51
|
Banarase TA, Sammeta SS, Wankhede NL, Mangrulkar SV, Rahangdale SR, Aglawe MM, Taksande BG, Upaganlawar AB, Umekar MJ, Kale MB. Mitophagy regulation in aging and neurodegenerative disease. Biophys Rev 2023; 15:239-255. [PMID: 37124925 PMCID: PMC10133433 DOI: 10.1007/s12551-023-01057-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are the primary cellular energy generators, supplying the majority of adenosine triphosphate through oxidative phosphorylation, which is necessary for neuron function and survival. Mitophagy is the metabolic process of eliminating dysfunctional or redundant mitochondria. It is a type of autophagy and it is crucial for maintaining mitochondrial and neuronal health. Impaired mitophagy leads to an accumulation of damaged mitochondria and proteins leading to the dysregulation of mitochondrial quality control processes. Recent research shows the vital role of mitophagy in neurons and the pathogenesis of major neurodegenerative diseases. Mitophagy also plays a major role in the process of aging. This review describes the alterations that are being caused in the mitophagy process at the molecular level in aging and in neurodegenerative diseases, particularly Alzheimer's, Parkinson's, and Huntington's diseases and amyotrophic lateral sclerosis, also looks at how mitophagy can be exploited as a therapeutic target for these diseases.
Collapse
Affiliation(s)
- Trupti A. Banarase
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shivkumar S. Sammeta
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Nitu L. Wankhede
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shubhada V. Mangrulkar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Sandip R. Rahangdale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Manish M. Aglawe
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Brijesh G. Taksande
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Aman B. Upaganlawar
- SNJB’s Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra India 423101
| | - Milind J. Umekar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Mayur B. Kale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| |
Collapse
|
52
|
Quintana-Cabrera R, Scorrano L. Determinants and outcomes of mitochondrial dynamics. Mol Cell 2023; 83:857-876. [PMID: 36889315 DOI: 10.1016/j.molcel.2023.02.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/13/2023] [Accepted: 02/13/2023] [Indexed: 03/09/2023]
Abstract
Mitochondria are not only central organelles in metabolism and energy conversion but are also platforms for cellular signaling cascades. Classically, the shape and ultrastructure of mitochondria were depicted as static. The discovery of morphological transitions during cell death and of conserved genes controlling mitochondrial fusion and fission contributed to establishing the concept that mitochondrial morphology and ultrastructure are dynamically regulated by mitochondria-shaping proteins. These finely tuned, dynamic changes in mitochondrial shape can in turn control mitochondrial function, and their alterations in human diseases suggest that this space can be explored for drug discovery. Here, we review the basic tenets and molecular mechanisms of mitochondrial morphology and ultrastructure, describing how they can coordinately define mitochondrial function.
Collapse
Affiliation(s)
| | - Luca Scorrano
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy; Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy.
| |
Collapse
|
53
|
Pla-Martín D, Babatz F, Schauss AC. Localization of Mitochondrial Nucleoids by Transmission Electron Microscopy Using the Transgenic Expression of the Mitochondrial Helicase Twinkle and APEX2. Methods Mol Biol 2023; 2615:173-188. [PMID: 36807792 DOI: 10.1007/978-1-0716-2922-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Reminiscent of their evolutionary origin, mitochondria contain their own genome (mtDNA) compacted into the mitochondrial chromosome or nucleoid (mt-nucleoid). Many mitochondrial disorders are characterized by disruption of mt-nucleoids, either by direct mutation of genes involved in mtDNA organization or by interfering with other vital proteins for mitochondrial function. Thus, changes in mt-nucleoid morphology, distribution, and structure are a common feature in many human diseases and can be exploited as an indicator of cellular fitness. Electron microscopy provides the highest possible resolution that can be achieved, delivering spatial and structural information about all cellular structures. Recently, the ascorbate peroxidase APEX2 has been used to increase transmission electron microscopy (TEM) contrast by inducing diaminobenzidine (DAB) precipitation. DAB has the ability to accumulate osmium during classical EM sample preparation and, due to its high electron density, provides strong contrast for TEM. Among the nucleoid proteins, the mitochondrial helicase Twinkle fused with APEX2 has been successfully used to target mt-nucleoids, providing a tool to visualize these subcellular structures with high contrast and with the resolution of an electron microscope. In the presence of H2O2, APEX2 catalyzes the polymerization of DAB, generating a brown precipitate that can be visualized in specific regions of the mitochondrial matrix. Here, we provide a detailed protocol to generate murine cell lines expressing a transgenic variant of Twinkle, suitable to target and visualize mt-nucleoids. We also describe all the necessary steps to validate the cell lines prior to electron microscopy imaging and offer examples of anticipated results.
Collapse
Affiliation(s)
- David Pla-Martín
- Center for Physiology, Faculty of Medicine and University Hospital, University of Cologne, Cologne, Germany. .,Center for Molecular Medicine Cologne, CMMC, University of Cologne, Cologne, Germany.
| | - Felix Babatz
- Cologne Cluster of Excellence on Cellular stress response in Aging-associated Disease, CECAD, University of Cologne, Cologne, Germany
| | - Astrid C Schauss
- Cologne Cluster of Excellence on Cellular stress response in Aging-associated Disease, CECAD, University of Cologne, Cologne, Germany
| |
Collapse
|
54
|
Li SJ, Liu H, Wu FF, Feng DY, Zhang S, Zheng J, Wang L, Tian F, Yang YL, Wang YY. Meshed neuronal mitochondrial networks empowered by AI-powered classifiers and immersive VR reconstruction. Front Neurosci 2023; 17:1059965. [PMID: 36816131 PMCID: PMC9932543 DOI: 10.3389/fnins.2023.1059965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Mitochondrial networks are defined as a continuous matrix lumen, but the morphological feature of neuronal mitochondrial networks is not clear due to the lack of suitable analysis techniques. The aim of the present study is to develop a framework to capture and analyze the neuronal mitochondrial networks by using 4-step process composed of 2D and 3D observation, primary and secondary virtual reality (VR) analysis, with the help of artificial intelligence (AI)-powered Aivia segmentation an classifiers. In order to fulfill this purpose, we first generated the PCs-Mito-GFP mice, in which green fluorescence protein (GFP) could be expressed on the outer mitochondrial membrane specifically on the cerebellar Purkinje cells (PCs), thus all mitochondria in the giant neuronal soma, complex dendritic arborization trees and long projection axons of Purkinje cells could be easily detected under a laser scanning confocal microscope. The 4-step process resolved the complicated neuronal mitochondrial networks into discrete neuronal mitochondrial meshes. Second, we measured the two parameters of the neuronal mitochondrial meshes, and the results showed that the surface area (μm2) of mitochondrial meshes was the biggest in dendritic trees (45.30 ± 53.21), the smallest in granular-like axons (3.99 ± 1.82), and moderate in soma (27.81 ± 22.22) and silk-like axons (17.50 ± 15.19). These values showed statistically different among different subcellular locations. The volume (μm3) of mitochondrial meshes was the biggest in dendritic trees (9.97 ± 12.34), the smallest in granular-like axons (0.43 ± 0.25), and moderate in soma (6.26 ± 6.46) and silk-like axons (3.52 ± 4.29). These values showed significantly different among different subcellular locations. Finally, we found both the surface area and the volume of mitochondrial meshes in dendritic trees and soma within the Purkinje cells in PCs-Mito-GFP mice after receiving the training with the simulating long-term pilot flight concentrating increased significantly. The precise reconstruction of neuronal mitochondrial networks is extremely laborious, the present 4-step workflow powered by artificial intelligence and virtual reality reconstruction could successfully address these challenges.
Collapse
Affiliation(s)
- Shu-Jiao Li
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Teaching Demonstration Center, School of Basic Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Hui Liu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Teaching Demonstration Center, School of Basic Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China,Department of Human Anatomy, Histology and Embryology, Medical School of Yan’an University, Yan’an, China
| | - Fei-Fei Wu
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Teaching Demonstration Center, School of Basic Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Da-Yun Feng
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Shuai Zhang
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Teaching Demonstration Center, School of Basic Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Jie Zheng
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Teaching Demonstration Center, School of Basic Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China,Department of Human Anatomy, Histology and Embryology, Medical School of Yan’an University, Yan’an, China
| | - Lu Wang
- Department of Human Anatomy, Histology and Embryology, Medical School of Yan’an University, Yan’an, China,Lu Wang,
| | - Fei Tian
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Teaching Demonstration Center, School of Basic Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China,Fei Tian,
| | - Yan-Ling Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China,Yan-Ling Yang,
| | - Ya-Yun Wang
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Teaching Demonstration Center, School of Basic Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China,State Key Laboratory of Military Stomatology, School of Stomatology, Air Force Medical University (Fourth Military Medical University), Xi’an, China,*Correspondence: Ya-Yun Wang, ,
| |
Collapse
|
55
|
Li Y, Xiong Z, Jiang Y, Zhou H, Yi L, Hu Y, Zhai X, Liu J, Tian F, Chen Y. Klf4 deficiency exacerbates myocardial ischemia/reperfusion injury in mice via enhancing ROCK1/DRP1 pathway-dependent mitochondrial fission. J Mol Cell Cardiol 2023; 174:115-132. [PMID: 36509022 DOI: 10.1016/j.yjmcc.2022.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022]
Abstract
RATIONAL Excessive mitochondrial fission is considered key process involved in myocardial ischemia/reperfusion (I/R) injury. However, the upstream mechanism remains largely unclear. Decreased level of Kruppel Like Factor 4 (KLF4) has been implicated in the pathogenesis of mitochondrial dysfunction and heart's adaption to stress. However, the role of Klf4 in I/R process is not fully elucidated. This study aims to investigate how Klf4 regulates mitochondrial dynamics and further clarify its underlying mechanism during cardiac I/R injury. METHODS Loss-of-function and gain-of-function strategies were applied to investigate the role of Klf4 in cardiac I/R injury via genetic ablation or intra-myocardial adenovirus injection. Mitochondrial dynamics was analyzed by confocal microscopy in vitro and transmission electron microscopy in vivo. Chromatin immunoprecipitation and luciferase reporter assay were performed to explore the underlying mechanisms. RESULTS KLF4 was downregulated in I/R heart. Cardiac-specific Klf4 knockout significantly exacerbated cardiac dysfunction in I/R mice. Mechanistically, Klf4 deficiency aggravated mitochondrial apoptosis, reduced ATP generation and boosted ROS overproduction via enhancing DRP1-dependent mitochondrial fission. ROCK1 was identified as a kinase regulating DRP1 activity at Ser616. Klf4 deficiency upregulated the expression of ROCK1 at transcriptional level, thus increasing S616-DRP1-mediated mitochondrial fission during I/R. Finally, reconstitution of Klf4 inhibited mitochondrial fission, restored mitochondrial function and alleviated I/R injury. CONCLUSION Our study provides the first evidence that Klf4 deficiency exacerbates myocardial I/R injury through regulating ROCK1 expression at transcriptional level to induce DRP1-mediated mitochondrial fission. Targeting mitochondrial dynamics by restoring Klf4 might be potentially cardio-protective strategies attenuating I/R injury.
Collapse
Affiliation(s)
- Yueyang Li
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhenyu Xiong
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yufan Jiang
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Hao Zhou
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Li Yi
- Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingyun Hu
- Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaofeng Zhai
- The Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jie Liu
- Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Feng Tian
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100853, China; Department of Cardiology, the First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
56
|
Malheiro RF, Carmo H, Carvalho F, Silva JP. Cannabinoid-mediated targeting of mitochondria on the modulation of mitochondrial function and dynamics. Pharmacol Res 2023; 187:106603. [PMID: 36516885 DOI: 10.1016/j.phrs.2022.106603] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Mitochondria play a critical role in the regulation of several biological processes (e.g., programmed cell death, inflammation, neurotransmission, cell differentiation). In recent years, accumulating findings have evidenced that cannabinoids, a group of endogenous and exogenous (synthetic and plant-derived) psychoactive compounds that bind to cannabinoid receptors, may modulate mitochondrial function and dynamics. As such, mitochondria have gained increasing interest as central mediators in cannabinoids' pharmacological and toxicological signatures. Here, we review the mechanisms underlying the cannabinoids' modulation of mitochondrial activity and dynamics, as well as the potential implications of such mitochondrial processes' disruption on cell homeostasis and disease. Interestingly, cannabinoids may target different mitochondrial processes (e.g., regulation of intracellular calcium levels, bioenergetic metabolism, apoptosis, and mitochondrial dynamics, including mitochondrial fission and fusion, transport, mitophagy, and biogenesis), by modulating multiple and complex signaling pathways. Of note, the outcome may depend on the experimental models used, as well as the chemical structure, concentration, and exposure settings to the cannabinoid, originating equivocal data. Notably, this interaction seems to represent not only an important feature of cannabinoids' toxicological signatures, with potential implications for the onset of distinct pathological conditions (e.g., cancer, neurodegenerative diseases, metabolic syndromes), but also an opportunity to develop novel therapeutic strategies for such pathologies, which is also discussed in this review.
Collapse
Affiliation(s)
- Rui Filipe Malheiro
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Helena Carmo
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - João Pedro Silva
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
57
|
Zhang W, Liu B, Wang Y, Zhang H, He L, Wang P, Dong M. Mitochondrial dysfunction in pulmonary arterial hypertension. Front Physiol 2022; 13:1079989. [PMID: 36589421 PMCID: PMC9795033 DOI: 10.3389/fphys.2022.1079989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/29/2022] [Indexed: 01/03/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by the increased pulmonary vascular resistance due to pulmonary vasoconstriction and vascular remodeling. PAH has high disability, high mortality and poor prognosis, which is becoming a more common global health issue. There is currently no drug that can permanently cure PAH patients. The pathogenesis of PAH is still not fully elucidated. However, the role of metabolic theory in the pathogenesis of PAH is becoming clearer, especially mitochondrial metabolism. With the deepening of mitochondrial researches in recent years, more and more studies have shown that the occurrence and development of PAH are closely related to mitochondrial dysfunction, including the tricarboxylic acid cycle, redox homeostasis, enhanced glycolysis, and increased reactive oxygen species production, calcium dysregulation, mitophagy, etc. This review will further elucidate the relationship between mitochondrial metabolism and pulmonary vasoconstriction and pulmonary vascular remodeling. It might be possible to explore more comprehensive and specific treatment strategies for PAH by understanding these mitochondrial metabolic mechanisms.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Bo Liu
- Department of Cardiovascular, Geratric Diseases Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Yazhou Wang
- Department of Cardiothoracic, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Hengli Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Lang He
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China,Correspondence: Mingqing Dong, ; Lang He, ; Pan Wang,
| | - Pan Wang
- Department of Critical Care Medicine, The Traditional Chinese Medicine Hospital of Wenjiang District, Chengdu, China,Correspondence: Mingqing Dong, ; Lang He, ; Pan Wang,
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China,Correspondence: Mingqing Dong, ; Lang He, ; Pan Wang,
| |
Collapse
|
58
|
Chowdhary S, Hadjantonakis AK. Journey of the mouse primitive endoderm: from specification to maturation. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210252. [PMID: 36252215 PMCID: PMC9574636 DOI: 10.1098/rstb.2021.0252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/25/2022] [Indexed: 12/22/2022] Open
Abstract
The blastocyst is a conserved stage and distinct milestone in the development of the mammalian embryo. Blastocyst stage embryos comprise three cell lineages which arise through two sequential binary cell fate specification steps. In the first, extra-embryonic trophectoderm (TE) cells segregate from inner cell mass (ICM) cells. Subsequently, ICM cells acquire a pluripotent epiblast (Epi) or extra-embryonic primitive endoderm (PrE, also referred to as hypoblast) identity. In the mouse, nascent Epi and PrE cells emerge in a salt-and-pepper distribution in the early blastocyst and are subsequently sorted into adjacent tissue layers by the late blastocyst stage. Epi cells cluster at the interior of the ICM, while PrE cells are positioned on its surface interfacing the blastocyst cavity, where they display apicobasal polarity. As the embryo implants into the maternal uterus, cells at the periphery of the PrE epithelium, at the intersection with the TE, break away and migrate along the TE as they mature into parietal endoderm (ParE). PrE cells remaining in association with the Epi mature into visceral endoderm. In this review, we discuss our current understanding of the PrE from its specification to its maturation. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- Sayali Chowdhary
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
59
|
The Role of Mitophagy in Various Neurological Diseases as a Therapeutic Approach. Cell Mol Neurobiol 2022:10.1007/s10571-022-01302-8. [DOI: 10.1007/s10571-022-01302-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022]
|
60
|
Kobayashi H. Recent advances in understanding the metabolic plasticity of ovarian cancer: A systematic review. Heliyon 2022; 8:e11487. [PMID: 36406733 PMCID: PMC9668530 DOI: 10.1016/j.heliyon.2022.e11487] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/03/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a gynecologic malignancy with a poor prognosis due to resistance to first-line chemotherapeutic agents. Some cancer cells are primarily dependent on glycolysis, but others favor mitochondrial oxidative phosphorylation (OXPHOS) over glycolysis. Changes in metabolic reprogramming have been reported to be involved in cancer cell survival. In this review, we summarize the metabolic profiles (e.g., metabolic heterogeneity, plasticity, and reprogramming) and adaptation to the dynamic tumor microenvironment and discuss potential novel therapeutic strategies. A literature search was performed between January 2000 and March 2022 in the PubMed and Google Scholar databases using a combination of specific terms. Ovarian cancer cells, including cancer stem cells, depend on glycolysis, OXPHOS, or both for survival. Several environmental stresses, such as nutrient starvation or glucose deprivation, hypoxic stress, acidification, and excessive reactive oxygen species (ROS) generation, reprogram the metabolic pathways to adapt. The interaction between tumors and adjacent stromal cells allows cancer cells to enhance mitochondrial energy metabolism. The metabolic reprogramming varies depending on genomic and epigenetic alterations of metabolism-related genes and the metabolic environment. Developing accurate and non-invasive methods for early identification of metabolic alterations could facilitate optimal cancer diagnosis and treatment. Cancer metabolism research has entered an exciting era where novel strategies targeting metabolic profiling will become more innovative.
Collapse
|
61
|
Xu Y, Yang X. Autophagy and pluripotency: self-eating your way to eternal youth. Trends Cell Biol 2022; 32:868-882. [PMID: 35490141 PMCID: PMC10433133 DOI: 10.1016/j.tcb.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 01/18/2023]
Abstract
Pluripotent stem cells (PSCs) can self-renew indefinitely in culture while retaining the potential to differentiate into virtually all normal cell types in the adult animal. Due to these remarkable properties, PSCs not only provide a superb system to investigate mammalian development and model diseases, but also hold promise for regenerative therapies. Autophagy is a self-digestive process that targets proteins, organelles, and other cellular contents for lysosomal degradation. Here, we review recent literature on the mechanistic role of different types of autophagy in embryonic development, embryonic stem cells (ESCs), and induced PSCs (iPSCs), focusing on their remodeling functions on protein, metabolism, and epigenetics. We present a perspective on unsolved issues and propose that autophagy is a promising target to modulate acquisition, maintenance, and directed differentiation of PSCs.
Collapse
Affiliation(s)
- Yi Xu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| | - Xiaolu Yang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
62
|
Wang Q, Yu P, Liu C, He X, Wang G. Mitochondrial fragmentation in liver cancer: Emerging player and promising therapeutic opportunities. Cancer Lett 2022; 549:215912. [PMID: 36103914 DOI: 10.1016/j.canlet.2022.215912] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/24/2022] [Accepted: 09/06/2022] [Indexed: 11/02/2022]
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related death worldwide. Enhanced mitochondrial fragmentation (MF) is associated with poor prognosis in HCC patients. However, its molecular mechanism in HCC remains elusive. Although enhanced MF activates effector T cells and dendritic cells, it induces immunoescape by decreasing the number and cytotoxicity of natural killer cells in the HCC immune microenvironment. Therefore, the influence of MF on the activity of different immune cells is a great challenge. Enhanced MF contributes to maintaining stemness by promoting the asymmetric division of liver cancer stem cells (LCSCs), suggesting that MF may become a potential target for HCC recurrence, metastasis, and chemotherapy resistance. Moreover, mechanistic studies suggest that MF may promote tumour progression through autophagy, oxidative stress, and metabolic reprogramming. Human-induced hepatocyte organoids are a recently developed system that can be genetically manipulated to mimic cancer initiation and identify potential preventive treatments. We can use it to screen MF-related candidate inhibitors of HCC progression and further explore the role of MF in hepatocarcinogenesis. We herein describe the mechanisms by which MF contributes to HCC development, discuss potential therapeutic approaches, and highlight the possibility that MF modulation has a synergistic effect with immunotherapy.
Collapse
Affiliation(s)
- Qian Wang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, China.
| | - Pengfei Yu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Chaoxu Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310006, China
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Gang Wang
- Department of General Surgery, The 74th Group Army Hospital, Guangzhou, 510318, China.
| |
Collapse
|
63
|
Comparative roadmaps of reprogramming and oncogenic transformation identify Bcl11b and Atoh8 as broad regulators of cellular plasticity. Nat Cell Biol 2022; 24:1350-1363. [PMID: 36075976 PMCID: PMC9481462 DOI: 10.1038/s41556-022-00986-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/27/2022] [Indexed: 12/22/2022]
Abstract
Coordinated changes of cellular plasticity and identity are critical for pluripotent reprogramming and oncogenic transformation. However, the sequences of events that orchestrate these intermingled modifications have never been comparatively dissected. Here, we deconvolute the cellular trajectories of reprogramming (via Oct4/Sox2/Klf4/c-Myc) and transformation (via Ras/c-Myc) at the single-cell resolution and reveal how the two processes intersect before they bifurcate. This approach led us to identify the transcription factor Bcl11b as a broad-range regulator of cell fate changes, as well as a pertinent marker to capture early cellular intermediates that emerge simultaneously during reprogramming and transformation. Multiomics characterization of these intermediates unveiled a c-Myc/Atoh8/Sfrp1 regulatory axis that constrains reprogramming, transformation and transdifferentiation. Mechanistically, we found that Atoh8 restrains cellular plasticity, independent of cellular identity, by binding a specific enhancer network. This study provides insights into the partitioned control of cellular plasticity and identity for both regenerative and cancer biology. Huyghe, Furlan et al. compare pluripotent reprogramming with oncogenic transformation and identify Bcl11b and Atoh8 as regulators of cellular plasticity in both processes, thus offering a unifying theory on the factors constraining cell fate changes.
Collapse
|
64
|
Vijayakumar G, Swetha US, Sudhagar S. Tamoxifen modulates mitochondrial dynamics through AMPK and MAPK during nutrition deprivation. Cell Biol Int 2022; 46:1661-1671. [PMID: 35819094 DOI: 10.1002/cbin.11853] [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: 02/14/2022] [Revised: 04/29/2022] [Accepted: 06/20/2022] [Indexed: 11/11/2022]
Abstract
The interaction of cancer cells with their tumor microenvironment determines key events in the progression of the disease, therapeutic efficacy, and the development of drug resistance. Here, we presented evidence that tamoxifen support breast cancer growth during nutrition deprivation by modulating mitochondrial dynamics through AMPK and MAPK signaling. Tamoxifen enhances mitochondrial fusion under nutrition-deprived conditions by suppressing Drp1 ser616 phosphorylation and upregulating Mfn1 levels. Tamoxifen-induced mitochondrial fusion is mediated by the activation of AMPK as evident by the pharmacological inhibition of AMPK reverse mitochondrial fusion. Interestingly, JNK activation by tamoxifen controls the mitochondrial fusion morphology by downregulating Mfn2. Collectively, tamoxifen support cell growth by enhancing mitochondrial fusion by regulating stress kinase signaling under nutrition deprivation condition.
Collapse
Affiliation(s)
- Gangipangi Vijayakumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Uppalapati S Swetha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Selvaraju Sudhagar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| |
Collapse
|
65
|
Cardiac disruption of SDHAF4-mediated mitochondrial complex II assembly promotes dilated cardiomyopathy. Nat Commun 2022; 13:3947. [PMID: 35803927 PMCID: PMC9270418 DOI: 10.1038/s41467-022-31548-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/15/2022] [Indexed: 12/30/2022] Open
Abstract
Succinate dehydrogenase, which is known as mitochondrial complex II, has proven to be a fascinating machinery, attracting renewed and increased interest in its involvement in human diseases. Herein, we find that succinate dehydrogenase assembly factor 4 (SDHAF4) is downregulated in cardiac muscle in response to pathological stresses and in diseased hearts from human patients. Cardiac loss of Sdhaf4 suppresses complex II assembly and results in subunit degradation and complex II deficiency in fetal mice. These defects are exacerbated in young adults with globally impaired metabolic capacity and activation of dynamin-related protein 1, which induces excess mitochondrial fission and mitophagy, thereby causing progressive dilated cardiomyopathy and lethal heart failure in animals. Targeting mitochondria via supplementation with fumarate or inhibiting mitochondrial fission improves mitochondrial dynamics, partially restores cardiac function and prolongs the lifespan of mutant mice. Moreover, the addition of fumarate is found to dramatically improve cardiac function in myocardial infarction mice. These findings reveal a vital role for complex II assembly in the development of dilated cardiomyopathy and provide additional insights into therapeutic interventions for heart diseases. Functional succinate dehydrogenase (SDH) complex is vital to mitochondrial homeostasis. Here the authors show that disruption of SDH assembly in the heart causes dilated cardiomyopathy via impairing the mitochondrial integrity and metabolism and that mitochondrial interventions can be an effective approach to ameliorate the disease progression.
Collapse
|
66
|
Coelho P, Fão L, Mota S, Rego AC. Mitochondrial function and dynamics in neural stem cells and neurogenesis: Implications for neurodegenerative diseases. Ageing Res Rev 2022; 80:101667. [PMID: 35714855 DOI: 10.1016/j.arr.2022.101667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/21/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
Mitochondria have been largely described as the powerhouse of the cell and recent findings demonstrate that this organelle is fundamental for neurogenesis. The mechanisms underlying neural stem cells (NSCs) maintenance and differentiation are highly regulated by both intrinsic and extrinsic factors. Mitochondrial-mediated switch from glycolysis to oxidative phosphorylation, accompanied by mitochondrial remodeling and dynamics are vital to NSCs fate. Deregulation of mitochondrial proteins, mitochondrial DNA, function, fission/fusion and metabolism underly several neurodegenerative diseases; data show that these impairments are already present in early developmental stages and NSC fate decisions. However, little is known about mitochondrial role in neurogenesis. In this Review, we describe the recent evidence covering mitochondrial role in neurogenesis, its impact in selected neurodegenerative diseases, for which aging is the major risk factor, and the recent advances in stem cell-based therapies that may alleviate neurodegenerative disorders-related neuronal deregulation through improvement of mitochondrial function and dynamics.
Collapse
Affiliation(s)
- Patrícia Coelho
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal.
| | - Lígia Fão
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| | - Sandra Mota
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; III, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - A Cristina Rego
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra Polo 1, Coimbra, Portugal; FMUC- Faculty of Medicine, University of Coimbra Polo 3, Coimbra, Portugal.
| |
Collapse
|
67
|
Basei FL, de Castro Ferezin C, Rodrigues de Oliveira AL, Muñoz JP, Zorzano A, Kobarg J. Nek4 regulates mitochondrial respiration and morphology. FEBS J 2022; 289:3262-3279. [DOI: 10.1111/febs.16343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/13/2021] [Accepted: 01/04/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Fernanda Luisa Basei
- Faculdade de Ciências Farmacêuticas Universidade Estadual de Campinas Brazil
- Institute for Research in Biomedicine (IRB Barcelona) The Barcelona Institute of Science and Technology Spain
| | - Camila de Castro Ferezin
- Faculdade de Ciências Farmacêuticas Universidade Estadual de Campinas Brazil
- Departamento de Bioquímica e Biologia Tecidual Instituto de Biologia Universidade Estadual de Campinas Brazil
| | - Ana Luisa Rodrigues de Oliveira
- Faculdade de Ciências Farmacêuticas Universidade Estadual de Campinas Brazil
- Departamento de Bioquímica e Biologia Tecidual Instituto de Biologia Universidade Estadual de Campinas Brazil
| | - Juan Pablo Muñoz
- Institute for Research in Biomedicine (IRB Barcelona) The Barcelona Institute of Science and Technology Spain
- Departament de Bioquímica i Biomedicina Molecular Facultat de Biologia Universitat de Barcelona Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) Instituto de Salud Carlos III Barcelona Spain
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona) The Barcelona Institute of Science and Technology Spain
- Departament de Bioquímica i Biomedicina Molecular Facultat de Biologia Universitat de Barcelona Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) Instituto de Salud Carlos III Barcelona Spain
| | - Jörg Kobarg
- Faculdade de Ciências Farmacêuticas Universidade Estadual de Campinas Brazil
- Departamento de Bioquímica e Biologia Tecidual Instituto de Biologia Universidade Estadual de Campinas Brazil
| |
Collapse
|
68
|
Metabolic Determinants in Cardiomyocyte Function and Heart Regenerative Strategies. Metabolites 2022; 12:metabo12060500. [PMID: 35736435 PMCID: PMC9227827 DOI: 10.3390/metabo12060500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Heart disease is the leading cause of mortality in developed countries. The associated pathology is characterized by a loss of cardiomyocytes that leads, eventually, to heart failure. In this context, several cardiac regenerative strategies have been developed, but they still lack clinical effectiveness. The mammalian neonatal heart is capable of substantial regeneration following injury, but this capacity is lost at postnatal stages when cardiomyocytes become terminally differentiated and transit to the fetal metabolic switch. Cardiomyocytes are metabolically versatile cells capable of using an array of fuel sources, and the metabolism of cardiomyocytes suffers extended reprogramming after injury. Apart from energetic sources, metabolites are emerging regulators of epigenetic programs driving cell pluripotency and differentiation. Thus, understanding the metabolic determinants that regulate cardiomyocyte maturation and function is key for unlocking future metabolic interventions for cardiac regeneration. In this review, we will discuss the emerging role of metabolism and nutrient signaling in cardiomyocyte function and repair, as well as whether exploiting this axis could potentiate current cellular regenerative strategies for the mammalian heart.
Collapse
|
69
|
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: 1.0] [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
|
70
|
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
|
71
|
Carmona-Carmona CA, Dalla Pozza E, Ambrosini G, Errico A, Dando I. Divergent Roles of Mitochondria Dynamics in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14092155. [PMID: 35565283 PMCID: PMC9105422 DOI: 10.3390/cancers14092155] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma is one of the most lethal neoplasia due to the lack of early diagnostic markers and effective therapies. The study of metabolic alterations of PDAC is of crucial importance since it would open the way to the discovery of new potential therapies. Mitochondria represent key organelles that regulate energy metabolism, and they remodel their structure by undergoing modifications by fusing with other mitochondria or dividing to generate smaller ones. The alterations of mitochondria arrangement may influence the metabolism of PDAC cells, thus supporting the proliferative needs of cancer. Shedding light on this topic regarding cancer and, more specifically, PDAC may help identify new potential strategies that hit cancer cells at their “core,” i.e., mitochondria. Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumors; it is often diagnosed at an advanced stage and is hardly treatable. These issues are strictly linked to the absence of early diagnostic markers and the low efficacy of treatment approaches. Recently, the study of the metabolic alterations in cancer cells has opened the way to important findings that can be exploited to generate new potential therapies. Within this scenario, mitochondria represent important organelles within which many essential functions are necessary for cell survival, including some key reactions involved in energy metabolism. These organelles remodel their shape by dividing or fusing themselves in response to cellular needs or stimuli. Interestingly, many authors have shown that mitochondrial dynamic equilibrium is altered in many different tumor types. However, up to now, it is not clear whether PDAC cells preferentially take advantage of fusion or fission processes since some studies reported a wide range of different results. This review described the role of both mitochondria arrangement processes, i.e., fusion and fission events, in PDAC, showing that a preference for mitochondria fragmentation could sustain tumor needs. In addition, we also highlight the importance of considering the metabolic arrangement and mitochondria assessment of cancer stem cells, which represent the most aggressive tumor cell type that has been shown to have distinctive metabolic features to that of differentiated tumor cells.
Collapse
Affiliation(s)
| | | | | | | | - Ilaria Dando
- Correspondence: (C.A.C.-C.); (I.D.); Tel.: +39-045-802-7174 (C.A.C.-C.); +39-045-802-7169 (I.D.)
| |
Collapse
|
72
|
Molugu K, Battistini GA, Heaster TM, Rouw J, Guzman EC, Skala MC, Saha K. Label-Free Imaging to Track Reprogramming of Human Somatic Cells. GEN BIOTECHNOLOGY 2022; 1:176-191. [PMID: 35586336 PMCID: PMC9092522 DOI: 10.1089/genbio.2022.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
The process of reprogramming patient samples to human-induced pluripotent stem cells (iPSCs) is stochastic, asynchronous, and inefficient, leading to a heterogeneous population of cells. In this study, we track the reprogramming status of patient-derived erythroid progenitor cells (EPCs) at the single-cell level during reprogramming with label-free live-cell imaging of cellular metabolism and nuclear morphometry to identify high-quality iPSCs. EPCs isolated from human peripheral blood of three donors were used for our proof-of-principle study. We found distinct patterns of autofluorescence lifetime for the reduced form of nicotinamide adenine dinucleotide (phosphate) and flavin adenine dinucleotide during reprogramming. Random forest models classified iPSCs with ∼95% accuracy, which enabled the successful isolation of iPSC lines from reprogramming cultures. Reprogramming trajectories resolved at the single-cell level indicated significant reprogramming heterogeneity along different branches of cell states. This combination of micropatterning, autofluorescence imaging, and machine learning provides a unique, real-time, and nondestructive method to assess the quality of iPSCs in a biomanufacturing process, which could have downstream impacts in regenerative medicine, cell/gene therapy, and disease modeling.
Collapse
Affiliation(s)
- Kaivalya Molugu
- Biophysics Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
| | - Giovanni A. Battistini
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
| | - Tiffany M. Heaster
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Jacob Rouw
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
| | | | - Melissa C. Skala
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
| |
Collapse
|
73
|
Apelin-13 Pretreatment Promotes the Cardioprotective Effect of Mesenchymal Stem Cells against Myocardial Infarction by Improving Their Survival. Stem Cells Int 2022; 2022:3742678. [PMID: 35355588 PMCID: PMC8960019 DOI: 10.1155/2022/3742678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/23/2022] Open
Abstract
Although mesenchymal stem cell- (MSC-) based therapy has shown promising results for myocardial infarction (MI), low cell survival heavily limits its beneficial effects. Apelin plays an essential regulatory role in cell proliferation. This study was aimed at determining whether Apelin-13 pretreatment could improve the survival of MSCs in the ischemic heart and enhance their cardioprotective efficacy against MI. MSCs were pretreated with or without Apelin-13 for 24 hours and then exposed to serum deprivation and hypoxia (SD/H) for 48 hours. The mitochondrial morphology of MSCs was assessed by MitoTracker staining. The apoptosis of MSCs was determined by TUNEL staining. The level of mitochondrial reactive oxygen species (ROS) of MSCs was detected by Mito-Sox staining. MSCs and Apelin-13-pretreated MSCs were transplanted into the peri-infarct region in a mouse MI model. Apelin-13 pretreatment protected MSCs against SD/H-induced mitochondrial fragmentation and apoptosis. Apelin-13 pretreatment reduced ROS generation induced by SD/H in MSCs. Furthermore, Apelin-13 pretreatment enhanced the angiogenesis of MSCs under SD/H conditions. Mechanistically, Apelin-13 pretreatment inhibited SD/H-induced MSC apoptosis by downregulating mitochondrial fission via activation of the ERK pathway, and these effects were partially abrogated by ERK inhibitor U0126. Apelin-13 pretreatment promoted the survival of MSCs in the ischemic heart. Moreover, transplantation with Apelin-13-pretreated MSCs improved heart function and increased angiogenesis accompanied by decreased fibrosis compared with MSC transplantation at 28 days following MI. These findings reveal that pretreatment with Apelin-13 improves MSCs survival and enhances their therapeutic efficacy for MI. Our study provides a novel approach to improve MSC-based therapy for cardiovascular disease.
Collapse
|
74
|
Caballano-Infantes E, Cahuana GM, Bedoya FJ, Salguero-Aranda C, Tejedo JR. The Role of Nitric Oxide in Stem Cell Biology. Antioxidants (Basel) 2022; 11:497. [PMID: 35326146 PMCID: PMC8944807 DOI: 10.3390/antiox11030497] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a gaseous biomolecule endogenously synthesized with an essential role in embryonic development and several physiological functions, such as regulating mitochondrial respiration and modulation of the immune response. The dual role of NO in embryonic stem cells (ESCs) has been previously reported, preserving pluripotency and cell survival or inducing differentiation with a dose-dependent pattern. In this line, high doses of NO have been used in vitro cultures to induce focused differentiation toward different cell lineages being a key molecule in the regenerative medicine field. Moreover, optimal conditions to promote pluripotency in vitro are essential for their use in advanced therapies. In this sense, the molecular mechanisms underlying stemness regulation by NO have been studied intensively over the current years. Recently, we have reported the role of low NO as a hypoxia-like inducer in pluripotent stem cells (PSCs), which supports using this molecule to maintain pluripotency under normoxic conditions. In this review, we stress the role of NO levels on stem cells (SCs) fate as a new approach for potential cell therapy strategies. Furthermore, we highlight the recent uses of NO in regenerative medicine due to their properties regulating SCs biology.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41092 Seville, Spain
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Javier Bedoya
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Salguero-Aranda
- Department of Pathology, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, CSIC-University of Seville, 41013 Seville, Spain;
- Spanish Biomedical Research Network Centre in Oncology-CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41004 Seville, Spain
| | - Juan R. Tejedo
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain; (G.M.C.); (F.J.B.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
75
|
Babaei-Abraki S, Karamali F, Nasr-Esfahani MH. The Role of Endoplasmic Reticulum and Mitochondria in Maintaining Redox Status and Glycolytic Metabolism in Pluripotent Stem Cells. Stem Cell Rev Rep 2022; 18:1789-1808. [PMID: 35141862 DOI: 10.1007/s12015-022-10338-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
Abstract
Pluripotent stem cells (PSCs), including embryonic stem cells and induced pluripotent stem cells (iPSCs), can be applicable for regenerative medicine. They strangely rely on glycolysis metabolism akin to aerobic glycolysis in cancer cells. Upon differentiation, PSCs undergo a metabolic shift from glycolysis to oxidative phosphorylation (OXPHOS). The metabolic shift depends on organelles maturation, transcriptome modification, and metabolic switching. Besides, metabolism-driven chromatin regulation is necessary for cell survival, self-renewal, proliferation, senescence, and differentiation. In this respect, mitochondria may serve as key organelle to adapt environmental changes with metabolic intermediates which are necessary for maintaining PSCs identity. The endoplasmic reticulum (ER) is another organelle whose role in cellular identity remains under-explored. The purpose of our article is to highlight the recent progress on these two organelles' role in maintaining PSCs redox status focusing on metabolism. Topics include redox status, metabolism regulation, mitochondrial dynamics, and ER stress in PSCs. They relate to the maintenance of stem cell properties and subsequent differentiation of stem cells into specific cell types.
Collapse
Affiliation(s)
- Shahnaz Babaei-Abraki
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.,Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Fereshteh Karamali
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
76
|
Ansari MY, Novak K, Haqqi TM. ERK1/2-mediated activation of DRP1 regulates mitochondrial dynamics and apoptosis in chondrocytes. Osteoarthritis Cartilage 2022; 30:315-328. [PMID: 34767958 PMCID: PMC8792336 DOI: 10.1016/j.joca.2021.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/19/2021] [Accepted: 11/01/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine the Dynamin-related protein 1 (DRP1) regulation of mitochondrial fission in chondrocytes under pathological conditions, an area which is underexplored in osteoarthritis pathogenesis. DESIGN DRP1 protein expression was determined by immunohistochemistry (IHC) or immunofluorescence (IF) staining of cartilage sections. IL-1β-induced DRP1 mRNA expression in chondrocytes was quantified by qPCR and protein expression by immunoblotting. Mitochondrial fragmentation in chondrocytes was visualized by MitoTracker staining or IF staining of mitochondrial marker proteins or by transient expression of mitoDsRed. Mitochondrial reactive oxygen species (ROS) levels were determined by MitoSOX staining. Apoptosis was determined by lactate dehydrogenase (LDH) release assay, Caspase 3/7 activity assay, propidium iodide (PI), and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and IF staining of cleaved caspase 3. Cytochrome c release was determined by confocal microscopy. Surgical destabilization of the medial meniscus (DMM) was used to induce osteoarthritis (OA) in mice. RESULTS Expression of DRP1 and mitochondrial damage was high in human OA cartilage and in the joints of mice subjected to DMM surgery which also showed increased chondrocytes apoptosis. IL-1β-induced mitochondrial network fragmentation and chondrocyte apoptosis via modulation of DRP1 expression and activity and induce apoptosis via Bax-mediated release of Cytochrome c. Pharmacological inhibition of DRP1 activity by Mdivi-1 blocked IL-1β-induced mitochondrial damage and apoptosis in chondrocytes. Additionally, IL-1β-induced activation of extracellular signal-regulated kinase 1/2 (ERK1/2) is crucial for DRP1 activation and induction of mitochondrial network fragmentation in chondrocytes as these were blocked by inhibiting ERK1/2 activation. CONCLUSIONS These findings demonstrate that ERK1/2 is a critical player in DRP1-mediated induction of mitochondrial fission and apoptosis in IL-1β-stimulated chondrocytes.
Collapse
Affiliation(s)
- Mohammad Y. Ansari
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, USA, 44272
| | - Kimberly Novak
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, USA, 44272
| | - Tariq M. Haqqi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, Ohio, USA, 44272,Corresponding author: Telephone number: +1 330 325 6704, TMH:
| |
Collapse
|
77
|
Cui G, Xu Y, Cao S, Shi K. Inducing somatic cells into pluripotent stem cells is an important platform to study the mechanism of early embryonic development. Mol Reprod Dev 2022; 89:70-85. [PMID: 35075695 DOI: 10.1002/mrd.23559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/16/2021] [Accepted: 01/10/2022] [Indexed: 01/24/2023]
Abstract
The early embryonic development starts with the totipotent zygote upon fertilization of differentiated sperm and egg, which undergoes a range of reprogramming and transformation to acquire pluripotency. Induced pluripotent stem cells (iPSCs), a nonclonal technique to produce stem cells, are originated from differentiated somatic cells via accomplishment of cell reprogramming, which shares common reprogramming process with early embryonic development. iPSCs are attractive in recent years due to the potentially significant applications in disease modeling, potential value in genetic improvement of husbandry animal, regenerative medicine, and drug screening. This review focuses on introducing the research advance of both somatic cell reprogramming and early embryonic development, indicating that the mechanisms of iPSCs also shares common features with that of early embryonic development in several aspects, such as germ cell factors, DNA methylation, histone modification, and/or X chromosome inactivation. As iPSCs can successfully avoid ethical concerns that are naturally present in the embryos and/or embryonic stem cells, the practicality of somatic cell reprogramming (iPSCs) could provide an insightful platform to elucidate the mechanisms underlying the early embryonic development.
Collapse
Affiliation(s)
- Guina Cui
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
| | - Yanwen Xu
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
| | - Shuyuan Cao
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
| | - Kerong Shi
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
78
|
Degitz C, Reime S, Thews O. Effect of Acidosis-Induced Signalling Pathways on Mitochondrial O2 Consumption of Tumour Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1395:231-235. [PMID: 36527642 DOI: 10.1007/978-3-031-14190-4_38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Signalling pathways such as ERK1/2, p38 or PI3K are activated in tumour cells by extracellular acidosis, which is a common phenomenon in human tumours. These signalling pathways can modulate the mitochondrial function and activity. The aim of the study was to evaluate the impact of extracellular acidosis on the mitochondrial O2 consumption and, in consequence, the potential role of ERK1/2, p38 and PI3K cascades on modulating the respiratory function. The O2 consumption rate (OCR) was measured at pH 7.4 and extracellular acidosis (pH 6.6) in combination with inhibition of the respective signalling pathway. The activity of the pathways was determined by phosphorylation-specific western blot using the cytosolic and mitochondrial fraction of cell lysates. The experiments were performed on a rat tumour cell line (subline AT1 of the rat R-3327 prostate carcinoma) and normal cells (NRK-49F fibroblasts). Acidosis increased the OCR of AT1 cells, especially the basal OCR and the O2 consumption, which is related to ATP production. In normal NRKF cells OCR was unaffected by low pH. Inhibition of ERK1/2 and PI3K, but not p38, reduced the acidosis-induced increase of the OCR significantly in AT1 tumour cells. In this cell line acidosis also led to an ERK1/2 and PI3K activation, predominantly in the mitochondrial fraction. These results indicate that extracellular acidosis activates cellular respiration in tumour cells, presumably by activating the ERK1/2 and/or the PI3K signalling cascade. This activation of ERK1/2 and PI3K is located primarily in the mitochondrial compartment of the cells.
Collapse
|
79
|
MITOCHONDRIA: Mitochondrial dynamics in the regulation of stem cells. Int J Biochem Cell Biol 2022; 144:106158. [DOI: 10.1016/j.biocel.2022.106158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/24/2022]
|
80
|
Pavlyuchenkova AN, Zinovkin RA, Makievskaya CI, Galkin II, Chelombitko MA. Mitochondria-targeted triphenylphosphonium-based compounds inhibit FcεRI-dependent degranulation of mast cells by preventing mitochondrial dysfunction through Erk1/2. Life Sci 2022; 288:120174. [PMID: 34826439 DOI: 10.1016/j.lfs.2021.120174] [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: 10/12/2021] [Revised: 11/11/2021] [Accepted: 11/19/2021] [Indexed: 11/27/2022]
Abstract
AIMS FcεRI-dependent activation and degranulation of mast cells (MC) play an important role in allergic diseases. We have previously demonstrated that triphenylphosphonium (TPP)-based antioxidant SkQ1 inhibits mast cell degranulation, but the exact mechanism of this inhibition is still unknown. This study focused on investigating the influence of TPP-based compounds SkQ1 and C12TPP on FcεRI-dependent mitochondrial dysfunction and signaling during MC degranulation. MAIN METHODS MC were sensitized by anti-dinitrophenyl IgE and stimulated by BSA-conjugated dinitrophenyl. The degranulation of MC was estimated by β-hexosaminidase release. The effect of TPP-based compounds on FcεRI-dependent signaling was determined by Western blot analysis for adapter molecule LAT, kinases Syk, PI3K, Erk1/2, and p38. Fluorescent microscopy was used to evaluate mitochondrial parameters such as morphology, membrane potential, reactive oxygen species and ATP level. KEY FINDINGS Pretreatment with TPP-based compounds significantly decreased FcεRI-dependent degranulation of MC. TPP-based compounds also prevented mitochondrial dysfunction (drop in mitochondrial ATP level and mitochondrial fission), and decreased Erk1/2 kinase phosphorylation. Selective Erk1/2 inhibition by U0126 also reduced β-hexosaminidase release and prevented mitochondrial fragmentation during FcεRI-dependent degranulation of MC. SIGNIFICANCE These findings expand the fundamental understanding of the role of mitochondria in the activation of MC. It also contributes to the rationale for the development of mitochondrial-targeted drugs for the treatment of allergic diseases.
Collapse
Affiliation(s)
| | - Roman A Zinovkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Ciara I Makievskaya
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Ivan I Galkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Maria A Chelombitko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
81
|
Wang D, Tian J, Yan Z, Yuan Q, Wu D, Liu X, Yang S, Guo S, Wang J, Yang Y, Xing J, An J, Huang Q. Mitochondrial fragmentation is crucial for c-Myc-driven hepatoblastoma-like liver tumor. Mol Ther 2022; 30:1645-1660. [PMID: 35085814 PMCID: PMC9077476 DOI: 10.1016/j.ymthe.2022.01.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/19/2021] [Accepted: 01/20/2022] [Indexed: 11/26/2022] Open
Abstract
Hepatoblastoma is the most common liver cancer in children, and the aggressive subtype often has a poor prognosis and lacks effective targeted therapy. Although aggressive hepatoblastoma (HB) is often accompanied by abnormally high expression of the transcription factor c-Myc, the underlying mechanism remains unclear. In this study, we found that mitochondrial fragmentation was enhanced by c-Myc overexpression in human aggressive HB tissues and was associated with poor prognosis. Then, a mouse model resembling human HB was established via hydrodynamic injection of c-Myc plasmids. We observed that liver-specific knockout of the mitochondrial fusion molecule MFN1 or overexpression of mitochondrial fission molecule DRP1 promoted the occurrence of c-Myc-driven liver cancer. In contrast, when MFN1 was overexpressed in the liver, tumor formation was delayed. In vitro experiments showed that c-Myc transcriptionally upregulated the expression of DRP1 and decreased MFN1 expression through upregulation of miR-373-3p. Moreover, enhanced mitochondrial fragmentation significantly promoted aerobic glycolysis and the proliferation of HB cells by significantly increasing reactive oxygen species (ROS) production and activating the RAC-alpha serine/threonine-protein kinase (AKT)/mammalian target of rapamycin (mTOR) and nuclear factor κB (NF-κB) pathways. Taken together, our results indicate that c-Myc-mediated mitochondrial fragmentation promotes the malignant transformation and progression of HB by activating ROS-mediated multi-oncogenic signaling.
Collapse
|
82
|
Regulation of mitochondrial morphology and metabolism by Jak-STAT pathway. JOURNAL OF ANIMAL REPRODUCTION AND BIOTECHNOLOGY 2021. [DOI: 10.12750/jarb.36.4.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
83
|
Cha Y, Kim T, Jeon J, Jang Y, Kim PB, Lopes C, Leblanc P, Cohen BM, Kim KS. SIRT2 regulates mitochondrial dynamics and reprogramming via MEK1-ERK-DRP1 and AKT1-DRP1 axes. Cell Rep 2021; 37:110155. [PMID: 34965411 PMCID: PMC8780843 DOI: 10.1016/j.celrep.2021.110155] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/01/2021] [Accepted: 12/01/2021] [Indexed: 02/08/2023] Open
Abstract
During somatic reprogramming, cellular energy metabolism fundamentally switches from predominantly mitochondrial oxidative phosphorylation toward glycolysis. This metabolic reprogramming, also called the Warburg effect, is critical for the induction of pluripotency, but its molecular mechanisms remain poorly defined. Notably, SIRT2 is consistently downregulated during the reprogramming process and regulates glycolytic switch. Here, we report that downregulation of SIRT2 increases acetylation of mitogen-activated protein kinase (MAPK) kinase-1 (MEK1) at Lys175, resulting in activation of extracellular signal-regulated kinases (ERKs) and subsequent activation of the pro-fission factor dynamin-related protein 1 (DRP1). In parallel, downregulation of SIRT2 hyperacetylates the serine/threonine protein kinase AKT1 at Lys20 in a non-canonical way, activating DRP1 and metabolic reprogramming. Together, our study identified two axes, SIRT2-MEK1-ERK-DRP1 and SIRT2-AKT1-DRP1, that critically link mitochondrial dynamics and oxidative phosphorylation to the somatic reprogramming process. These upstream signals, together with SIRT2’s role in glycolytic switching, may underlie the Warburg effect observed in human somatic cell reprogramming. Mitochondrial remodeling has critical roles for the somatic cell reprogramming process. Cha et al. report the functional role of SIRT2 in mitochondrial dynamics and remodeling during the human somatic cell reprogramming process. They identify two axes, SIRT2-MEK1-ERK-DRP1 and SIRT2-AKT1-DRP1, that link SIRT2 downregulation to mitochondrial remodeling and somatic cell reprogramming.
Collapse
Affiliation(s)
- Young Cha
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA.
| | - Taewoo Kim
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Jeha Jeon
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Yongwoo Jang
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA; Department of Biomedical Engineering, Hanyang University, Seoul 04763, Korea
| | - Patrick B Kim
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Claudia Lopes
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Pierre Leblanc
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Bruce M Cohen
- Department of Psychiatry and Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Kwang-Soo Kim
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA.
| |
Collapse
|
84
|
Zhang Y, Zheng W, Ren P, Hu H, Tong X, Zhang S, Li X, Wang H, Jiang J, Jin J, Yang W, Cao L, He Y, Ma Y, Zhang Y, Gu Y, Hu L, Luo K, Gong F, Lu G, Lin G, Fan H, Zhang S. Biallelic mutations in MOS cause female infertility characterized by human early embryonic arrest and fragmentation. EMBO Mol Med 2021; 13:e14887. [PMID: 34779126 PMCID: PMC8649871 DOI: 10.15252/emmm.202114887] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 01/26/2023] Open
Abstract
Early embryonic arrest and fragmentation (EEAF) is a common phenomenon leading to female infertility, but the genetic determinants remain largely unknown. The Moloney sarcoma oncogene (MOS) encodes a serine/threonine kinase that activates the ERK signaling cascade during oocyte maturation in vertebrates. Here, we identified four rare variants of MOS in three infertile female individuals with EEAF that followed a recessive inheritance pattern. These MOS variants encoded proteins that resulted in decreased phosphorylated ERK1/2 level in cells and oocytes, and displayed attenuated rescuing effects on cortical F-actin assembly. Using oocyte-specific Erk1/2 knockout mice, we verified that MOS-ERK signal pathway inactivation in oocytes caused EEAF as human. The RNA sequencing data revealed that maternal mRNA clearance was disrupted in human mature oocytes either with MOS homozygous variant or with U0126 treatment, especially genes relative to mitochondrial function. Mitochondrial dysfunction was observed in oocytes with ERK1/2 deficiency or inactivation. In conclusion, this study not only uncovers biallelic MOS variants causes EEAF but also demonstrates that MOS-ERK signaling pathway drives human oocyte cytoplasmic maturation to prevent EEAF.
Collapse
Affiliation(s)
- Yin‐Li Zhang
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Wei Zheng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Peipei Ren
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Huiling Hu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Xiaomei Tong
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Shuo‐Ping Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Xiang Li
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Haichao Wang
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | | | - Jiamin Jin
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Weijie Yang
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Lanrui Cao
- Life Sciences InstituteZhejiang UniversityHangzhouChina
| | - Yuanlin He
- Department of EpidemiologyCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Yerong Ma
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Yingyi Zhang
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| | - Yifan Gu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Liang Hu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Keli Luo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Fei Gong
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Guang‐Xiu Lu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC‐XiangyaChangshaChina
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive EngineeringCentral South UniversityChangshaChina
| | - Heng‐Yu Fan
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
- Life Sciences InstituteZhejiang UniversityHangzhouChina
| | - Songying Zhang
- Assisted Reproduction UnitDepartment of Obstetrics and GynecologySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
85
|
Chang YW, Song ZH, Chen CC. FAK regulates cardiomyocyte mitochondrial fission and function through Drp1. FEBS J 2021; 289:1897-1910. [PMID: 34739186 DOI: 10.1111/febs.16263] [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: 03/04/2021] [Revised: 09/19/2021] [Accepted: 11/03/2021] [Indexed: 11/29/2022]
Abstract
Loss of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1) in cardiomyocytes results in energy shortage and heart failure. We aim to understand the intracellular signal pathway and extracellular factors regulating Drp1 phosphorylation and mitochondrial morphology and function in cardiomyocytes. We found cyclic mechanical stretching induced mitochondrial fission through Drp1 and focal adhesion kinase (FAK) in neonatal rat ventricular myocytes (NRVMs). FAK regulated phosphorylation of Drp1 and mitochondrial Drp1 levels. Extracellular fibronectin activated Drp1 and caused mitochondrial fission through FAK and extracellular signal-regulated kinase 1/2 (ERK1/2). Fibronectin increased NRVMs oxygen consumption rate and ATP content via FAK-ERK1/2-Drp1. Inhibition of the FAK-ERK1/2-Drp1 pathway caused cellular energy shortage. In addition, the FAK-ERK1/2-Drp1 pathway was rapidly activated by adrenergic agonists and contributed to agonists-stimulated NRVMs respiration. Interestingly, fibronectin limited the adrenergic agonists-induced NRVMs respiration by restricting phosphorylation of Drp1. Our results suggest that extracellular fibronectin and adrenergic stimulations use the FAK-ERK1/2-Drp1 pathway to regulate mitochondrial morphology and function in cardiomyocytes.
Collapse
Affiliation(s)
- Yu-Wang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | - Zong-Han Song
- Institute of Biomedical Sciences, Academia Sinica, Taiwan
| | | |
Collapse
|
86
|
Bakare AB, Meshrkey F, Lowe B, Molder C, Rao RR, Zhan J, Iyer S. MitoCellPhe reveals mitochondrial morphologies in single fibroblasts and clustered stem cells. Am J Physiol Cell Physiol 2021; 321:C735-C748. [PMID: 34469204 PMCID: PMC8560386 DOI: 10.1152/ajpcell.00231.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 08/16/2021] [Indexed: 11/22/2022]
Abstract
Mitochondria are dynamic organelles that differ significantly in their morphologies across cell types, reflecting specific cellular needs and stages in development. Despite the wide biological significance in disease and in health, delineating mitochondrial morphologies in complex systems remains challenging. Here, we present the Mitochondrial Cellular Phenotype (MitoCellPhe) tool developed for quantifying mitochondrial morphologies and demonstrate its utility in delineating differences in mitochondrial morphologies in a human fibroblast and human induced pluripotent stem cell (hiPSC) line. MitoCellPhe generates 24 parameters, allowing for a comprehensive analysis of mitochondrial structures and importantly allows for quantification to be performed on mitochondria in images containing single cells or clusters of cells. With this tool, we were able to validate previous findings that show networks of mitochondria in healthy fibroblast cell lines and a more fragmented morphology in hiPSCs. Using images generated from control and diseased fibroblasts and hiPSCs, we also demonstrate the efficacy of the toolset in delineating differences in morphologies between healthy and the diseased state in both stem cell (hiPSC) and differentiated fibroblast cells. Our results demonstrate that MitoCellPhe enables high-throughput, sensitive, detailed, and quantitative mitochondrial morphological assessment and thus enables better biological insights into mitochondrial dynamics in health and disease.
Collapse
Affiliation(s)
- Ajibola B Bakare
- Department of Biological Sciences, J. William Fulbright College of Arts and Sciences, University of Arkansas, Fayetteville, Arkansas
| | - Fibi Meshrkey
- Department of Biological Sciences, J. William Fulbright College of Arts and Sciences, University of Arkansas, Fayetteville, Arkansas
- Department of Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Benjamin Lowe
- Department of Computer Science and Computer Engineering, College of Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Carson Molder
- Department of Computer Science and Computer Engineering, College of Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Raj R Rao
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Justin Zhan
- Department of Computer Science and Computer Engineering, College of Engineering, University of Arkansas, Fayetteville, Arkansas
| | - Shilpa Iyer
- Department of Biological Sciences, J. William Fulbright College of Arts and Sciences, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|
87
|
Spurlock B, Parker D, Basu MK, Hjelmeland A, GC S, Liu S, Siegal GP, Gunter A, Moran A, Mitra K. Fine-tuned repression of Drp1-driven mitochondrial fission primes a 'stem/progenitor-like state' to support neoplastic transformation. eLife 2021; 10:e68394. [PMID: 34545812 PMCID: PMC8497058 DOI: 10.7554/elife.68394] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Gene knockout of the master regulator of mitochondrial fission, Drp1, prevents neoplastic transformation. Also, mitochondrial fission and its opposing process of mitochondrial fusion are emerging as crucial regulators of stemness. Intriguingly, stem/progenitor cells maintaining repressed mitochondrial fission are primed for self-renewal and proliferation. Using our newly derived carcinogen transformed human cell model, we demonstrate that fine-tuned Drp1 repression primes a slow cycling 'stem/progenitor-like state', which is characterized by small networks of fused mitochondria and a gene-expression profile with elevated functional stem/progenitor markers (Krt15, Sox2 etc) and their regulators (Cyclin E). Fine tuning Drp1 protein by reducing its activating phosphorylation sustains the neoplastic stem/progenitor cell markers. Whereas, fine-tuned reduction of Drp1 protein maintains the characteristic mitochondrial shape and gene-expression of the primed 'stem/progenitor-like state' to accelerate neoplastic transformation, and more complete reduction of Drp1 protein prevents it. Therefore, our data highlights a 'goldilocks' level of Drp1 repression supporting stem/progenitor state dependent neoplastic transformation.
Collapse
Affiliation(s)
- Brian Spurlock
- Department of Genetics, University of Alabama at BirminghamBirminghamUnited States
| | - Danitra Parker
- Department of Genetics, University of Alabama at BirminghamBirminghamUnited States
| | - Malay Kumar Basu
- Departments of Pathology, University of Alabama at BirminghamBirminghamUnited States
| | - Anita Hjelmeland
- Department of Cell Development and Integrative Biology, University of Alabama at BirminghamBirminghamUnited States
| | - Sajina GC
- Department of Cell Development and Integrative Biology, University of Alabama at BirminghamBirminghamUnited States
| | - Shanrun Liu
- Department of Genetics, University of Alabama at BirminghamBirminghamUnited States
| | - Gene P Siegal
- Departments of Pathology, Surgery, Genetics and Cell and Developmental Biology, University of Alabama at BirminghamBirminghamUnited States
| | - Alan Gunter
- Department of Genetics, University of Alabama at BirminghamBirminghamUnited States
| | - Aida Moran
- Department of Genetics, University of Alabama at BirminghamBirminghamUnited States
| | - Kasturi Mitra
- Department of Genetics, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
88
|
Oxygen as a Master Regulator of Human Pluripotent Stem Cell Function and Metabolism. J Pers Med 2021; 11:jpm11090905. [PMID: 34575682 PMCID: PMC8466012 DOI: 10.3390/jpm11090905] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) offer numerous possibilities in science and medicine, particularly when combined with precise genome editing methods. hiPSCs are artificially generated equivalents of human embryonic stem cells (hESCs), which possess an unlimited ability to self-renew and the potential to differentiate into any cell type of the human body. Importantly, generating patient-specific hiPSCs enables personalized drug testing or autologous cell therapy upon differentiation into a desired cell line. However, to ensure the highest standard of hiPSC-based biomedical products, their safety and reliability need to be proved. One of the key factors influencing human pluripotent stem cell (hPSC) characteristics and function is oxygen concentration in their microenvironment. In recent years, emerging data have pointed toward the beneficial effect of low oxygen pressure (hypoxia) on both hiPSCs and hESCs. In this review, we examine the state-of-the-art research on the oxygen impact on hiPSC functions and activity with an emphasis on their niche, metabolic state, reprogramming efficiency, and differentiation potential. We also discuss the similarities and differences between PSCs and cancer stem cells (CSCs) with respect to the role of oxygen in both cell types.
Collapse
|
89
|
Zhu Y, Kuang L, Wu Y, Deng H, She H, Zhou Y, Zhang J, Liu L, Li T. Protective Effects of Inhibition of Mitochondrial Fission on Organ Function After Sepsis. Front Pharmacol 2021; 12:712489. [PMID: 34566637 PMCID: PMC8457550 DOI: 10.3389/fphar.2021.712489] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023] Open
Abstract
Sepsis-associated organ dysfunction plays a critical role in its high mortality, mainly in connection with mitochondrial dysfunction. Whether the inhibition of mitochondrial fission is beneficial to sepsis-related organ dysfunction and underlying mechanisms are unknown. Cecal ligation and puncture induced sepsis in rats and dynamic related protein 1 knockout mice, lipopolysaccharide-treated vascular smooth muscle cells and cardiomyocytes, were used to explore the effects of inhibition of mitochondrial fission and specific mechanisms. Our study showed that mitochondrial fission inhibitor Mdivi-1 could antagonize sepsis-induced organ dysfunction including heart, vascular smooth muscle, liver, kidney, and intestinal functions, and prolonged animal survival. The further study showed that mitochondrial functions such as mitochondrial membrane potential, adenosine-triphosphate contents, reactive oxygen species, superoxide dismutase and malonaldehyde were recovered after Mdivi-1 administration via improving mitochondrial morphology. And sepsis-induced inflammation and apoptosis in heart and vascular smooth muscle were alleviated through inhibition of mitochondrial fission and mitochondrial function improvement. The parameter trends in lipopolysaccharide-stimulated cardiomyocytes and vascular smooth muscle cells were similar in vivo. Dynamic related protein 1 knockout preserved sepsis-induced organ dysfunction, and the animal survival was prolonged. Taken together, this finding provides a novel effective candidate therapy for severe sepsis/septic shock and other critical clinical diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Shock and Transfusion Department, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
90
|
HSPA9/Mortalin mediates axo-protection and modulates mitochondrial dynamics in neurons. Sci Rep 2021; 11:17705. [PMID: 34489498 PMCID: PMC8421332 DOI: 10.1038/s41598-021-97162-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 08/23/2021] [Indexed: 12/21/2022] Open
Abstract
Mortalin is a mitochondrial chaperone protein involved in quality control of proteins imported into the mitochondrial matrix, which was recently described as a sensor of neuronal stress. Mortalin is down-regulated in neurons of patients with neurodegenerative diseases and levels of Mortalin expression are correlated with neuronal fate in animal models of Alzheimer's disease or cerebral ischemia. To date, however, the links between Mortalin levels, its impact on mitochondrial function and morphology and, ultimately, the initiation of neurodegeneration, are still unclear. In the present study, we used lentiviral vectors to over- or under-express Mortalin in primary neuronal cultures. We first analyzed the early events of neurodegeneration in the axonal compartment, using oriented neuronal cultures grown in microfluidic-based devices. We observed that Mortalin down-regulation induced mitochondrial fragmentation and axonal damage, whereas its over-expression conferred protection against axonal degeneration mediated by rotenone exposure. We next demonstrated that Mortalin levels modulated mitochondrial morphology by acting on DRP1 phosphorylation, thereby further illustrating the crucial implication of mitochondrial dynamics on neuronal fate in degenerative diseases.
Collapse
|
91
|
Prieto J, García-Cañaveras JC, León M, Sendra R, Ponsoda X, Izpisúa Belmonte JC, Lahoz A, Torres J. c-MYC Triggers Lipid Remodelling During Early Somatic Cell Reprogramming to Pluripotency. Stem Cell Rev Rep 2021; 17:2245-2261. [PMID: 34476741 PMCID: PMC8599373 DOI: 10.1007/s12015-021-10239-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 11/30/2022]
Abstract
Metabolic rewiring and mitochondrial dynamics remodelling are hallmarks of cell reprogramming, but the roles of the reprogramming factors in these changes are not fully understood. Here we show that c-MYC induces biosynthesis of fatty acids and increases the rate of pentose phosphate pathway. Time-course profiling of fatty acids and complex lipids during cell reprogramming using lipidomics revealed a profound remodelling of the lipid content, as well as the saturation and length of their acyl chains, in a c-MYC-dependent manner. Pluripotent cells displayed abundant cardiolipins and scarce phosphatidylcholines, with a prevalence of monounsaturated acyl chains. Cells undergoing cell reprogramming showed an increase in mitochondrial membrane potential that paralleled that of mitochondrial-specific cardiolipins. We conclude that c-MYC controls the rewiring of somatic cell metabolism early in cell reprogramming by orchestrating cell proliferation, synthesis of macromolecular components and lipid remodelling, all necessary processes for a successful phenotypic transition to pluripotency.
Collapse
Affiliation(s)
- Javier Prieto
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, 46100, Burjassot, Valencia, Spain.,Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | | | - Marian León
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, 46100, Burjassot, Valencia, Spain
| | - Ramón Sendra
- Departamento de Bioquímica y Biología Molecular, Universitat de València, 46100, Burjassot, Valencia, Spain
| | - Xavier Ponsoda
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, 46100, Burjassot, Valencia, Spain
| | | | - Agustín Lahoz
- Biomarkers and Precision Medicine Unit, Instituto de Investigación Sanitaria La Fe, 46026, Valencia, Spain.
| | - Josema Torres
- Departamento Biología Celular, Biología Funcional y Antropología Física, Universitat de València, 46100, Burjassot, Valencia, Spain. .,Instituto de Investigación Sanitaria (INCLIVA), 46010, Valencia, Spain.
| |
Collapse
|
92
|
Caballano-Infantes E, Díaz I, Hitos AB, Cahuana GM, Martínez-Ruiz A, Soria-Juan B, Rodríguez-Griñolo R, Hmadcha A, Martín F, Soria B, Tejedo JR, Bedoya FJ. Stemness of Human Pluripotent Cells: Hypoxia-Like Response Induced by Low Nitric Oxide. Antioxidants (Basel) 2021; 10:antiox10091408. [PMID: 34573040 PMCID: PMC8472328 DOI: 10.3390/antiox10091408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022] Open
Abstract
The optimization of conditions to promote the stemness of pluripotent cells in vitro is instrumental for their use in advanced therapies. We show here that exposure of human iPSCs and human ESCs to low concentrations of the chemical NO donor DETA/NO leads to stabilization of hypoxia-inducible factors (HIF-1α and HIF-2α) under normoxia, with this effect being dependent on diminished Pro 402 hydroxylation and decreased degradation by the proteasome. Moreover, the master genes of pluripotency, NANOG and OCT-4, were upregulated. NO also induces a shift in the metabolic profile of PSCs, with an increased expression of hypoxia response genes in glycolysis. Furthermore, a reduction in the mitochondrial membrane potential with lower oxygen consumption and increased expression of mitochondrial fusion regulators, such as DRP1, was observed. The results reported here indicate that NO mimics hypoxia response in human PSCs and enhances their stemness properties when cultured under normoxic conditions.
Collapse
Affiliation(s)
- Estefanía Caballano-Infantes
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Correspondence: (E.C.-I.); (F.J.B.)
| | - Irene Díaz
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Ana Belén Hitos
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Gladys Margot Cahuana
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28009 Madrid, Spain;
| | | | - Rosario Rodríguez-Griñolo
- Departamento de Economía, Métodos Cuantitativo e Historia Económica, Universidad Pablo de Olavide, 41013 Seville, Spain;
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Franz Martín
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Bernat Soria
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
- ISABIAL and Institute of Bioengineering, University Miguel Hernández de Elche, 03010 Alicante, Spain
| | - Juan R. Tejedo
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
| | - Francisco Javier Bedoya
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, 41013 Seville, Spain; (I.D.); (A.B.H.); (A.H.); (F.M.); (J.R.T.)
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain;
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM, Instituto de Salud Carlos III, 08036 Madrid, Spain;
- Correspondence: (E.C.-I.); (F.J.B.)
| |
Collapse
|
93
|
Yuan Y, Chen J, Ge X, Deng J, Xu X, Zhao Y, Wang H. Activation of ERK-Drp1 signaling promotes hypoxia-induced Aβ accumulation by upregulating mitochondrial fission and BACE1 activity. FEBS Open Bio 2021; 11:2740-2755. [PMID: 34403210 PMCID: PMC8487051 DOI: 10.1002/2211-5463.13273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/29/2021] [Accepted: 08/16/2021] [Indexed: 12/20/2022] Open
Abstract
Hypoxia is a risk factor for Alzheimer's disease (AD). Besides, mitochondrial fission is increased in response to hypoxia. In this study, we sought to investigate whether hypoxia‐induced mitochondrial fission plays a critical role in regulating amyloid‐β (Aβ) production. Hypoxia significantly activated extracellular signal‐regulated kinase (ERK), increased phosphorylation of dynamin‐related protein 1 (Drp1) at serine 616, and decreased phosphorylation of Drp1 at serine 637. Importantly, hypoxia triggered mitochondrial dysfunction, elevated β‐secretase 1 (BACE1) and γ‐secretase activities, and promoted Aβ accumulation in HEK293 cells transfected with β‐amyloid precursor protein (APP) plasmid harboring the Swedish and Indiana familial Alzheimer's disease mutations (APPSwe/Ind HEK293 cells). Then, we investigated whether the ERK inhibitor PD325901 and Drp1 inhibitor mitochondrial division inhibitor‐1 (Mdivi‐1) would attenuate hypoxia‐induced mitochondrial fission and Aβ generation in APPSwe/Ind HEK293 cells. PD325901 and Mdivi‐1 inhibited phosphorylation of Drp1 at serine 616, resulting in reduced mitochondrial fission under hypoxia. Furthermore, hypoxia‐induced mitochondrial dysfunction, BACE1 activation, and Aβ accumulation were downregulated by PD325901 and Mdivi‐1. Our data demonstrate that hypoxia induces mitochondrial fission, impairs mitochondrial function, and facilitates Aβ generation. The ERK–Drp1 signaling pathway is partly involved in the hypoxia‐induced Aβ generation by regulating mitochondrial fission and BACE1 activity. Therefore, inhibition of hypoxia‐induced mitochondrial fission may prevent or slow the progression of AD.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Jingjiong Chen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Xuhua Ge
- Department of General Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiangshan Deng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Xiaofeng Xu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| | - Hongmei Wang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China
| |
Collapse
|
94
|
Jacobs RA, Aboouf MA, Koester-Hegmann C, Muttathukunnel P, Laouafa S, Arias-Reyes C, Thiersch M, Soliz J, Gassmann M, Schneider Gasser EM. Erythropoietin promotes hippocampal mitochondrial function and enhances cognition in mice. Commun Biol 2021; 4:938. [PMID: 34354241 PMCID: PMC8342552 DOI: 10.1038/s42003-021-02465-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 07/19/2021] [Indexed: 11/22/2022] Open
Abstract
Erythropoietin (EPO) improves neuronal mitochondrial function and cognition in adults after brain injury and in those afflicted by psychiatric disorders. However, the influence of EPO on mitochondria and cognition during development remains unexplored. We previously observed that EPO stimulates hippocampal-specific neuronal maturation and synaptogenesis early in postnatal development in mice. Here we show that EPO promotes mitochondrial respiration in developing postnatal hippocampus by increasing mitochondrial content and enhancing cellular respiratory potential. Ultrastructurally, mitochondria profiles and total vesicle content were greater in presynaptic axon terminals, suggesting that EPO enhances oxidative metabolism and synaptic transmission capabilities. Behavioural tests of hippocampus-dependent memory at early adulthood, showed that EPO improves spatial and short-term memory. Collectively, we identify a role for EPO in the murine postnatal hippocampus by promoting mitochondrial function throughout early postnatal development, which corresponds to enhanced cognition by early adulthood. Robert Jacobs, Mostafa Aboouf, et al. examined the effect of erythropoietin (EPO) in hippocampal mitochondrial function and memory in two mouse models: one overexpressing EPO in the brain, and juvenile mice treated during three days with a high dose of intraperitoneal EPO. Their results suggest that erythropoietin in the neonatal brain may impact spatial memory by increasing mitochondrial content.
Collapse
Affiliation(s)
- Robert A Jacobs
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Department of Human Physiology & Nutrition, University of Colorado, Colorado Springs, CO, USA
| | - Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland.,Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Christina Koester-Hegmann
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Paola Muttathukunnel
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland
| | - Sofien Laouafa
- Faculty of Medicine, Centre Hospitalier Universitaire de Québec (CHUQ), Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Christian Arias-Reyes
- Faculty of Medicine, Centre Hospitalier Universitaire de Québec (CHUQ), Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland
| | - Jorge Soliz
- Faculty of Medicine, Centre Hospitalier Universitaire de Québec (CHUQ), Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland
| | - Edith M Schneider Gasser
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland. .,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland. .,Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland.
| |
Collapse
|
95
|
Zhao HL, Zhang J, Zhu Y, Wu Y, Yan QG, Peng XY, Xiang XM, Tian KL, Li T, Liu LM. Protective effects of HBOC on pulmonary vascular leakage after haemorrhagic shock and the underlying mechanisms. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 48:1272-1281. [PMID: 33084450 DOI: 10.1080/21691401.2020.1835937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Volume resuscitation is an important early treatment for haemorrhagic shock. Haemoglobin-based oxygen carrier (HBOC) can expand the volume and provide oxygen for tissues. Vascular leakage is common complication in the process of haemorrhagic shock and resuscitation. The aim of this study was to observe the effects of HBOC (a bovine-derived, cross-linked tetramer haemoglobin oxygen-carrying solution, 0.5 g/L) on vascular leakage in rats after haemorrhagic shock. A haemorrhagic shock rat model and hypoxic vascular endothelial cells (VECs) were used. The role of intercellular junctions and endothelial glycocalyx in the protective effects of HBOC and the relationship with mitochondrial function were analysed. After haemorrhagic shock, the pulmonary vascular permeability to FITC-BSA, Evans Blue was increased, endothelial glycocalyx was destroyed and the expression of intercellular junction proteins was decreased. After haemorrhagic shock, a small volume of HBOC solution (6 ml/kg) protected pulmonary vascular permeability, increased structural thickness of endothelial glycocalyx, the levels of its components and increased expression levels of the intercellular junction proteins ZO-1, VE-cadherin and occludin. Moreover, HBOC significantly increased oxygen delivery and consumption in rats, improved VEC mitochondrial function and structure. In conclusion, HBOC mitigates endothelial leakage by protecting endothelial glycocalyx and intercellular junctions through improving mitochondrial function and tissue oxygen delivery.
Collapse
Affiliation(s)
- Hong Liang Zhao
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Jie Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Yue Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Qing Guang Yan
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Xiao Yong Peng
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Xin Ming Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Kun Lun Tian
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| | - Liang Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, P.R. China
| |
Collapse
|
96
|
Dasgupta A, Chen KH, Lima PDA, Mewburn J, Wu D, Al-Qazazi R, Jones O, Tian L, Potus F, Bonnet S, Archer SL. PINK1-induced phosphorylation of mitofusin 2 at serine 442 causes its proteasomal degradation and promotes cell proliferation in lung cancer and pulmonary arterial hypertension. FASEB J 2021; 35:e21771. [PMID: 34275172 DOI: 10.1096/fj.202100361r] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/06/2021] [Accepted: 06/17/2021] [Indexed: 12/11/2022]
Abstract
Impaired mitochondrial fusion, due in part to decreased mitofusin 2 (Mfn2) expression, contributes to unrestricted cell proliferation and apoptosis-resistance in hyperproliferative diseases like pulmonary arterial hypertension (PAH) and non-small cell lung cancer (NSCLC). We hypothesized that Mfn2 levels are reduced due to increased proteasomal degradation of Mfn2 triggered by its phosphorylation at serine 442 (S442) and investigated the potential kinase mediators. Mfn2 expression was decreased and Mfn2 S442 phosphorylation was increased in pulmonary artery smooth muscle cells from PAH patients and in NSCLC cells. Mfn2 phosphorylation was mediated by PINK1 and protein kinase A (PKA), although only PINK1 expression was increased in these diseases. We designed a S442 phosphorylation deficient Mfn2 construct (PD-Mfn2) and a S442 constitutively phosphorylated Mfn2 construct (CP-Mfn2). The effects of these modified Mfn2 constructs on Mfn2 expression and biological function were compared with those of the wildtype Mfn2 construct (WT-Mfn2). WT-Mfn2 increased Mfn2 expression and mitochondrial fusion in both PAH and NSCLC cells resulting in increased apoptosis and decreased cell proliferation. Compared to WT-Mfn2, PD-Mfn2 caused greater Mfn2 expression, suppression of proliferation, apoptosis induction, and cell cycle arrest. Conversely, CP-Mfn2 caused only a small increase in Mfn2 expression and did not restore mitochondrial fusion, inhibit cell proliferation, or induce apoptosis. Silencing PINK1 or PKA, or proteasome blockade using MG132, increased Mfn2 expression, enhanced mitochondrial fusion and induced apoptosis. In a xenotransplantation NSCLC model, PD-Mfn2 gene therapy caused greater tumor regression than did therapy with WT-Mfn2. Mfn2 deficiency in PAH and NSCLC reflects proteasomal degradation triggered by Mfn2-S442 phosphorylation by PINK1 and/or PKA. Inhibiting Mfn2 phosphorylation has potential therapeutic benefit in PAH and lung cancer.
Collapse
Affiliation(s)
- Asish Dasgupta
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Kuang-Hueih Chen
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Patricia D A Lima
- Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Translational Institute of Medicine (TIME), Queen's University, Kingston, ON, Canada
| | - Jeffrey Mewburn
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Ruaa Al-Qazazi
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Oliver Jones
- Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Translational Institute of Medicine (TIME), Queen's University, Kingston, ON, Canada
| | - Lian Tian
- Department of Medicine, Queen's University, Kingston, ON, Canada.,Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Francois Potus
- Department of Medicine, Queen's University, Kingston, ON, Canada.,Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec Research Center, Laval University, Quebec City, QC, Canada
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec Research Center, Laval University, Quebec City, QC, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, ON, Canada.,Queen's Cardiopulmonary Unit (QCPU), Department of Medicine, Translational Institute of Medicine (TIME), Queen's University, Kingston, ON, Canada
| |
Collapse
|
97
|
Cen X, Zhang M, Zhou M, Ye L, Xia H. Mitophagy Regulates Neurodegenerative Diseases. Cells 2021; 10:1876. [PMID: 34440645 PMCID: PMC8392649 DOI: 10.3390/cells10081876] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play an essential role in supplying energy for the health and survival of neurons. Mitophagy is a metabolic process that removes dysfunctional or redundant mitochondria. This process preserves mitochondrial health. However, defective mitophagy triggers the accumulation of damaged mitochondria, causing major neurodegenerative disorders. This review introduces molecular mechanisms and signaling pathways behind mitophagy regulation. Furthermore, we focus on the recent advances in understanding the potential role of mitophagy in the pathogenesis of major neurodegenerative diseases (Parkinson's, Alzheimer's, Huntington's, etc.) and aging. The findings will help identify the potential interventions of mitophagy regulation and treatment strategies of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xufeng Cen
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Manke Zhang
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
| | - Mengxin Zhou
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
| | - Lingzhi Ye
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Hongguang Xia
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| |
Collapse
|
98
|
Mitochondrial quality control in intervertebral disc degeneration. Exp Mol Med 2021; 53:1124-1133. [PMID: 34272472 PMCID: PMC8333068 DOI: 10.1038/s12276-021-00650-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/18/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is a common and early-onset pathogenesis in the human lifespan that can increase the risk of low back pain. More clarification of the molecular mechanisms associated with the onset and progression of IDD is likely to help establish novel preventive and therapeutic strategies. Recently, mitochondria have been increasingly recognized as participants in regulating glycolytic metabolism, which has historically been regarded as the main metabolic pathway in intervertebral discs due to their avascular properties. Indeed, mitochondrial structural and functional disruption has been observed in degenerated nucleus pulposus (NP) cells and intervertebral discs. Multilevel and well-orchestrated strategies, namely, mitochondrial quality control (MQC), are involved in the maintenance of mitochondrial integrity, mitochondrial proteostasis, the mitochondrial antioxidant system, mitochondrial dynamics, mitophagy, and mitochondrial biogenesis. Here, we address the key evidence and current knowledge of the role of mitochondrial function in the IDD process and consider how MQC strategies contribute to the protective and detrimental properties of mitochondria in NP cell function. The relevant potential therapeutic treatments targeting MQC for IDD intervention are also summarized. Further clarification of the functional and synergistic mechanisms among MQC mechanisms may provide useful clues for use in developing novel IDD treatments.
Collapse
|
99
|
Cui P, Zhang P, Yuan L, Wang L, Guo X, Cui G, Zhang Y, Li M, Zhang X, Li X, Yin Y, Yu Z. HIF-1α Affects the Neural Stem Cell Differentiation of Human Induced Pluripotent Stem Cells via MFN2-Mediated Wnt/β-Catenin Signaling. Front Cell Dev Biol 2021; 9:671704. [PMID: 34235146 PMCID: PMC8256873 DOI: 10.3389/fcell.2021.671704] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/28/2021] [Indexed: 11/20/2022] Open
Abstract
Hypoxia-inducible factor 1α (HIF-1α) plays pivotal roles in maintaining pluripotency, and the developmental potential of pluripotent stem cells (PSCs). However, the mechanisms underlying HIF-1α regulation of neural stem cell (NSC) differentiation of human induced pluripotent stem cells (hiPSCs) remains unclear. In this study, we demonstrated that HIF-1α knockdown significantly inhibits the pluripotency and self-renewal potential of hiPSCs. We further uncovered that the disruption of HIF-1α promotes the NSC differentiation and development potential in vitro and in vivo. Mechanistically, HIF-1α knockdown significantly enhances mitofusin2 (MFN2)-mediated Wnt/β-catenin signaling, and excessive mitochondrial fusion could also promote the NSC differentiation potential of hiPSCs via activating the β-catenin signaling. Additionally, MFN2 significantly reverses the effects of HIF-1α overexpression on the NSC differentiation potential and β-catenin activity of hiPSCs. Furthermore, Wnt/β-catenin signaling inhibition could also reverse the effects of HIF-1α knockdown on the NSC differentiation potential of hiPSCs. This study provided a novel strategy for improving the directed differentiation efficiency of functional NSCs. These findings are important for the development of potential clinical interventions for neurological diseases caused by metabolic disorders.
Collapse
Affiliation(s)
- Peng Cui
- Institute of Precision of Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ping Zhang
- Department of Hematology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lin Yuan
- Institute of Precision of Medicine, Peking University Shenzhen Hospital, Shenzhen, China.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Li Wang
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xin Guo
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guanghui Cui
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yanmin Zhang
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Minghua Li
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaowei Zhang
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaoqiang Li
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuxin Yin
- Institute of Precision of Medicine, Peking University Shenzhen Hospital, Shenzhen, China.,Institute of Systems Biomedicine, Department of Pathology, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| | - Zhendong Yu
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
100
|
Brown KE, Fisher AG. Reprogramming lineage identity through cell-cell fusion. Curr Opin Genet Dev 2021; 70:15-23. [PMID: 34087754 DOI: 10.1016/j.gde.2021.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/12/2021] [Accepted: 04/23/2021] [Indexed: 12/23/2022]
Abstract
The conversion of differentiated cells to a pluripotent state through somatic cell nuclear transfer provided the first unequivocal evidence that differentiation was reversible. In more recent times, introducing a combination of key transcription factors into terminally differentiated mammalian cells was shown to drive their conversion to induced pluripotent stem cells (iPSCs). These discoveries were transformative, but the relatively slow speed (2-3 weeks) and low efficiency of reprogramming (0.1-1%) made deciphering the underlying molecular mechanisms difficult and complex. Cell fusion provides an alternative reprogramming approach that is both efficient and tractable, particularly when combined with modern multi-omics analysis of individual cells. Here we review the history and the recent advances in cell-cell fusion that are enabling a better understanding cell fate conversion, and we discuss how this knowledge could be used to shape improved strategies for regenerative medicine.
Collapse
Affiliation(s)
- Karen E Brown
- Epigenetic Memory Group, MRC London Institute of Medical Sciences (LMS), Imperial College London, Du Cane Road, London W12 0NN, UK.
| | - Amanda G Fisher
- Epigenetic Memory Group, MRC London Institute of Medical Sciences (LMS), Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|