
- Ressearch Unit in Cell Biology (URBC), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium; (I.A.); (S.M.)
| | - Sébastien Meurant
- Ressearch Unit in Cell Biology (URBC), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium; (I.A.); (S.M.)
| | - Patricia Renard
- Ressearch Unit in Cell Biology (URBC), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium; (I.A.); (S.M.)
- Mass Spectrometry Platform (MaSUN), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium
| |
Collapse
|
5
|
Anitha A, Thanseem I, Iype M, Thomas SV. Mitochondrial dysfunction in cognitive neurodevelopmental disorders: Cause or effect? Mitochondrion 2023; 69:18-32. [PMID: 36621534 DOI: 10.1016/j.mito.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Mitochondria have a crucial role in brain development and neurogenesis, both in embryonic and adult brains. Since the brain is the highest energy consuming organ, it is highly vulnerable to mitochondrial dysfunction. This has been implicated in a range of brain disorders including, neurodevelopmental conditions, psychiatric illnesses, and neurodegenerative diseases. Genetic variations in mitochondrial DNA (mtDNA), and nuclear DNA encoding mitochondrial proteins, have been associated with several cognitive disorders. However, it is not yet clear whether mitochondrial dysfunction is a primary cause of these conditions or a secondary effect. Our review article deals with this topic, and brings out recent advances in mitochondria-oriented therapies. Mitochondrial dysfunction could be involved in the pathogenesis of a subset of disorders involving cognitive impairment. In these patients, mitochondrial dysfunction could be the cause of the condition, rather than the consequence. There are vast areas in this topic that remains to be explored and elucidated.
Collapse
Affiliation(s)
- Ayyappan Anitha
- Dept. of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India.
| | - Ismail Thanseem
- Dept. of Neurogenetics, Institute for Communicative and Cognitive Neurosciences (ICCONS), Shoranur, Palakkad 679 523, Kerala, India
| | - Mary Iype
- Dept. of Pediatric Neurology, Government Medical College, Thiruvananthapuram 695 011, Kerala, India; Dept. of Neurology, ICCONS, Thiruvananthapuram 695 033, Kerala, India
| | - Sanjeev V Thomas
- Dept. of Neurology, ICCONS, Thiruvananthapuram 695 033, Kerala, India
| |
Collapse
|
6
|
Correia B, Sousa MI, Branco AF, Rodrigues AS, Ramalho-Santos J. Leucine and Arginine Availability Modulate Mouse Embryonic Stem Cell Proliferation and Metabolism. Int J Mol Sci 2022; 23:ijms232214286. [PMID: 36430764 PMCID: PMC9694364 DOI: 10.3390/ijms232214286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
Amino acids are crucial nutrients involved in several cellular and physiological processes, including fertilization and early embryo development. In particular, Leucine and Arginine have been shown to stimulate implantation, as lack of both in a blastocyst culture system is able to induce a dormant state in embryos. The aim of this work was to evaluate the effects of Leucine and Arginine withdrawal on pluripotent mouse embryonic stem cell status, notably, their growth, self-renewal, as well as glycolytic and oxidative metabolism. Our results show that the absence of both Leucine and Arginine does not affect mouse embryonic stem cell pluripotency, while reducing cell proliferation through cell-cycle arrest. Importantly, these effects are not related to Leukemia Inhibitory Factor (LIF) and are reversible when both amino acids are reconstituted in the culture media. Moreover, a lack of these amino acids is related to a reduction in glycolytic and oxidative metabolism and decreased protein translation in mouse embryonic stem cells (mESCs), while maintaining their pluripotent status.
Collapse
Affiliation(s)
- Bibiana Correia
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Azinhaga de Santa Comba, Polo 3, 3000-354 Coimbra, Portugal
| | - Maria Inês Sousa
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Azinhaga de Santa Comba, Polo 3, 3000-354 Coimbra, Portugal
| | - Ana Filipa Branco
- CNC—Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Azinhaga de Santa Comba, Polo 3, 3000-354 Coimbra, Portugal
| | - Ana Sofia Rodrigues
- CNC—Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Azinhaga de Santa Comba, Polo 3, 3000-354 Coimbra, Portugal
| | - João Ramalho-Santos
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
- CNC—Center for Neuroscience and Cell Biology, CIBB, University of Coimbra, Azinhaga de Santa Comba, Polo 3, 3000-354 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
7
|
Tournaire G, Loopmans S, Stegen S, Rinaldi G, Eelen G, Torrekens S, Moermans K, Carmeliet P, Ghesquière B, Thienpont B, Fendt SM, van Gastel N, Carmeliet G. Skeletal progenitors preserve proliferation and self-renewal upon inhibition of mitochondrial respiration by rerouting the TCA cycle. Cell Rep 2022; 40:111105. [PMID: 35905715 PMCID: PMC9380255 DOI: 10.1016/j.celrep.2022.111105] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 04/13/2022] [Accepted: 06/25/2022] [Indexed: 11/25/2022] Open
Abstract
A functional electron transport chain (ETC) is crucial for supporting bioenergetics and biosynthesis. Accordingly, ETC inhibition decreases proliferation in cancer cells but does not seem to impair stem cell proliferation. However, it remains unclear how stem cells metabolically adapt. In this study, we show that pharmacological inhibition of complex III of the ETC in skeletal stem and progenitor cells induces glycolysis side pathways and reroutes the tricarboxylic acid (TCA) cycle to regenerate NAD+ and preserve cell proliferation. These metabolic changes also culminate in increased succinate and 2-hydroxyglutarate levels that inhibit Ten-eleven translocation (TET) DNA demethylase activity, thereby preserving self-renewal and multilineage potential. Mechanistically, mitochondrial malate dehydrogenase and reverse succinate dehydrogenase activity proved to be essential for the metabolic rewiring in response to ETC inhibition. Together, these data show that the metabolic plasticity of skeletal stem and progenitor cells allows them to bypass ETC blockade and preserve their self-renewal. Skeletal stem/progenitor cells can proliferate upon electron transport chain blockade Succinate dehydrogenase is reversed with fumarate functioning as electron acceptor Pyruvate and aspartate are critical for NAD+ regeneration and proliferation Metabolic changes prevent DNA demethylation and preserve self-renewal
Collapse
Affiliation(s)
- Guillaume Tournaire
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, O&N1bis Herestraat 49, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Shauni Loopmans
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, O&N1bis Herestraat 49, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, O&N1bis Herestraat 49, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Gianmarco Rinaldi
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology and Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sophie Torrekens
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, O&N1bis Herestraat 49, 3000 Leuven, Belgium
| | - Karen Moermans
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, O&N1bis Herestraat 49, 3000 Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Expertise Center, Department of Oncology, KU Leuven/VIB Center for Cancer Biology Leuven, Leuven, Belgium
| | - Bernard Thienpont
- Laboratory of Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB Center for Cancer Biology, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology and Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | | | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, O&N1bis Herestraat 49, 3000 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.
| |
Collapse
|
8
|
Gyllenhammer LE, Rasmussen JM, Bertele N, Halbing A, Entringer S, Wadhwa PD, Buss C. Maternal Inflammation During Pregnancy and Offspring Brain Development: The Role of Mitochondria. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2022; 7:498-509. [PMID: 34800727 PMCID: PMC9086015 DOI: 10.1016/j.bpsc.2021.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/20/2021] [Accepted: 11/04/2021] [Indexed: 01/06/2023]
Abstract
The association between maternal immune activation (MIA) during pregnancy and risk for offspring neuropsychiatric disorders has been increasingly recognized over the past several years. Among the mechanistic pathways that have been described through which maternal inflammation during pregnancy may affect fetal brain development, the role of mitochondria has received little attention. In this review, the role of mitochondria as a potential mediator of the association between MIA during pregnancy and offspring brain development and risk for psychiatric disorders will be proposed. As a basis for this postulation, convergent evidence is presented supporting the obligatory role of mitochondria in brain development, the role of mitochondria as mediators and initiators of inflammatory processes, and evidence of mitochondrial dysfunction in preclinical MIA exposure models and human neurodevelopmental disorders. Elucidating the role of mitochondria as a potential mediator of MIA-induced alterations in brain development and neurodevelopmental disease risk may not only provide new insight into the pathophysiology of mental health disorders that have their origins in exposure to infection/immune activation during pregnancy but also offer new therapeutic targets.
Collapse
Affiliation(s)
- Lauren E Gyllenhammer
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California
| | - Jerod M Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California
| | - Nina Bertele
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Amy Halbing
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Einstein Center for Neurosciences Berlin, Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California; Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California; Department of Psychiatry and Human Behavior, University of California, Irvine, School of Medicine, Irvine, California; Department of Obstetrics and Gynecology, University of California, Irvine, School of Medicine, Irvine, California; Department of Epidemiology, University of California, Irvine, School of Medicine, Irvine, California
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, California; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, California; Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
9
|
Romero-Morales AI, Gama V. Revealing the Impact of Mitochondrial Fitness During Early Neural Development Using Human Brain Organoids. Front Mol Neurosci 2022; 15:840265. [PMID: 35571368 PMCID: PMC9102998 DOI: 10.3389/fnmol.2022.840265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial homeostasis -including function, morphology, and inter-organelle communication- provides guidance to the intrinsic developmental programs of corticogenesis, while also being responsive to environmental and intercellular signals. Two- and three-dimensional platforms have become useful tools to interrogate the capacity of cells to generate neuronal and glia progeny in a background of metabolic dysregulation, but the mechanistic underpinnings underlying the role of mitochondria during human neurogenesis remain unexplored. Here we provide a concise overview of cortical development and the use of pluripotent stem cell models that have contributed to our understanding of mitochondrial and metabolic regulation of early human brain development. We finally discuss the effects of mitochondrial fitness dysregulation seen under stress conditions such as metabolic dysregulation, absence of developmental apoptosis, and hypoxia; and the avenues of research that can be explored with the use of brain organoids.
Collapse
Affiliation(s)
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
10
|
Zhang S, Zhao J, Quan Z, Li H, Qing H. Mitochondria and Other Organelles in Neural Development and Their Potential as Therapeutic Targets in Neurodegenerative Diseases. Front Neurosci 2022; 16:853911. [PMID: 35450015 PMCID: PMC9016280 DOI: 10.3389/fnins.2022.853911] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/07/2022] [Indexed: 12/19/2022] Open
Abstract
The contribution of organelles to neural development has received increasing attention. Studies have shown that organelles such as mitochondria, endoplasmic reticulum (ER), lysosomes, and endosomes play important roles in neurogenesis. Specifically, metabolic switching, reactive oxygen species production, mitochondrial dynamics, mitophagy, mitochondria-mediated apoptosis, and the interaction between mitochondria and the ER all have roles in neurogenesis. Lysosomes and endosomes can regulate neurite growth and extension. Moreover, metabolic reprogramming represents a novel strategy for generating functional neurons. Accordingly, the exploration and application of mechanisms underlying metabolic reprogramming will be beneficial for neural conversion and regenerative medicine. There is adequate evidence implicating the dysfunction of cellular organelles—especially mitochondria—in neurodegenerative disorders, and that improvement of mitochondrial function may reverse the progression of these diseases through the reinforcement of adult neurogenesis. Therefore, these organelles have potential as therapeutic targets for the treatment of neurodegenerative diseases. In this review, we discuss the function of these organelles, especially mitochondria, in neural development, focusing on their potential as therapeutic targets in neurodegenerative disorders, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Shuyuan Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Juan Zhao
- Aerospace Medical Center, Aerospace Center Hospital, Beijing, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
- *Correspondence: Hui Li,
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
- Hong Qing,
| |
Collapse
|
11
|
Detection of Pathological Markers of Neurodegenerative Diseases following Microfluidic Direct Conversion of Patient Fibroblasts into Neurons. Int J Mol Sci 2022; 23:ijms23042147. [PMID: 35216271 PMCID: PMC8879457 DOI: 10.3390/ijms23042147] [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: 12/28/2021] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/28/2022] Open
Abstract
Neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease are clinically diagnosed using neuropsychological and cognitive tests, expensive neuroimaging-based approaches (MRI and PET) and invasive and time-consuming lumbar puncture for cerebrospinal fluid (CSF) sample collection to detect biomarkers. Thus, a rapid, simple and cost-effective approach to more easily access fluids and tissues is in great need. Here, we exploit the chemical direct reprogramming of patient skin fibroblasts into neurons (chemically induced neurons, ciNs) as a novel strategy for the rapid detection of different pathological markers of neurodegenerative diseases. We found that FAD fibroblasts have a reduced efficiency of reprogramming, and converted ciNs show a less complex neuronal network. In addition, ciNs from patients show misfolded protein accumulation and mitochondria ultrastructural abnormalities, biomarkers commonly associated with neurodegeneration. Moreover, for the first time, we show that microfluidic technology, in combination with chemical reprogramming, enables on-chip examination of disease pathological processes and may have important applications in diagnosis. In conclusion, ciNs on microfluidic devices represent a small-scale, non-invasive and cost-effective high-throughput tool for protein misfolding disease diagnosis and may be useful for new biomarker discovery, disease mechanism studies and design of personalised therapies.
Collapse
|
12
|
Liu W, Chen G. Regulation of energy metabolism in human pluripotent stem cells. Cell Mol Life Sci 2021; 78:8097-8108. [PMID: 34773132 PMCID: PMC11071932 DOI: 10.1007/s00018-021-04016-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
All living organisms need energy to carry out their essential functions. The importance of energy metabolism is increasingly recognized in human pluripotent stem cells. Energy production is not only essential for cell survival and proliferation, but also critical for pluripotency and cell fate determination. Thus, energy metabolism is an important target in cellular regulation and stem cell applications. In this review, we will discuss key factors that influence energy metabolism and their association with stem cell functions.
Collapse
Affiliation(s)
- Weiwei Liu
- Faculty of Health Sciences, Centre of Reproduction, Development and Aging, University of Macau, Taipa, Macau SAR, China
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Guokai Chen
- Faculty of Health Sciences, Centre of Reproduction, Development and Aging, University of Macau, Taipa, Macau SAR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
13
|
Correia B, Sousa MI, Ramalho-Santos J. Glycolytic Profiling of Mouse Embryonic Stem Cells (mESCs). METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2520:151-159. [PMID: 34724190 DOI: 10.1007/7651_2021_449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mouse embryonic stem cells (mESCs) can be captured in vitro in different pluripotency states through media modulation, mimicking their natural environment during early embryo development. As highly proliferative cells, mESCs prefer to use glycolysis to support the energetic and biosynthetic demands, even in the presence of oxygen. Indeed, glycolysis can not only supply ATP at a much faster rate, when compared to other catabolic pathways, but also provides biosynthetic substrates to meet anabolic requirements. Considering that ESCs cultured in different media conditions display distinct metabolic requirements, it is of utmost importance to have a robust metabolic characterization methodology to understand how subtle metabolic variations may be coupled to ESC identity. Here we describe how to profile the glycolytic activity of naive mouse ESC, using the established Seahorse XFe24 Live-cell Metabolic Assay. This may be a useful protocol for understanding how the glycolytic function of mESCs changes in certain circumstances and how is it coupled to diverse pluripotency/differentiation phenotypes.
Collapse
Affiliation(s)
- Bibiana Correia
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3,University of Coimbra, Coimbra, Portugal
| | - Maria Inês Sousa
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3,University of Coimbra, Coimbra, Portugal
| | - João Ramalho-Santos
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, Coimbra, Portugal.
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3,University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
14
|
Brunetti D, Dykstra W, Le S, Zink A, Prigione A. Mitochondria in neurogenesis: Implications for mitochondrial diseases. Stem Cells 2021; 39:1289-1297. [PMID: 34089537 DOI: 10.1002/stem.3425] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/24/2021] [Indexed: 06/12/2023]
Abstract
Mitochondria are organelles with recognized key roles in cellular homeostasis, including bioenergetics, redox, calcium signaling, and cell death. Mitochondria are essential for neuronal function, given the high energy demands of the human brain. Consequently, mitochondrial diseases affecting oxidative phosphorylation (OXPHOS) commonly exhibit neurological impairment. Emerging evidence suggests that mitochondria are important not only for mature postmitotic neurons but also for the regulation of neural progenitor cells (NPCs) during the process of neurogenesis. These recent findings put mitochondria as central regulator of cell fate decisions during brain development. OXPHOS mutations may disrupt the function of NPCs and thereby impair the metabolic programming required for neural fate commitment. Promoting the mitochondrial function of NPCs could therefore represent a novel interventional approach against incurable mitochondrial diseases.
Collapse
Affiliation(s)
- Dario Brunetti
- Mitochondrial Medicine Laboratory, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy
| | - Werner Dykstra
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Stephanie Le
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Annika Zink
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Alessandro Prigione
- Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
15
|
High Glycolytic Activity Enhances Stem Cell Reprogramming of Fahd1-KO Mouse Embryonic Fibroblasts. Cells 2021; 10:cells10082040. [PMID: 34440809 PMCID: PMC8392800 DOI: 10.3390/cells10082040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play a key role in metabolic transitions involved in the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs), but the underlying molecular mechanisms remain largely unexplored. To obtain new insight into the mechanisms of cellular reprogramming, we studied the role of FAH domain-containing protein 1 (FAHD1) in the reprogramming of murine embryonic fibroblasts (MEFs) into iPSCs and their subsequent differentiation into neuronal cells. MEFs from wild type (WT) and Fahd1-knock-out (KO) mice were reprogrammed into iPSCs and characterized for alterations in metabolic parameters and the expression of marker genes indicating mitochondrial biogenesis. Fahd1-KO MEFs showed a higher reprogramming efficiency accompanied by a significant increase in glycolytic activity as compared to WT. We also observed a strong increase of mitochondrial DNA copy number and expression of biogenesis marker genes in Fahd1-KO iPSCs relative to WT. Neuronal differentiation of iPSCs was accompanied by increased expression of mitochondrial biogenesis genes in both WT and Fahd1-KO neurons with higher expression in Fahd1-KO neurons. Together these observations establish a role of FAHD1 as a potential negative regulator of reprogramming and add additional insight into mechanisms by which FAHD1 modulates mitochondrial functions.
Collapse
|
16
|
da Costa RT, dos Santos MB, Silva ICS, de Almeida RP, Teruel MS, Carrettiero DC, Ribeiro CAJ. Methylmalonic Acid Compromises Respiration and Reduces the Expression of Differentiation Markers of SH-SY5Y Human Neuroblastoma Cells. ACS Chem Neurosci 2021; 12:2608-2618. [PMID: 34191487 DOI: 10.1021/acschemneuro.1c00119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Methylmalonic acidemia is a rare metabolic disorder caused by the deficient activity of l-methylmalonyl-CoA mutase or its cofactor 5-deoxyadenosylcobalamin and is characterized by accumulation of methylmalonic acid (MMA) and alternative metabolites. The brain is one of the most affected tissues and neurologic symptoms, characterized by seizures, mental retardation, psychomotor abnormalities, and coma, commonly appear in newborns. The molecular mechanisms of neuropathogenesis in methylmalonic acidemia are still poorly understood, specifically regarding the impairments in neuronal development, maturation, and differentiation. In this study, we investigated the effects of MMA in both undifferentiated and differentiated phenotypes of SH-SY5Y human neuroblastoma cells. We observed an increase in glucose consumption and reduction in respiratory parameters of both undifferentiated and differentiated cells after exposition to MMA, suggesting that differentiated cells are slightly more prone to perturbations in respiratory parameters by MMA than undifferentiated cells. Next, we performed qPCR of mature neuronal-specific gene markers and measured mitochondrial functioning to evaluate the role of MMA during differentiation. Our results showed that MMA impairs the respiratory parameters only at the late stage of differentiation and downregulates the transcriptional gene profile of mature neuronal markers neuron-specific enolase (ENO2) and synaptophysin (SYP). Altogether, our findings point out important changes observed during neuronal maturation and energetic stress vulnerability that can play a role in the neurological clinical symptoms at the newborn period and reveal important molecular mechanisms that could help the screening of targets to new approaches in the therapies of this disease.
Collapse
Affiliation(s)
- Renata T. da Costa
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas (CCNH), São Bernardo do Campo, SP 09606-070, Brazil
| | - Marcella B. dos Santos
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas (CCNH), São Bernardo do Campo, SP 09606-070, Brazil
| | - Izabel C. S. Silva
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas (CCNH), São Bernardo do Campo, SP 09606-070, Brazil
| | - Raquel P. de Almeida
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas (CCNH), São Bernardo do Campo, SP 09606-070, Brazil
| | - Marcela S. Teruel
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas (CCNH), São Bernardo do Campo, SP 09606-070, Brazil
| | - Daniel C. Carrettiero
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas (CCNH), São Bernardo do Campo, SP 09606-070, Brazil
| | - César A. J. Ribeiro
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas (CCNH), São Bernardo do Campo, SP 09606-070, Brazil
| |
Collapse
|
17
|
Yuan F, Wang N, Chen Y, Huang X, Yang Z, Xu Y, You K, Zhang J, Wang G, Zhuang Y, Pan T, Xiong Y, Yu X, Yang F, Li Y. Calcitriol promotes the maturation of hepatocyte-like cells derived from human pluripotent stem cells. J Steroid Biochem Mol Biol 2021; 211:105881. [PMID: 33766737 DOI: 10.1016/j.jsbmb.2021.105881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/07/2021] [Accepted: 03/18/2021] [Indexed: 11/23/2022]
Abstract
Human hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) represent a promising cell source for the assessment of hepatotoxicity and pharmaceutical safety testing. However, the hepatic functionality of HLCs remains significantly inferior to primary human hepatocytes. The bioactive vitamin D (VD), calcitriol, promotes the differentiation of many types of cells, and its deficiency is correlated to the severity of liver diseases. Whether calcitriol contributes to the differentiation of HLCs needs to be explored. Here, we found that the supplementation of calcitriol improved the functionalities of hPSCs-derived HLCs in P450 activities, urea production, and albumin secretion. Moreover, calcitriol also enhanced mitochondrial respiratory function with increased protein expression levels of the subunit of respiratory enzyme complexes in HLCs. Further analyses showed that the mitochondrial biogenesis regulators and mitophagy were increased by calcitriol, thus improving the mitochondrial quality. These improvements in functionality and mitochondrial condition were dependent on vitamin D receptor (VDR) because the improvements were abolished under VDR-deficient conditions. Our finding provides a cost-effective chemical process for HLC maturation to meet the demand for basic research and potential clinic applications.
Collapse
Affiliation(s)
- Fang Yuan
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; School of Life Sciences, University of Science and Technology of China, 230027, Hefei, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Ning Wang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yan Chen
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Xinping Huang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Zhen Yang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yingying Xu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Kai You
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Jiaye Zhang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Guodong Wang
- The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Yuanqi Zhuang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Tingcai Pan
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yue Xiong
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Xiaorui Yu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; School of Life Sciences, University of Science and Technology of China, 230027, Hefei, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Fan Yang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yinxiong Li
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese, Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China.
| |
Collapse
|
18
|
Qiu YY, Zhang HS, Tang Y, Liu FY, Pang JQ, Zhang XY, Xiong H, Liang YS, Zhao HY, Chen SJ. Mitochondrial dysfunction resulting from the down-regulation of bone morphogenetic protein 5 may cause microtia. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:418. [PMID: 33842639 PMCID: PMC8033356 DOI: 10.21037/atm-21-831] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Bone morphogenetic protein 5 (BMP5) has been identified as one of the important risk factors for microtia; however, the link between them has yet to be clarified. In this study, we aimed to demonstrate the relationship of BMP5 with mitochondrial function and investigate the specific role of mitochondria in regulating microtia development. Methods BMP5 expression was measured in auricular cartilage tissues from patients with and without microtia. The effects of BMP5 knockdown on cellular function and mitochondrial function were also analyzed in vitro. Changes in genome-wide expression profiles were measured in BMP5-knockdown cells. Finally, the specific impact of BMP5 down-regulation on mitochondrial fat oxidation was analyzed in vitro. Results BMP5 expression was down-regulated in the auricular cartilage tissues of microtia patients. BMP5 down-regulation inhibited various cellular functions in vitro, including cell proliferation, mobility, and cytoactivity. The functional integrity of mitochondria was also damaged, accompanied by a decrease in mitochondrial membrane potential, reactive oxygen species (ROS) neutralization, and reduced adenosine triphosphate (ATP) production. Carnitine O-palmitoyltransferase 2 and diacylglycerol acyltransferase 2, two of the key regulators of mitochondrial lipid oxidation, were also found to be decreased by BMP5 down-regulation. Conclusions Down-regulation of BMP5 affects glycerolipid metabolism and fatty acid degradation, leading to mitochondrial dysfunction, reduced ATP production, and changes in cell function, and ultimately resulting in microtia. This research provides supporting evidence for an important role of BMP5 down-regulation in affecting mitochondrial metabolism in cells, and sheds new light on the mechanisms underlying the pathogenesis of microtia.
Collapse
Affiliation(s)
- Yin-Yi Qiu
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hua-Song Zhang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Otolaryngology, Longgang E.N.T Hospital & Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| | - Yuan Tang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fei-Yi Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen, University, Guangzhou, China
| | - Jia-Qi Pang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xue-Yuan Zhang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Xiong
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu-Shuang Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen, University, Guangzhou, China
| | - Hui-Ying Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen, University, Guangzhou, China
| | - Sui-Jun Chen
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
19
|
Correia B, Sousa MI, Branco AF, Ramalho-Santos J. Monitoring Mitochondrial Function in Mouse Embryonic Stem Cells (mESCs). Methods Mol Biol 2021; 2310:47-56. [PMID: 34095997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mouse embryonic stem cells (mESCs) can be grown in culture, recapitulating the different states of pluripotency of their in vivo counterparts, with notably different metabolic profiles. mESCs in a naïve pluripotent state present an ambivalent metabolism, using both glycolysis and oxidative phosphorylation as energy sources. Here, we describe a method to evaluate the oxidative function of naïve mESCs using the Seahorse Extracellular Flux Analyzer coupled to flow cytometry analysis of mitochondrial transmembrane potential using the TMRM fluorescence probe, thus assessing both oxygen consumption and mitochondrial membrane potential. This may be a useful protocol for understanding how mitochondrial oxidative function and potential of mESCs change in certain circumstances, and how is it related with various pluripotency/differentiation phenotypes.
Collapse
Affiliation(s)
- Bibiana Correia
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal
| | - Maria Inês Sousa
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal
| | - Ana F Branco
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal
| | - João Ramalho-Santos
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
20
|
Li D, Ding Z, Gui M, Hou Y, Xie K. Metabolic Enhancement of Glycolysis and Mitochondrial Respiration Are Essential for Neuronal Differentiation. Cell Reprogram 2020; 22:291-299. [DOI: 10.1089/cell.2020.0034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Ding Li
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, China
| | - Zhexu Ding
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, China
| | - Manjin Gui
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, China
| | - Yanmei Hou
- Ausnutria Hyproca Nutrition Co. Ltd., Changsha, China
| | - Kui Xie
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, China
- Ausnutria Hyproca Nutrition Co. Ltd., Changsha, China
- Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
21
|
Lu J, Li Y, Mollinari C, Garaci E, Merlo D, Pei G. Amyloid-β Oligomers-induced Mitochondrial DNA Repair Impairment Contributes to Altered Human Neural Stem Cell Differentiation. Curr Alzheimer Res 2020; 16:934-949. [PMID: 31642778 DOI: 10.2174/1567205016666191023104036] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/25/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Amyloid-β42 oligomers (Aβ42O), the proximate effectors of neurotoxicity observed in Alzheimer's disease (AD), can induce mitochondrial oxidative stress and impair mitochondrial function besides causing mitochondrial DNA (mtDNA) damage. Aβ42O also regulate the proliferative and differentiative properties of stem cells. OBJECTIVE We aimed to study whether Aβ42O-induced mtDNA damage is involved in the regulation of stem cell differentiation. METHOD Human iPSCs-derived neural stem cell (NSC) was applied to investigate the effect of Aβ42O on reactive oxygen species (ROS) production and DNA damage using mitoSOX staining and long-range PCR lesion assay, respectively. mtDNA repair activity was measured by non-homologous end joining (NHEJ) in vitro assay using mitochondria isolates and the expression and localization of NHEJ components were determined by Western blot and immunofluorescence assay. The expressions of Tuj-1 and GFAP, detected by immunofluorescence and qPCR, respectively, were examined as an index of neurons and astrocytes production. RESULTS We show that in NSC Aβ42O treatment induces ROS production and mtDNA damage and impairs DNA end joining activity. NHEJ components, such as Ku70/80, DNA-PKcs, and XRCC4, are localized in mitochondria and silencing of XRCC4 significantly exacerbates the effect of Aβ42O on mtDNA integrity. On the contrary, pre-treatment with Phytic Acid (IP6), which specifically stimulates DNA-PK-dependent end-joining, inhibits Aβ42O-induced mtDNA damage and neuronal differentiation alteration. CONCLUSION Aβ42O-induced mtDNA repair impairment may change cell fate thus shifting human NSC differentiation toward an astrocytic lineage. Repair stimulation counteracts Aβ42O neurotoxicity, suggesting mtDNA repair pathway as a potential target for the treatment of neurodegenerative disorders like AD.
Collapse
Affiliation(s)
- Jing Lu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yi Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
| | - Cristiana Mollinari
- Department of Neuroscience, Istituto Superiore di Sanita, Rome, Italy.,Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Enrico Garaci
- IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.,Telematic University San Raffaele, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Daniela Merlo
- Department of Neuroscience, Istituto Superiore di Sanita, Rome, Italy
| | - Gang Pei
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.,Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
22
|
Lord T, Nixon B. Metabolic Changes Accompanying Spermatogonial Stem Cell Differentiation. Dev Cell 2020; 52:399-411. [PMID: 32097651 DOI: 10.1016/j.devcel.2020.01.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/27/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Male fertility is driven by spermatogonial stem cells (SSCs) that self-renew while also giving rise to differentiating spermatogonia. Spermatogonial transitions are accompanied by a shift in gene expression, however, whether equivalent changes in metabolism occur remains unexplored. In this review, we mined recently published scRNA-seq databases from mouse and human testes to compare expression profiles of spermatogonial subsets, focusing on metabolism. Comparisons revealed a conserved upregulation of genes involved in mitochondrial function, biogenesis, and oxidative phosphorylation in differentiating spermatogonia, while gene expression in SSCs reflected a glycolytic cell. Here, we also discuss the relationship between metabolism and the external microenvironment within which spermatogonia reside.
Collapse
Affiliation(s)
- Tessa Lord
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, the University of Newcastle, Callaghan, Newcastle, NSW 2300, Australia; Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, Newcastle, NSW 2305, Australia.
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, the University of Newcastle, Callaghan, Newcastle, NSW 2300, Australia; Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, Newcastle, NSW 2305, Australia
| |
Collapse
|
23
|
Harvey AJ. Mitochondria in early development: linking the microenvironment, metabolism and the epigenome. Reproduction 2020; 157:R159-R179. [PMID: 30870807 DOI: 10.1530/rep-18-0431] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 02/04/2019] [Indexed: 12/24/2022]
Abstract
Mitochondria, originally of bacterial origin, are highly dynamic organelles that have evolved a symbiotic relationship within eukaryotic cells. Mitochondria undergo dynamic, stage-specific restructuring and redistribution during oocyte maturation and preimplantation embryo development, necessary to support key developmental events. Mitochondria also fulfil a wide range of functions beyond ATP synthesis, including the production of intracellular reactive oxygen species and calcium regulation, and are active participants in the regulation of signal transduction pathways. Communication between not only mitochondria and the nucleus, but also with other organelles, is emerging as a critical function which regulates preimplantation development. Significantly, perturbations and deficits in mitochondrial function manifest not only as reduced quality and/or poor oocyte and embryo development but contribute to post-implantation failure, long-term cell function and adult disease. A growing body of evidence indicates that altered availability of metabolic co-factors modulate the activity of epigenetic modifiers, such that oocyte and embryo mitochondrial activity and dynamics have the capacity to establish long-lasting alterations to the epigenetic landscape. It is proposed that preimplantation embryo development may represent a sensitive window during which epigenetic regulation by mitochondria is likely to have significant short- and long-term effects on embryo, and offspring, health. Hence, mitochondrial integrity, communication and metabolism are critical links between the environment, the epigenome and the regulation of embryo development.
Collapse
Affiliation(s)
- Alexandra J Harvey
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
24
|
Agriesti F, Tataranni T, Pacelli C, Scrima R, Laurenzana I, Ruggieri V, Cela O, Mazzoccoli C, Salerno M, Sessa F, Sani G, Pomara C, Capitanio N, Piccoli C. Nandrolone induces a stem cell-like phenotype in human hepatocarcinoma-derived cell line inhibiting mitochondrial respiratory activity. Sci Rep 2020; 10:2287. [PMID: 32041983 PMCID: PMC7010785 DOI: 10.1038/s41598-020-58871-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022] Open
Abstract
Nandrolone is a testosterone analogue with anabolic properties commonly abused worldwide, recently utilized also as therapeutic agent in chronic diseases, cancer included. Here we investigated the impact of nandrolone on the metabolic phenotype in HepG2 cell line. The results attained show that pharmacological dosage of nandrolone, slowing cell growth, repressed mitochondrial respiration, inhibited the respiratory chain complexes I and III and enhanced mitochondrial reactive oxygen species (ROS) production. Intriguingly, nandrolone caused a significant increase of stemness-markers in both 2D and 3D cultures, which resulted to be CxIII-ROS dependent. Notably, nandrolone negatively affected differentiation both in healthy hematopoietic and mesenchymal stem cells. Finally, nandrolone administration in mice confirmed the up-regulation of stemness-markers in liver, spleen and kidney. Our observations show, for the first time, that chronic administration of nandrolone, favoring maintenance of stem cells in different tissues would represent a precondition that, in addition to multiple hits, might enhance risk of carcinogenesis raising warnings about its abuse and therapeutic utilization.
Collapse
Affiliation(s)
- Francesca Agriesti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy
| | - Tiziana Tataranni
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto c/o OO.RR., 71100, Foggia, Italy
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto c/o OO.RR., 71100, Foggia, Italy
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy
| | - Vitalba Ruggieri
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy
| | - Olga Cela
- Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto c/o OO.RR., 71100, Foggia, Italy
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy
| | - Monica Salerno
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania - A.O.U. "Policlinico - V. Emanuele", via S. Sofia, 87 - Sector 10, Building B - 95123, Catania, Italy
| | - Francesco Sessa
- Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto c/o OO.RR., 71100, Foggia, Italy
| | - Gabriele Sani
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Roma, Italy.,Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Cristoforo Pomara
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania - A.O.U. "Policlinico - V. Emanuele", via S. Sofia, 87 - Sector 10, Building B - 95123, Catania, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto c/o OO.RR., 71100, Foggia, Italy
| | - Claudia Piccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy. .,Department of Clinical and Experimental Medicine, University of Foggia, via L. Pinto c/o OO.RR., 71100, Foggia, Italy.
| |
Collapse
|
25
|
Prieto J, Ponsoda X, Izpisua Belmonte JC, Torres J. Mitochondrial dynamics and metabolism in induced pluripotency. Exp Gerontol 2020; 133:110870. [PMID: 32045634 DOI: 10.1016/j.exger.2020.110870] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/20/2019] [Accepted: 02/05/2020] [Indexed: 12/15/2022]
Abstract
Somatic cells can be reprogrammed to pluripotency by either ectopic expression of defined factors or exposure to chemical cocktails. During reprogramming, somatic cells undergo dramatic changes in a wide range of cellular processes, such as metabolism, mitochondrial morphology and function, cell signaling pathways or immortalization. Regulation of these processes during cell reprograming lead to the acquisition of a pluripotent state, which enables indefinite propagation by symmetrical self-renewal without losing the ability of reprogrammed cells to differentiate into all cell types of the adult. In this review, recent data from different laboratories showing how these processes are controlled during the phenotypic transformation of a somatic cell into a pluripotent stem cell will be discussed.
Collapse
Affiliation(s)
- Javier Prieto
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Calle Dr. Moliner 50, 46100 Burjassot, Spain; Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Xavier Ponsoda
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Calle Dr. Moliner 50, 46100 Burjassot, Spain; Instituto de Investigación Sanitaria (INCLIVA), Avenida de Menéndez y Pelayo 4, 46010, Valencia, Spain
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Josema Torres
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Calle Dr. Moliner 50, 46100 Burjassot, Spain; Instituto de Investigación Sanitaria (INCLIVA), Avenida de Menéndez y Pelayo 4, 46010, Valencia, Spain.
| |
Collapse
|
26
|
Chiu SP, Lee YW, Wu LY, Tung TH, Gomez S, Lo CM, Wang JY. Application of ECIS to Assess FCCP-Induced Changes of MSC Micromotion and Wound Healing Migration. SENSORS 2019; 19:s19143210. [PMID: 31330904 PMCID: PMC6679573 DOI: 10.3390/s19143210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
Electric cell-substrate impedance sensing (ECIS) is an emerging technique for sensitively monitoring morphological changes of adherent cells in tissue culture. In this study, human mesenchymal stem cells (hMSCs) were exposed to different concentrations of carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP) for 20 h and their subsequent concentration-dependent responses in micromotion and wound healing migration were measured by ECIS. FCCP disrupts ATP synthesis and results in a decrease in cell migration rates. To detect the change of cell micromotion in response to FCCP challenge, time-series resistances of cell-covered electrodes were monitored and the values of variance were calculated to verify the difference. While Seahorse XF-24 extracellular flux analyzer can detect the effect of FCCP at 3 μM concentration, the variance calculation of the time-series resistances measured at 4 kHz can detect the effect of FCCP at concentrations as low as 1 μM. For wound healing migration, the recovery resistance curves were fitted by sigmoid curve and the hill slope showed a concentration-dependent decline from 0.3 μM to 3 μM, indicating a decrease in cell migration rate. Moreover, dose dependent incline of the inflection points from 0.3 μM to 3 μM FCCP implied the increase of the half time for wound recovery migration. Together, our results demonstrate that partial uncoupling of mitochondrial oxidative phosphorylation reduces micromotion and wound healing migration of hMSCs. The ECIS method used in this study offers a simple and sensitive approach to investigate stem cell migration and its regulation by mitochondrial dynamics.
Collapse
Affiliation(s)
- Sheng-Po Chiu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei 11490, Taiwan
| | - Yu-Wei Lee
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan
| | - Ling-Yi Wu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei 11490, Taiwan
| | - Tse-Hua Tung
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan
| | - Sofia Gomez
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
27
|
Rastogi A, Joshi P, Contreras E, Gama V. Remodeling of mitochondrial morphology and function: an emerging hallmark of cellular reprogramming. Cell Stress 2019; 3:181-194. [PMID: 31225513 PMCID: PMC6558935 DOI: 10.15698/cst2019.06.189] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Research in the stem cell field has traditionally focused on understanding key transcriptional factors that provide pluripotent cell identity. However, much less is known about other critical non-transcriptional signaling networks that govern stem cell identity. Although we continue to gain critical insights into the mechanisms underlying mitochondrial morphology and function during cellular reprogramming – the process of reverting the fate of a differentiated cell into a stem cell, many uncertainties remain. Recent studies suggest an emerging landscape in which mitochondrial morphology and function have an active role in maintaining and regulating changes in cell identity. In this review, we will focus on these emerging concepts as crucial modulators of cellular reprogramming. Recognition of the widespread applicability of these concepts will increase our understanding of the mitochondrial mechanisms involved in cell identity, cell fate and disease.
Collapse
Affiliation(s)
- Anuj Rastogi
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Piyush Joshi
- Neuroscience Program, Vanderbilt University Medical Center, Nashville, TN 37240.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37240
| | - Ela Contreras
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Vivian Gama
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37240.,Neuroscience Program, Vanderbilt University Medical Center, Nashville, TN 37240.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37240.,Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37240
| |
Collapse
|
28
|
Chicherin IV, Dashinimaev E, Baleva M, Krasheninnikov I, Levitskii S, Kamenski P. Cytochrome c Oxidase on the Crossroads of Transcriptional Regulation and Bioenergetics. Front Physiol 2019; 10:644. [PMID: 31231235 PMCID: PMC6558401 DOI: 10.3389/fphys.2019.00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 05/07/2019] [Indexed: 11/13/2022] Open
Abstract
Mitochondria are the organelles of eukaryotic cells responsible for the ATP production by means of the electron transfer chain (ETC). Its work is under strict genetic control providing the correct assembly of the enzyme complexes and the interface to adapt the energetic demands of the cell to the environment. These mechanisms are particularly developed in the cells with high energy consumption, like neurons and myocytes. This review summarizes several aspects of the involvement of the ETC complexes in the transcriptional control mechanisms of the neurons and other cells. Their influence on the differentiation of neurons is also discussed.
Collapse
Affiliation(s)
- Ivan Vladimirovich Chicherin
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, Russia
| | - Erdem Dashinimaev
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.,Pirogov Russian National Research Medical University, Moscow, Russia
| | - Mariia Baleva
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Igor Krasheninnikov
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Sergey Levitskii
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Piotr Kamenski
- Department of Molecular Biology, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
29
|
Princz A, Kounakis K, Tavernarakis N. Mitochondrial contributions to neuronal development and function. Biol Chem 2019; 399:723-739. [PMID: 29476663 DOI: 10.1515/hsz-2017-0333] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
Abstract
Mitochondria are critical to tissues and organs characterized by high-energy demands, such as the nervous system. They provide essential energy and metabolites, and maintain Ca2+ balance, which is imperative for proper neuronal function and development. Emerging findings further underline the role of mitochondria in neurons. Technical advances in the last decades made it possible to investigate key mechanisms in neuronal development and the contribution of mitochondria therein. In this article, we discuss the latest findings relevant to the involvement of mitochondria in neuronal development, placing emphasis on mitochondrial metabolism and dynamics. In addition, we survey the role of mitochondrial energy metabolism and Ca2+ homeostasis in proper neuronal function, and the involvement of mitochondria in axon myelination.
Collapse
Affiliation(s)
- Andrea Princz
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
- Department of Biology, University of Crete, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
| | - Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, N. Plastira 100, Vassilika Vouton, Heraklion 70013, Crete, Greece
| |
Collapse
|
30
|
Pharmacological targeting of mitochondria in cancer stem cells: An ancient organelle at the crossroad of novel anti-cancer therapies. Pharmacol Res 2019; 139:298-313. [DOI: 10.1016/j.phrs.2018.11.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/13/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|
31
|
Xie K, Ngo S, Rong J, Sheppard A. Modulation of mitochondrial respiration underpins neuronal differentiation enhanced by lutein. Neural Regen Res 2019; 14:87-99. [PMID: 30531082 PMCID: PMC6262990 DOI: 10.4103/1673-5374.243713] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Lutein is a dietary carotenoid of particular nutritional interest as it is preferentially taken up by neural tissues. Often linked with beneficial effects on vision, a broader role for lutein in neuronal differentiation has emerged recently, although the underlying mechanisms for these effects are not yet clear. The purpose of this study was to investigate the effect of lutein on neuronal differentiation and explore the associated underpinning mechanisms. We found that lutein treatment enhanced the differentiation of SH-SY5Y cells, specifically increasing neuronal arborization and expression of the neuronal process filament protein microtubule-associated protein 2. This effect was mediated by the intracellular phosphoinositide-3-kinase (PI3K) signaling pathway. While PI3K activity is a known trigger of neuronal differentiation, more recently it has also been shown to modulate the metabolic state of cells. Our analysis of bioenergetics found that lutein treatment increased glucose consumption, rates of glycolysis and enhanced respiratory activity of mitochondrial complexes. Concomitantly, the generation of reactive oxygen species was increased (consistent with previous reports that reactive oxygen species promote neuronal differentiation), as well as the production of the key metabolic intermediate acetyl-CoA, an essential determinant of epigenetic status in the cell. We suggest that lutein-stimulated neuronal differentiation is mediated by PI3K-dependent modulation of mitochondrial respiration and signaling, and that the consequential metabolic shifts initiate epigenetically dependent transcriptomic reprogramming in support of this morphogenesis. These observations support the potential importance of micronutrients supplementation to neurogenesis, both during normal development and in regenerative repair.
Collapse
Affiliation(s)
- Kui Xie
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Sherry Ngo
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Jing Rong
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Allan Sheppard
- Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
32
|
Perestrelo T, Correia M, Ramalho-Santos J, Wirtz D. Metabolic and Mechanical Cues Regulating Pluripotent Stem Cell Fate. Trends Cell Biol 2018; 28:1014-1029. [DOI: 10.1016/j.tcb.2018.09.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/30/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
|
33
|
Pashkovskaia N, Gey U, Rödel G. Mitochondrial ROS direct the differentiation of murine pluripotent P19 cells. Stem Cell Res 2018; 30:180-191. [PMID: 29957443 DOI: 10.1016/j.scr.2018.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/07/2018] [Accepted: 06/10/2018] [Indexed: 01/20/2023] Open
Abstract
ROS are frequently associated with deleterious effects caused by oxidative stress. Despite the harmful effects of non-specific oxidation, ROS also function as signal transduction molecules that regulate various biological processes, including stem cell proliferation and differentiation. Here we show that mitochondrial ROS level determines cell fate during differentiation of the pluripotent stem cell line P19. As stem cells in general, P19 cells are characterized by a low respiration activity, accompanied by a low level of ROS formation. Nevertheless, we found that P19 cells contain fully assembled mitochondrial electron transport chain supercomplexes (respirasomes), suggesting that low respiration activity may serve as a protective mechanism against ROS. Upon elevated mitochondrial ROS formation, the proliferative potential of P19 cells is decreased due to longer S phase of the cell cycle. Our data show that besides being harmful, mitochondrial ROS production regulates the differentiation potential of P19 cells: elevated mitochondrial ROS level favours trophoblast differentiation, whereas preventing neuron differentiation. Therefore, our results suggest that mitochondrial ROS level serves as an important factor that directs differentiation towards certain cell types while preventing others.
Collapse
Affiliation(s)
| | - Uta Gey
- Institute of Genetics, Technische Universität Dresden, Dresden 01217, Germany
| | - Gerhard Rödel
- Institute of Genetics, Technische Universität Dresden, Dresden 01217, Germany
| |
Collapse
|
34
|
Xie K, Sheppard A. Dietary Micronutrients Promote Neuronal Differentiation by Modulating the Mitochondrial‐Nuclear Dialogue. Bioessays 2018; 40:e1800051. [DOI: 10.1002/bies.201800051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 04/30/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Kui Xie
- Liggins InstituteUniversity of AucklandGraftonAuckland 1023New Zealand
| | - Allan Sheppard
- Liggins InstituteUniversity of AucklandGraftonAuckland 1023New Zealand
| |
Collapse
|
35
|
Trixl L, Amort T, Wille A, Zinni M, Ebner S, Hechenberger C, Eichin F, Gabriel H, Schoberleitner I, Huang A, Piatti P, Nat R, Troppmair J, Lusser A. RNA cytosine methyltransferase Nsun3 regulates embryonic stem cell differentiation by promoting mitochondrial activity. Cell Mol Life Sci 2018; 75:1483-1497. [PMID: 29103146 PMCID: PMC5852174 DOI: 10.1007/s00018-017-2700-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 10/17/2017] [Accepted: 10/26/2017] [Indexed: 12/14/2022]
Abstract
Chemical modifications of RNA have been attracting increasing interest because of their impact on RNA fate and function. Therefore, the characterization of enzymes catalyzing such modifications is of great importance. The RNA cytosine methyltransferase NSUN3 was recently shown to generate 5-methylcytosine in the anticodon loop of mitochondrial tRNAMet. Further oxidation of this position is required for normal mitochondrial translation and function in human somatic cells. Because embryonic stem cells (ESCs) are less dependent on oxidative phosphorylation than somatic cells, we examined the effects of catalytic inactivation of Nsun3 on self-renewal and differentiation potential of murine ESCs. We demonstrate that Nsun3-mutant cells show strongly reduced mt-tRNAMet methylation and formylation as well as reduced mitochondrial translation and respiration. Despite the lower dependence of ESCs on mitochondrial activity, proliferation of mutant cells was reduced, while pluripotency marker gene expression was not affected. By contrast, ESC differentiation was skewed towards the meso- and endoderm lineages at the expense of neuroectoderm. Wnt3 was overexpressed in early differentiating mutant embryoid bodies and in ESCs, suggesting that impaired mitochondrial function disturbs normal differentiation programs by interfering with cellular signalling pathways. Interestingly, basal levels of reactive oxygen species (ROS) were not altered in ESCs, but Nsun3 inactivation attenuated induction of mitochondrial ROS upon stress, which may affect gene expression programs upon differentiation. Our findings not only characterize Nsun3 as an important regulator of stem cell fate but also provide a model system to study the still incompletely understood interplay of mitochondrial function with stem cell pluripotency and differentiation.
Collapse
Affiliation(s)
- Lukas Trixl
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Thomas Amort
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Alexandra Wille
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Manuela Zinni
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Susanne Ebner
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Clara Hechenberger
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Felix Eichin
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Hanna Gabriel
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Ines Schoberleitner
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Anming Huang
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | | | - Roxana Nat
- Institute for Neuroscience, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, 6020, Innsbruck, Austria
| | - Alexandra Lusser
- Division of Molecular Biology, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Innovations in agriculture and medicine as well as industrial and domestic technologies are essential for the growing and aging global population. These advances generally require the use of novel natural or synthetic chemical agents with the potential to affect human health. Here, we attempt to highlight environmental chemicals and select drugs with the potential to exacerbate aging by directly affecting molecular aging cascades focusing particular attention on the brain. Finally, we call attention to some potential fruitful areas of research, particularly with advanced molecular profiling that could aid in prevention or mitigation of environmental chemical toxic influences in the periphery and the brain. RECENT FINDINGS We briefly summarize new research and highlight a recent study designed to prospectively identify agrochemicals with the potential to induce neurological diseases and place these discoveries into the already rich neurodegeneration and aging literature. Collectively, the research reviewed briefly here highlight chemicals with the true potential to accelerate aging, particularly in the brain, by eliciting elevated free radical stress and mitochondrial dysfunction. We make general recommendations about improved methodological approaches toward identification and regulation of chemicals that are gerontogenic to the brain.
Collapse
Affiliation(s)
- Brandon L Pearson
- DZNE, German Center for Neurodegenerative Diseases, Sigmund-Freud Str 27, 53127, Bonn, Germany.
| | - Dan Ehninger
- DZNE, German Center for Neurodegenerative Diseases, Sigmund-Freud Str 27, 53127, Bonn, Germany
| |
Collapse
|
37
|
García-Prat L, Sousa-Victor P, Muñoz-Cánoves P. Proteostatic and Metabolic Control of Stemness. Cell Stem Cell 2018; 20:593-608. [PMID: 28475885 DOI: 10.1016/j.stem.2017.04.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Adult stem cells, particularly those resident in tissues with little turnover, are largely quiescent and only activate in response to regenerative demands, while embryonic stem cells continuously replicate, suggesting profoundly different regulatory mechanisms within distinct stem cell types. In recent years, evidence linking metabolism, mitochondrial dynamics, and protein homeostasis (proteostasis) as fundamental regulators of stem cell function has emerged. Here, we discuss new insights into how these networks control potency, self-renewal, differentiation, and aging of highly proliferative embryonic stem cells and quiescent adult stem cells, with a focus on hematopoietic and muscle stem cells and implications for anti-aging research.
Collapse
Affiliation(s)
- Laura García-Prat
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), E-08003 Barcelona, Spain; Spanish National Center on Cardiovascular Research (CNIC), E-28029 Madrid, Spain; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Pedro Sousa-Victor
- Paul F. Glenn Center for Biology of Aging Research, Buck Institute for Research on Aging, Novato, CA 94945-1400, USA
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), E-08003 Barcelona, Spain; Spanish National Center on Cardiovascular Research (CNIC), E-28029 Madrid, Spain; ICREA, E-08010 Barcelona, Spain.
| |
Collapse
|
38
|
Russell OM, Fruh I, Rai PK, Marcellin D, Doll T, Reeve A, Germain M, Bastien J, Rygiel KA, Cerino R, Sailer AW, Lako M, Taylor RW, Mueller M, Lightowlers RN, Turnbull DM, Helliwell SB. Preferential amplification of a human mitochondrial DNA deletion in vitro and in vivo. Sci Rep 2018; 8:1799. [PMID: 29379065 PMCID: PMC5789095 DOI: 10.1038/s41598-018-20064-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 12/27/2017] [Indexed: 01/19/2023] Open
Abstract
We generated induced pluripotent stem cells (iPSCs) from patient fibroblasts to yield cell lines containing varying degrees of heteroplasmy for a m.13514 A > G mtDNA point mutation (2 lines) and for a ~6 kb single, large scale mtDNA deletion (3 lines). Long term culture of the iPSCs containing a single, large-scale mtDNA deletion showed consistent increase in mtDNA deletion levels with time. Higher levels of mtDNA heteroplasmy correlated with increased respiratory deficiency. To determine what changes occurred in deletion level during differentiation, teratomas comprising all three embryonic germ layers were generated from low (20%) and intermediate heteroplasmy (55%) mtDNA deletion clones. Regardless of whether iPSCs harbouring low or intermediate mtDNA heteroplasmy were used, the final levels of heteroplasmy in all teratoma germ layers increased to a similar high level (>60%). Thus, during human stem cell division, cells not only tolerate high mtDNA deletion loads but seem to preferentially replicate deleted mtDNA genomes. This has implications for the involvement of mtDNA deletions in both disease and ageing.
Collapse
Affiliation(s)
- Oliver M Russell
- Wellcome Centre for Mitochondrial Research, Institutes of Neuroscience and Cellular and Molecular Bioscience, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, Tyne and Wear, UK
| | - Isabelle Fruh
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Pavandeep K Rai
- Wellcome Centre for Mitochondrial Research, Institutes of Neuroscience and Cellular and Molecular Bioscience, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, Tyne and Wear, UK
| | - David Marcellin
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Thierry Doll
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Amy Reeve
- Wellcome Centre for Mitochondrial Research, Institutes of Neuroscience and Cellular and Molecular Bioscience, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, Tyne and Wear, UK
| | - Mitchel Germain
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Julie Bastien
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Karolina A Rygiel
- Wellcome Centre for Mitochondrial Research, Institutes of Neuroscience and Cellular and Molecular Bioscience, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, Tyne and Wear, UK
| | - Raffaele Cerino
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Andreas W Sailer
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Institutes of Neuroscience and Cellular and Molecular Bioscience, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, Tyne and Wear, UK
| | - Matthias Mueller
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland
| | - Robert N Lightowlers
- Wellcome Centre for Mitochondrial Research, Institutes of Neuroscience and Cellular and Molecular Bioscience, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, Tyne and Wear, UK
| | - Doug M Turnbull
- Wellcome Centre for Mitochondrial Research, Institutes of Neuroscience and Cellular and Molecular Bioscience, Newcastle University Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, Tyne and Wear, UK.
| | - Stephen B Helliwell
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel, CH-4056, Switzerland.
| |
Collapse
|
39
|
Keller A, Dziedzicka D, Zambelli F, Markouli C, Sermon K, Spits C, Geens M. Genetic and epigenetic factors which modulate differentiation propensity in human pluripotent stem cells. Hum Reprod Update 2018; 24:162-175. [PMID: 29377992 DOI: 10.1093/humupd/dmx042] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/23/2017] [Accepted: 12/22/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Human pluripotent stem cell (hPSC) lines are known to have a bias in their differentiation. This gives individual cell lines a propensity to preferentially differentiate towards one germ layer or cell type over others. Chromosomal aberrations, mitochondrial mutations, genetic diversity and epigenetic variance are the main drivers of this phenomenon, and can lead to a wide range of phenotypes. OBJECTIVE AND RATIONALE Our aim is to provide a comprehensive overview of the different factors which influence differentiation propensity. Specifically, we sought to highlight known genetic variances and their mechanisms, in addition to more general observations from larger abnormalities. Furthermore, we wanted to provide an up-to-date list of a growing number of predictive indicators which are able to identify differentiation propensity before the initiation of differentiation. As differentiation propensity can lead to difficulties in both research as well as clinical translation, our thorough overview could be a useful tool. SEARCH METHODS Combinations of the following key words were applied as search criteria in the PubMed database: embryonic stem cells, induced pluripotent stem cells, differentiation propensity (also: potential, efficiency, capacity, bias, variability), epigenetics, chromosomal abnormalities, genetic aberrations, X chromosome inactivation, mitochondrial function, mitochondrial metabolism, genetic diversity, reprogramming, predictive marker, residual stem cell, clinic. Only studies in English were included, ranging from 2000 to 2017, with a majority ranging from 2010 to 1017. Further manuscripts were added from cross-references. OUTCOMES Differentiation propensity is affected by a wide variety of (epi)genetic factors. These factors clearly lead to a loss of differentiation capacity, preference towards certain cell types and oftentimes, phenotypes which begin to resemble cancer. Broad changes in (epi)genetics, such as aneuploidies or wide-ranging modifications to the epigenetic landscape tend to lead to extensive, less definite changes in differentiation capacity, whereas more specific abnormalities often have precise ramifications in which certain cell types become more preferential. Furthermore, there appears to be a greater, though often less considered, contribution to differentiation propensity by factors such as mitochondria and inherent genetic diversity. Varied differentiation capacity can also lead to potential consequences in the clinical translation of hPSC, including the occurrence of residual undifferentiated stem cells, and the transplantation of potentially transformed cells. WIDER IMPLICATIONS As hPSC continue to advance towards the clinic, our understanding of them progresses as well. As a result, the challenges faced become more numerous, but also more clear. If the transition to the clinic is to be achieved with a minimum number of potential setbacks, thorough evaluation of the cells will be an absolute necessity. Altered differentiation propensity represents at least one such hurdle, for which researchers and eventually clinicians will need to find solutions. Already, steps are being taken to tackle the issue, though further research will be required to evaluate any long-term risks it poses.
Collapse
Affiliation(s)
- Alexander Keller
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Dominika Dziedzicka
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Filippo Zambelli
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Christina Markouli
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Karen Sermon
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Claudia Spits
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| | - Mieke Geens
- Research group Reproduction and Genetics (REGE), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Jette, Belgium
| |
Collapse
|
40
|
Khacho M, Slack RS. Mitochondrial activity in the regulation of stem cell self-renewal and differentiation. Curr Opin Cell Biol 2017; 49:1-8. [DOI: 10.1016/j.ceb.2017.11.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 10/25/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022]
|
41
|
Kim HB, Baik KY, Choung PH, Chung JH. Pulse frequency dependency of photobiomodulation on the bioenergetic functions of human dental pulp stem cells. Sci Rep 2017; 7:15927. [PMID: 29162863 PMCID: PMC5698451 DOI: 10.1038/s41598-017-15754-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/01/2017] [Indexed: 11/09/2022] Open
Abstract
Photobiomodulation (PBM) therapy contributes to pain relief, wound healing, and tissue regeneration. The pulsed wave (PW) mode has been reported to be more effective than the continuous wave (CW) mode when applying PBM to many biological systems. However, the reason for the higher effectiveness of PW-PBM is poorly understood. Herein, we suggest using delayed luminescence (DL) as a reporter of mitochondrial activity after PBM treatment. DL originates mainly from mitochondrial electron transport chain systems, which produce reactive oxygen species (ROS) and adenosine triphosphate (ATP). The decay time of DL depends on the pulse frequencies of applied light, which correlate with the biological responses of human dental pulp stem cells (hDPSCs). Using a low-power light whose wavelength is 810 nm and energy density is 38 mJ/cm2, we find that a 300-Hz pulse frequency prolonged the DL pattern and enhanced alkaline phosphatase activity. In addition, we analyze mitochondrial morphological changes and their volume density and find evidence supporting mitochondrial physiological changes from PBM treatment. Our data suggest a new methodology for determining the effectiveness of PBM and the specific pulse frequency dependency of PBM in the differentiation of hDPSCs.
Collapse
Affiliation(s)
- Hong Bae Kim
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ku Youn Baik
- Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Pill-Hoon Choung
- Department of Oral and Maxillofacial Surgery and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jong Hoon Chung
- Department of Biosystems & Biomaterials Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea. .,Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
42
|
Perestrelo T, Chen W, Correia M, Le C, Pereira S, Rodrigues AS, Sousa MI, Ramalho-Santos J, Wirtz D. Pluri-IQ: Quantification of Embryonic Stem Cell Pluripotency through an Image-Based Analysis Software. Stem Cell Reports 2017; 9:697-709. [PMID: 28712847 PMCID: PMC5549834 DOI: 10.1016/j.stemcr.2017.06.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/12/2017] [Accepted: 06/13/2017] [Indexed: 02/07/2023] Open
Abstract
Image-based assays, such as alkaline phosphatase staining or immunocytochemistry for pluripotent markers, are common methods used in the stem cell field to assess pluripotency. Although an increased number of image-analysis approaches have been described, there is still a lack of software availability to automatically quantify pluripotency in large images after pluripotency staining. To address this need, we developed a robust and rapid image processing software, Pluri-IQ, which allows the automatic evaluation of pluripotency in large low-magnification images. Using mouse embryonic stem cells (mESC) as a model, we combined an automated segmentation algorithm with a supervised machine-learning platform to classify colonies as pluripotent, mixed, or differentiated. In addition, Pluri-IQ allows the automatic comparison between different culture conditions. This efficient user-friendly open-source software can be easily implemented in images derived from pluripotent cells or cells that express pluripotent markers (e.g., OCT4-GFP) and can be routinely used, decreasing image assessment bias. Open-source software to evaluate pluripotency in low-magnification images Automatic colony detection and segmentation Supervised machine-learning platform with high characterization accuracy Software tools for easy data validation, visualization, and data analysis comparison
Collapse
Affiliation(s)
- Tânia Perestrelo
- PhD Program in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra 3030-789, Portugal; Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal; Institute for Nanobiotechnology at Johns Hopkins University, Baltimore, MD 21218, USA; Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Weitong Chen
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Marcelo Correia
- PhD Program in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra 3030-789, Portugal; Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
| | - Christopher Le
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sandro Pereira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
| | - Ana S Rodrigues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal
| | - Maria I Sousa
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal; Department of Life Sciences, University of Coimbra, Coimbra 3000-456, Portugal
| | - João Ramalho-Santos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra 3004-504, Portugal; Department of Life Sciences, University of Coimbra, Coimbra 3000-456, Portugal.
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Johns Hopkins Physical Sciences - Oncology Center, The Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
43
|
Ducray AD, Felser A, Zielinski J, Bittner A, Bürgi JV, Nuoffer JM, Frenz M, Mevissen M. Effects of silica nanoparticle exposure on mitochondrial function during neuronal differentiation. J Nanobiotechnology 2017; 15:49. [PMID: 28676089 PMCID: PMC5496409 DOI: 10.1186/s12951-017-0284-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/17/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Nanomedicine offers a promising tool for therapies of brain diseases, but potential effects on neuronal health and neuronal differentiation need to be investigated to assess potential risks. The aim of this study was to investigate effects of silica-indocyanine green/poly (ε-caprolactone) nanoparticles (PCL-NPs) engineered for laser tissue soldering in the brain before and during differentiation of SH-SY5Y cells. Considering adaptations in mitochondrial homeostasis during neuronal differentiation, metabolic effects of PCL-NP exposure before and during neuronal differentiation were studied. In addition, kinases of the PI3 kinase (PI3-K/Akt) and the MAP kinase (MAP-K/ERK) pathways related to neuronal differentiation and mitochondrial function were investigated. RESULTS Differentiation resulted in a decrease in the cellular respiration rate and the extracellular acidification rate (ECAR). PCL-NP exposure impaired mitochondrial function depending on the time of exposure. The cellular respiration rate was significantly reduced compared to differentiated controls when PCL-NPs were given before differentiation. The shift in ECAR was less pronounced in PCL-NP exposure during differentiation. Differentiation and PCL-NP exposure had no effect on expression levels and the enzymatic activity of respiratory chain complexes. The activity of the glycolytic enzyme phosphofructokinase was significantly reduced after differentiation with the effect being more pronounced after PCL-NP exposure before differentiation. The increase in mitochondrial membrane potential observed after differentiation was not found in SH-SY5Y cells exposed to PCL-NPs before differentiation. The cellular adenosine triphosphate (ATP) production significantly dropped during differentiation, and this effect was independent of the PCL-NP exposure. Differentiation and nanoparticle exposure had no effect on superoxide levels at the endpoint of the experiments. A slight decrease in the expression of the neuronal differentiation markers was found after PCL-NP exposure, but no morphological variation was observed. CONCLUSIONS PCL-NP exposure affects mitochondrial function depending on the time of exposure before and during neuronal differentiation. PCL-NP exposure during differentiation was associated with impaired mitochondrial function, which may affect differentiation. Considering the importance of adaptations in cellular respiration for neuronal differentiation and function, further studies are needed to unravel the underlying mechanisms and consequences to assess the possible risks including neurodegeneration.
Collapse
Affiliation(s)
- Angélique D Ducray
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Andrea Felser
- Institute of Clinical Chemistry, University Hospital Bern, 3010, Bern, Switzerland
| | - Jana Zielinski
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Aniela Bittner
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Julia V Bürgi
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland
| | - Jean-Marc Nuoffer
- Institute of Clinical Chemistry, University Hospital Bern, 3010, Bern, Switzerland
| | - Martin Frenz
- Institute of Applied Physics, University of Bern, Sidlerstrasse 5, 3012, Bern, Switzerland
| | - Meike Mevissen
- Division of Pharmacology and Toxicology, Vetsuisse Faculty, University of Bern, Laenggassstrasse 124, 3012, Bern, Switzerland.
| |
Collapse
|
44
|
Gibson GE, Thakkar A. Mitochondria/metabolic reprogramming in the formation of neurons from peripheral cells: Cause or consequence and the implications to their utility. Neurochem Int 2017. [PMID: 28627365 DOI: 10.1016/j.neuint.2017.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The induction of pluripotent stem cells (iPSC) from differentiated cells such as fibroblasts and their subsequent conversion to neural progenitor cells (NPC) and finally to neurons is intriguing scientifically, and its potential to medicine is nearly infinite, but unrealized. A better understanding of the changes at each step of the transformation will enable investigators to better model neurological disease. Each step of conversion from a differentiated cell to an iPSC to a NPC to neurons requires large changes in glycolysis including aerobic glycolysis, the pentose shunt, the tricarboxylic acid cycle, the electron transport chain and in the production of reactive oxygen species (ROS). These mitochondrial/metabolic changes are required and their manipulation modifies conversions. These same mitochondrial/metabolic processes are altered in common neurological diseases so that factors related to the disease may alter the cellular transformation at each step including the final phenotype. A lack of understanding of these interactions could compromise the validity of the disease comparisons in iPSC derived neurons. Both the complexity and potential of iPSC derived cells for understanding and treating disease remain great.
Collapse
Affiliation(s)
- Gary E Gibson
- Weil Cornell Medicine, Brain and Mind Research Institute, Burke Medical Research, White Plains, NY 10605, United States.
| | - Ankita Thakkar
- Weil Cornell Medicine, Brain and Mind Research Institute, Burke Medical Research, White Plains, NY 10605, United States
| |
Collapse
|
45
|
Yoffe Y, David M, Kalaora R, Povodovski L, Friedlander G, Feldmesser E, Ainbinder E, Saada A, Bialik S, Kimchi A. Cap-independent translation by DAP5 controls cell fate decisions in human embryonic stem cells. Genes Dev 2017; 30:1991-2004. [PMID: 27664238 PMCID: PMC5066241 DOI: 10.1101/gad.285239.116] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/18/2016] [Indexed: 12/21/2022]
Abstract
In this study, Yoffe et al. provide insight into a new regulatory mechanism that is critical for stem cell fate decisions toward several cell lineages. They found that DAP5-mediated translation of a specific set of proteins is critical for the transition from pluripotency to differentiation, highlighting the importance of cap-independent translation in stem cell fate decisions. Multiple transcriptional and epigenetic changes drive differentiation of embryonic stem cells (ESCs). This study unveils an additional level of gene expression regulation involving noncanonical, cap-independent translation of a select group of mRNAs. This is driven by death-associated protein 5 (DAP5/eIF4G2/NAT1), a translation initiation factor mediating IRES-dependent translation. We found that the DAP5 knockdown from human ESCs (hESCs) resulted in persistence of pluripotent gene expression, delayed induction of differentiation-associated genes in different cell lineages, and defective embryoid body formation. The latter involved improper cellular organization, lack of cavitation, and enhanced mislocalized apoptosis. RNA sequencing of polysome-associated mRNAs identified candidates with reduced translation efficiency in DAP5-depleted hESCs. These were enriched in mitochondrial proteins involved in oxidative respiration, a pathway essential for differentiation, the significance of which was confirmed by the aberrant mitochondrial morphology and decreased oxidative respiratory activity in DAP5 knockdown cells. Further analysis identified the chromatin modifier HMGN3 as a cap-independent DAP5 translation target whose knockdown resulted in defective differentiation. Thus, DAP5-mediated translation of a specific set of proteins is critical for the transition from pluripotency to differentiation, highlighting the importance of cap-independent translation in stem cell fate decisions.
Collapse
Affiliation(s)
- Yael Yoffe
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Maya David
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rinat Kalaora
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Lital Povodovski
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gilgi Friedlander
- Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ester Feldmesser
- Bioinformatics Unit, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elena Ainbinder
- Stem Cell Core Unit, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ann Saada
- Monique and Jacques Roboh Department of Genetic Research, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; Department of Genetics and Metabolic Diseases, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Shani Bialik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
46
|
Abstract
INTRODUCTION Research reveals cardiac regeneration exists at levels previously deemed unattainable. Clinical trials using stem cells demonstrate promising cardiomyogenic and regenerative potential but insufficient contractile recovery. Incomplete understanding of the biology of administered cells likely contributes to inconsistent patient outcomes. Metabolism is a core component of many well-characterized stem cell types, and metabolic changes fundamentally alter stem cell fate from self-renewal to lineage commitment, and vice versa. However, the metabolism of stem cells currently studied for cardiac regeneration remains incompletely understood. Areas covered: Key metabolic features of stem cells are reviewed and unique stem cell metabolic characteristics are discussed. Metabolic changes altering stem cell fate are considered from quiescence and self-renewal to lineage commitment. Key metabolic concepts are applied toward examining cardiac regeneration through stem cell-based approaches, and clinical implications of current cell therapies are evaluated to identify potential areas of improvement. Expert commentary: The metabolism and biology of stem cells used for cardiac therapy remain poorly characterized. A growing appreciation for the fundamental relationship between stem cell functionality and metabolic phenotype is developing. Future studies unraveling links between cardiac stem cell metabolism and regenerative potential may considerably improve treatment strategies and therapeutic outcomes.
Collapse
Affiliation(s)
- Dieter A Kubli
- a San Diego State University , Integrated Regenerative Research Institute , San Diego , CA , USA
| | - Mark A Sussman
- a San Diego State University , Integrated Regenerative Research Institute , San Diego , CA , USA
| |
Collapse
|
47
|
Distinct Effects of miR-210 Reduction on Neurogenesis: Increased Neuronal Survival of Inflammation But Reduced Proliferation Associated with Mitochondrial Enhancement. J Neurosci 2017; 37:3072-3084. [PMID: 28188219 DOI: 10.1523/jneurosci.1777-16.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 01/19/2023] Open
Abstract
Neurogenesis is essential to brain development and plays a central role in the response to brain injury. Stroke and head trauma stimulate proliferation of endogenous neural stem cells (NSCs); however, the survival of young neurons is sharply reduced by postinjury inflammation. Cellular mitochondria are critical to successful neurogenesis and are a major target of inflammatory injury. Mitochondrial protection was shown to improve survival of young neurons. This study tested whether reducing cellular microRNA-210 (miR-210) would enhance mitochondrial function and improve survival of young murine neurons under inflammatory conditions. Several studies have demonstrated the potential of miR-210 inhibition to enhance and protect mitochondrial function through upregulation of mitochondrial proteins. Here, miR-210 inhibition significantly increased neuronal survival and protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase in differentiating NSC cultures exposed to inflammatory mediators. Unexpectedly, we found that reducing miR-210 significantly attenuated NSC proliferation upon induction of differentiation. Further investigation revealed that increased mitochondrial function suppressed the shift to primarily glycolytic metabolism and reduced mitochondrial length characteristic of dividing cells. Activation of AMP-regulated protein kinase-retinoblastoma signaling is important in NSC proliferation and the reduction of this activation observed by miR-210 inhibition is one mechanism contributing to the reduced proliferation. Postinjury neurogenesis occurs as a burst of proliferation that peaks in days, followed by migration and differentiation over weeks. Our studies suggest that mitochondrial protective miR-210 inhibition should be delayed until after the initial burst of proliferation, but could be beneficial during the prolonged differentiation stage.SIGNIFICANCE STATEMENT Increasing the success of endogenous neurogenesis after brain injury holds therapeutic promise. Postinjury inflammation markedly reduces newborn neuron survival. This study found that enhancement of mitochondrial function by reducing microRNA-210 (miR-210) levels could improve survival of young neurons under inflammatory conditions. miR-210 inhibition protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase. Conversely, we observed decreased precursor cell proliferation likely due to suppression of the AMP-regulated protein kinase-retinoblastoma axis with miR-210 inhibition. Therefore, mitochondrial protection is a double-edged sword: early inhibition reduces proliferation, but inhibition later significantly increases neuroblast survival. This explains in part the contradictory published reports of the effects of miR-210 on neurogenesis.
Collapse
|
48
|
Correia M, Perestrelo T, Rodrigues AS, Ribeiro MF, Pereira SL, Sousa MI, Ramalho-Santos J. Sirtuins in metabolism, stemness and differentiation. Biochim Biophys Acta Gen Subj 2017; 1861:3444-3455. [DOI: 10.1016/j.bbagen.2016.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/16/2016] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
|
49
|
Almeida AS, Vieira HLA. Role of Cell Metabolism and Mitochondrial Function During Adult Neurogenesis. Neurochem Res 2016; 42:1787-1794. [DOI: 10.1007/s11064-016-2150-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 12/15/2022]
|
50
|
Lapel M, Weston P, Strassheim D, Karoor V, Burns N, Lyubchenko T, Paucek P, Stenmark KR, Gerasimovskaya EV. Glycolysis and oxidative phosphorylation are essential for purinergic receptor-mediated angiogenic responses in vasa vasorum endothelial cells. Am J Physiol Cell Physiol 2016; 312:C56-C70. [PMID: 27856430 PMCID: PMC5283894 DOI: 10.1152/ajpcell.00250.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/03/2016] [Indexed: 11/24/2022]
Abstract
Angiogenesis is an energy-demanding process; however, the role of cellular energy pathways and their regulation by extracellular stimuli, especially extracellular nucleotides, remain largely unexplored. Using metabolic inhibitors of glycolysis (2-deoxyglucose) and oxidative phosphorylation (OXPHOS) (oligomycin, rotenone, and FCCP), we demonstrate that glycolysis and OXPHOS are both essential for angiogenic responses of vasa vasorum endothelial cell (VVEC). Treatment with P2R agonists, ATP, and 2-methylthioadenosine diphosphate trisodium salt (MeSADP), but not P1 receptor agonist, adenosine, increased glycolytic activity in VVEC (measured by extracellular acidification rate and lactate production). Stimulation of glycolysis was accompanied by increased levels of phospho-phosphofructokinase B3, hexokinase (HK), and GLUT-1, but not lactate dehydrogenase. Moreover, extracellular ATP and MeSADP, and to a lesser extent adenosine, increased basal and maximal oxygen consumption rates in VVEC. These effects were potentiated when the cells were cultured in 20 mM galactose and 5 mM glucose compared with 25 mM glucose. Treatment with P2R agonists decreased phosphorylation of pyruvate dehydrogenase (PDH)-E1α and increased succinate dehydrogenase (SDH), cytochrome oxidase IV, and β-subunit of F1F0 ATP synthase expression. In addition, P2R stimulation transiently elevated mitochondrial Ca2+ concentration, implying involvement of mitochondria in VVEC angiogenic activation. We also demonstrated a critical role of phosphatidylinositol 3-kinase and Akt pathways in lactate production, PDH-E1α phosphorylation, and the expression of HK, SDH, and GLUT-1 in ATP-stimulated VVEC. Together, our findings suggest that purinergic and metabolic regulation of VVEC energy pathways is essential for VV angiogenesis and may contribute to pathologic vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Martin Lapel
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | - Philip Weston
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | - Derek Strassheim
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | - Vijaya Karoor
- Department of Medicine, University of Colorado Denver, Aurora, Colorado; and
| | - Nana Burns
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | - Taras Lyubchenko
- Department of Medicine, University of Colorado Denver, Aurora, Colorado; and
| | - Petr Paucek
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | | |
Collapse
|