1
|
Chan KY, Yu Y, Kong Y, Cheng L, Yao R, Yin Chair PS, Wang P, Wang R, Sun WY, He RR, Min J, Wang F, Björklund M. GPX4-dependent ferroptosis sensitivity is a fitness trade-off for cell enlargement. iScience 2025; 28:112363. [PMID: 40330887 PMCID: PMC12053632 DOI: 10.1016/j.isci.2025.112363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/20/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Despite wide variation, each cell type has an optimal size. Maintaining optimal size is essential for cellular fitness and function but the biological basis for this remains elusive. Here, we performed fitness analysis involving genome-wide CRISPR-Cas9 knockout data from tens of human cell lines and identified that cell size influences the essentiality of genes related to mitochondria and membrane repair. These genes also included glutathione peroxidase 4 (GPX4), which safeguards membranes from oxidative damage and prevents ferroptosis-iron-dependent death. Growth beyond normal size, with or without cell-cycle arrest, increased lipid peroxidation, resulting in a ferroptosis-sensitive state. Proteomic analysis revealed cell-cycle-independent superscaling of endoplasmic reticulum, accumulation of iron, and lipidome remodeling. Even slight increases from normal cell size sensitized proliferating cells to ferroptosis as evidenced by deep-learning-based single-cell analysis. Thus, lipid peroxidation may be a fitness trade-off that constrains cell enlargement and contributes to the establishment of an optimal cell size.
Collapse
Affiliation(s)
- Kuan Yoow Chan
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Yini Yu
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Yidi Kong
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Ling Cheng
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Renzhi Yao
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Phoebe Sha Yin Chair
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Ping Wang
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Rong Wang
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Wan-Yang Sun
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mikael Björklund
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9JZ, UK
| |
Collapse
|
2
|
Cerullo D, Mantzouratou P, Lavecchia AM, Balsamo M, Corna D, Brunelli L, Xinaris C. Triiodothyronine protects infarcted myocardium by reducing apoptosis and preserving mitochondria. Basic Res Cardiol 2025:10.1007/s00395-025-01106-z. [PMID: 40232385 DOI: 10.1007/s00395-025-01106-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/13/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025]
Abstract
Myocardial infarction (MI) is a leading cause of heart failure, with thyroid hormone (TH) signaling playing a key role in heart function and postinfarct recovery. Despite evidence of TH administration's safety in cardiac patients, inconsistent therapeutic outcomes and limited understanding of its mechanisms hinder clinical translation. This study aims to investigate the long-term effect of acute triiodothyronine (T3) administration following MI and to elucidate the mechanisms of its cardioprotective actions. To this end, two doses (40 μg/kg) of T3 were administered immediately after injury and 24 h later in a cryoinjury mouse model of left ventricle (LV) infarction. Remarkably T3 administration significantly reduced scar expansion. Echocardiographic analysis conducted 28 days post-injury revealed that T3 administration improved LV remodeling and prevented LV hypertrophy. At molecular level, T3 administration strongly reduced apoptosis in the peri-infarcted area, without inducing cardiac cell proliferation. Furthermore, T3 prevented the accumulation of long-chain acylcarnitines and the subsequent mitochondrial damage. These findings demonstrate that acute T3 treatment following MI improves long-term LV function and reduces LV remodeling by limiting apoptosis in the peri-infarct region and by preserving mitochondrial function and structural integrity.
Collapse
Affiliation(s)
- Domenico Cerullo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Molecular Medicine, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
| | - Polyxeni Mantzouratou
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Molecular Medicine, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
| | - Angelo M Lavecchia
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Molecular Medicine, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
| | - Melissa Balsamo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Molecular Medicine, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
| | - Daniela Corna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Molecular Medicine, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
| | - Laura Brunelli
- Environmental Health Sciences Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy
| | - Christodoulos Xinaris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Molecular Medicine, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy.
| |
Collapse
|
3
|
He S, Li Z, Xie L, Lin R, Yan B, Li B, Luo L, Xv Y, Wen H, Liang Y, Huang C, Li Z. Biomimetic gene delivery system coupled with extracellular vesicle-encapsulated AAV for improving diabetic wound through promoting vascularization and remodeling of inflammatory microenvironment. J Nanobiotechnology 2025; 23:242. [PMID: 40128816 PMCID: PMC11931832 DOI: 10.1186/s12951-025-03261-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
Adeno-associated virus (AAV)-mediated gene transfer has demonstrated potential in effectively promoting re-epithelialization and angiogenesis. AAV vector has a safety profile; however, the relatively low delivery efficacy in chronic wound with an inflammatory microenvironment and external exposure has limited its prospective clinical translation. Here, we generated AAV-containing EVs (EV-AAVs) from cultured HEK 293T cells and confirmed that the gene transfer efficiency of VEGF-EV-AAV significantly surpassed that of free AAV. Subsequently, a biomimetic gene delivery system VEGF-EV-AAV/MSC-Exo@FHCCgel developing, and synergistically enhances anti-inflammation and transfection efficiency in the combination of human umbilical cord mesenchymal stem cell-derived exosomes (hUC-MSC-Exo). Upon reaching physiological temperature, this hydrogel system transitions to a gel state, maintaining AAV bioactivity and facilitating a sustained release of the encapsulated vesicles. The encapsulation strategy enables the vesicles to rapidly fuse with endothelial cell membranes, ensuring controlled expression of endogenous VEGF. Results revealed that VEGF-EV-AAV/MSC-Exo@FHCCgel alleviates mitochondrial function in endotheliocyte under oxidative stress. Furthermore, it eliminates senescent macrophages by inhabitation of cyclic GMP-AMP (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway to promote efferocytosis. The system increases Treg cells accumulation, leading to a reduction of inflammatory cytokines. Collectively, the biomimetic gene delivery system represents a promising multi-faceted strategy for chronic wound healing.
Collapse
Affiliation(s)
- Shan He
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhenhao Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Xie
- Department of Radiology, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Rongtian Lin
- R&D Center, Guangdong Luofushan Sinopharm Co., Ltd., Huizhou, 516100, China
| | - Biying Yan
- Department of Biology, Faculty of Arts and Sciences, Beijing Normal University at Zhuhai, Zhuhai, 519087, China
- Center for Biological Science and Technology, Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Zhuhai-Macao Biotechnology Joint Laboratory, Beijing Normal University at Zhuhai, Zhuhai, 519087, China
| | - Bixiang Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lingxi Luo
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Youshan Xv
- Huiqiao Medical Center (International Medical Service), NanFang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huangding Wen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yaxuan Liang
- Department of Biology, Faculty of Arts and Sciences, Beijing Normal University at Zhuhai, Zhuhai, 519087, China.
- Center for Biological Science and Technology, Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, Zhuhai-Macao Biotechnology Joint Laboratory, Beijing Normal University at Zhuhai, Zhuhai, 519087, China.
| | - Cong Huang
- Department of Ultrasound, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| | - Zhiqing Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
4
|
de Calbiac H, Imbard A, de Lonlay P. Cellular mechanisms of acute rhabdomyolysis in inherited metabolic diseases. J Inherit Metab Dis 2025; 48:e12781. [PMID: 39135340 DOI: 10.1002/jimd.12781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 12/28/2024]
Abstract
Acute rhabdomyolysis (RM) constitutes a life-threatening emergency resulting from the (acute) breakdown of skeletal myofibers, characterized by a plasma creatine kinase (CK) level exceeding 1000 IU/L in response to a precipitating factor. Genetic predisposition, particularly inherited metabolic diseases, often underlie RM, contributing to recurrent episodes. Both sporadic and congenital forms of RM share common triggers. Considering the skeletal muscle's urgent need to rapidly adjust to environmental cues, sustaining sufficient energy levels and functional autophagy and mitophagy processes are vital for its preservation and response to stressors. Crucially, the composition of membrane lipids, along with lipid and calcium transport, and the availability of adenosine triphosphate (ATP), influence membrane biophysical properties, membrane curvature in skeletal muscle, calcium channel signaling regulation, and determine the characteristics of autophagic organelles. Consequently, a genetic defect involving ATP depletion, aberrant calcium release, abnormal lipid metabolism and/or lipid or calcium transport, and/or impaired anterograde trafficking may disrupt autophagy resulting in RM. The complex composition of lipid membranes also alters Toll-like receptor signaling and viral replication. In response, infections, recognized triggers of RM, stimulate increased levels of inflammatory cytokines, affecting skeletal muscle integrity, energy metabolism, and cellular trafficking, while elevated temperatures can reduce the activity of thermolabile enzymes. Overall, several mechanisms can account for RMs and may be associated in the same disease-causing RM.
Collapse
Affiliation(s)
- Hortense de Calbiac
- INSERM U1151, Institut Necker Enfants-Malades (INEM), Université Paris Cité, Paris, France
| | - Apolline Imbard
- Service de Biochimie, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Faculté de pharmacie, LYPSIS, Université Paris Saclay, Orsay, France
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Filière G2M, MetabERN, Paris, France
| | - Pascale de Lonlay
- INSERM U1151, Institut Necker Enfants-Malades (INEM), Université Paris Cité, Paris, France
- Reference Center for Inherited Metabolic Diseases, Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Filière G2M, MetabERN, Paris, France
| |
Collapse
|
5
|
Lanz MC, Zhang S, Swaffer MP, Ziv I, Götz LH, Kim J, McCarthy F, Jarosz DF, Elias JE, Skotheim JM. Genome dilution by cell growth drives starvation-like proteome remodeling in mammalian and yeast cells. Nat Struct Mol Biol 2024; 31:1859-1871. [PMID: 39048803 DOI: 10.1038/s41594-024-01353-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
Cell size is tightly controlled in healthy tissues and single-celled organisms, but it remains unclear how cell size influences physiology. Increasing cell size was recently shown to remodel the proteomes of cultured human cells, demonstrating that large and small cells of the same type can be compositionally different. In the present study, we utilize the natural heterogeneity of hepatocyte ploidy and yeast genetics to establish that the ploidy-to-cell size ratio is a highly conserved determinant of proteome composition. In both mammalian and yeast cells, genome dilution by cell growth elicits a starvation-like phenotype, suggesting that growth in large cells is restricted by genome concentration in a manner that mimics a limiting nutrient. Moreover, genome dilution explains some proteomic changes ascribed to yeast aging. Overall, our data indicate that genome concentration drives changes in cell composition independently of external environmental cues.
Collapse
Affiliation(s)
- Michael C Lanz
- Department of Biology, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA.
| | - Shuyuan Zhang
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Inbal Ziv
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | - Jacob Kim
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Frank McCarthy
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| | - Joshua E Elias
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA
| | - Jan M Skotheim
- Department of Biology, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub San Francisco, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Bruszt K, Horvath O, Ordog K, Toth S, Juhasz K, Vamos E, Fekete K, Gallyas F, Toth K, Halmosi R, Deres L. Cardiac effects of OPA1 protein promotion in a transgenic animal model. PLoS One 2024; 19:e0310394. [PMID: 39570915 PMCID: PMC11581344 DOI: 10.1371/journal.pone.0310394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/30/2024] [Indexed: 11/24/2024] Open
Abstract
Mitochondria form a dynamic network in cells, regulated by the balance between mitochondrial fusion and fission. The inhibition of mitochondrial fission can have positive effects in acute ischemic/reperfusion injury models by preventing the fall in mitochondrial membrane potential associated with fission processes. However, inhibition of fission in chronic models is disadvantageous because it obstructs the elimination of damaged mitochondrial fragments. OPA1, in view of previous results, is a possible therapeutic target as a fusion promoter and structure stabilizer protein. We used transgenic mice in which the OMA1 cleavage sites of OPA1 were deleted. This resulted in a higher representation of L-OPA1 compared to S-OPA1. After genotyping and model validation, all animals were examined by echocardiograph on two occasions, at weeks 11 and 36. Histological samples were taken from hearts to examine mitochondrial morphology and structure remodeling. The signaling pathways related to mitochondrial dynamic processes were evaluated. Cardiomyocytes were isolated from neonatal mice to determine the efficiency of mitochondrial respiration using the SeaHorse assay method. OPA1 protein promotion has a negative effect on systolic function during aging. We confirmed that volume overload and ventricular remodeling did not manifest. The reason behind the loss of pump function might be, at least partly, due to the energy deficit caused by mitochondrial respiratory failure and damage in mitochondrial quality control pathways.
Collapse
Affiliation(s)
- Kitti Bruszt
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Orsolya Horvath
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Katalin Ordog
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Szilard Toth
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Kata Juhasz
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Eszter Vamos
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Ferenc Gallyas
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Kalman Toth
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Robert Halmosi
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Laszlo Deres
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| |
Collapse
|
7
|
Chadha Y, Khurana A, Schmoller KM. Eukaryotic cell size regulation and its implications for cellular function and dysfunction. Physiol Rev 2024; 104:1679-1717. [PMID: 38900644 PMCID: PMC11495193 DOI: 10.1152/physrev.00046.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/24/2024] [Accepted: 06/19/2024] [Indexed: 06/22/2024] Open
Abstract
Depending on cell type, environmental inputs, and disease, the cells in the human body can have widely different sizes. In recent years, it has become clear that cell size is a major regulator of cell function. However, we are only beginning to understand how the optimization of cell function determines a given cell's optimal size. Here, we review currently known size control strategies of eukaryotic cells and the intricate link of cell size to intracellular biomolecular scaling, organelle homeostasis, and cell cycle progression. We detail the cell size-dependent regulation of early development and the impact of cell size on cell differentiation. Given the importance of cell size for normal cellular physiology, cell size control must account for changing environmental conditions. We describe how cells sense environmental stimuli, such as nutrient availability, and accordingly adapt their size by regulating cell growth and cell cycle progression. Moreover, we discuss the correlation of pathological states with misregulation of cell size and how for a long time this was considered a downstream consequence of cellular dysfunction. We review newer studies that reveal a reversed causality, with misregulated cell size leading to pathophysiological phenotypes such as senescence and aging. In summary, we highlight the important roles of cell size in cellular function and dysfunction, which could have major implications for both diagnostics and treatment in the clinic.
Collapse
Affiliation(s)
- Yagya Chadha
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Arohi Khurana
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
8
|
Mangalam M, Isoyama Y, Ogata H, Nose-Ogura S, Kayaba M, Nagai N, Kiyono K. Multi-scaling allometry in human development, mammalian morphology, and tree growth. Sci Rep 2024; 14:19957. [PMID: 39198500 PMCID: PMC11358500 DOI: 10.1038/s41598-024-69199-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Various animal and plant species exhibit allometric relationships among their respective traits, wherein one trait undergoes expansion as a power-law function of another due to constraints acting on growth processes. For instance, the acknowledged consensus posits that tree height scales with the two-thirds power of stem diameter. In the context of human development, it is posited that body weight scales with the second power of height. This prevalent allometric relationship derives its nomenclature from fitting two variables linearly within a logarithmic framework, thus giving rise to the term "power-law relationship." Here, we challenge the conventional assumption that a singular power-law equation adequately encapsulates the allometric relationship between any two traits. We strategically leverage quantile regression analysis to demonstrate that the scaling exponent characterizing this power-law relationship is contingent upon the centile within these traits' distributions. This observation fundamentally underscores the proposition that individuals occupying disparate segments of the distribution may employ distinct growth strategies, as indicated by distinct power-law exponents. We introduce the innovative concept of "multi-scale allometry" to encapsulate this newfound insight. Through a comprehensive reevaluation of (i) the height-weight relationship within a cohort comprising 7, 863, 520 Japanese children aged 5-17 years for which the age, sex, height, and weight were recorded as part of a national study, (ii) the stem-diameter-height and crown-radius-height relationships within an expansive sample of 498, 838 georeferenced and taxonomically standardized records of individual trees spanning diverse geographical locations, and (iii) the brain-size-body-size relationship within an extensive dataset encompassing 1, 552 mammalian species, we resolutely substantiate the viability of multi-scale allometric analysis. This empirical substantiation advocates a paradigm shift from uni-scaling to multi-scaling allometric modeling, thereby affording greater prominence to the inherent growth processes that underlie the morphological diversity evident throughout the living world.
Collapse
Affiliation(s)
- Madhur Mangalam
- Division of Biomechanics and Research Development, Department of Biomechanics, Center for Research in Human Movement Variability, University of Nebraska at Omaha, Omaha, NE, 68182, USA.
| | - Yosuke Isoyama
- Graduate School of Engineering Science, Osaka University, Osaka, 560-8531, Japan
| | - Hitomi Ogata
- Graduate School of Humanities and Social Sciences, Hiroshima University, Hiroshima, 739-8521, Japan
| | - Sayaka Nose-Ogura
- Department of Sports Medicine and Research, Japan High-Performance Sport Center, Japan Institute Sports Sciences, Tokyo, 115-0056, Japan
- Department of Obstetrics and Gynecology, University of Tokyo Hospital, Tokyo, 113-8655, Japan
| | - Momoko Kayaba
- Faculty of Medicine, University of Tsukuba, Tsukuba, 305-8577, Japan
| | - Narumi Nagai
- School of Human Science and Environment, University of Hyogo, Himeji, 670-0092, Japan
| | - Ken Kiyono
- Graduate School of Engineering Science, Osaka University, Osaka, 560-8531, Japan
| |
Collapse
|
9
|
Manohar S, Neurohr GE. Too big not to fail: emerging evidence for size-induced senescence. FEBS J 2024; 291:2291-2305. [PMID: 37986656 DOI: 10.1111/febs.16983] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023]
Abstract
Cellular senescence refers to a permanent and stable state of cell cycle exit. This process plays an important role in many cellular functions, including tumor suppression. It was first noted that senescence is associated with increased cell size in the early 1960s; however, how this contributes to permanent cell cycle exit was poorly understood until recently. In this review, we discuss new findings that identify increased cell size as not only a consequence but also a cause of permanent cell cycle exit. We highlight recent insights into how increased cell size alters normal cellular physiology and creates homeostatic imbalances that contribute to senescence induction. Finally, we focus on the potential clinical implications of these findings in the context of cell cycle arrest-causing cancer therapeutics and speculate on how tumor cell size changes may impact outcomes in patients treated with these drugs.
Collapse
Affiliation(s)
- Sandhya Manohar
- Department of Biology, Institute for Biochemistry, ETH Zürich, Switzerland
| | - Gabriel E Neurohr
- Department of Biology, Institute for Biochemistry, ETH Zürich, Switzerland
| |
Collapse
|
10
|
Kim M, Gorelick AN, Vàzquez-García I, Williams MJ, Salehi S, Shi H, Weiner AC, Ceglia N, Funnell T, Park T, Boscenco S, O'Flanagan CH, Jiang H, Grewal D, Tang C, Rusk N, Gammage PA, McPherson A, Aparicio S, Shah SP, Reznik E. Single-cell mtDNA dynamics in tumors is driven by coregulation of nuclear and mitochondrial genomes. Nat Genet 2024; 56:889-899. [PMID: 38741018 PMCID: PMC11096122 DOI: 10.1038/s41588-024-01724-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 03/20/2024] [Indexed: 05/16/2024]
Abstract
The extent of cell-to-cell variation in tumor mitochondrial DNA (mtDNA) copy number and genotype, and the phenotypic and evolutionary consequences of such variation, are poorly characterized. Here we use amplification-free single-cell whole-genome sequencing (Direct Library Prep (DLP+)) to simultaneously assay mtDNA copy number and nuclear DNA (nuDNA) in 72,275 single cells derived from immortalized cell lines, patient-derived xenografts and primary human tumors. Cells typically contained thousands of mtDNA copies, but variation in mtDNA copy number was extensive and strongly associated with cell size. Pervasive whole-genome doubling events in nuDNA associated with stoichiometrically balanced adaptations in mtDNA copy number, implying that mtDNA-to-nuDNA ratio, rather than mtDNA copy number itself, mediated downstream phenotypes. Finally, multimodal analysis of DLP+ and single-cell RNA sequencing identified both somatic loss-of-function and germline noncoding variants in mtDNA linked to heteroplasmy-dependent changes in mtDNA copy number and mitochondrial transcription, revealing phenotypic adaptations to disrupted nuclear/mitochondrial balance.
Collapse
Affiliation(s)
- Minsoo Kim
- Tri-Institutional PhD Program in Computational Biology & Medicine, Weill Cornell Medicine, New York City, NY, USA
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Alexander N Gorelick
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ignacio Vàzquez-García
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Marc J Williams
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Sohrab Salehi
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Hongyu Shi
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Adam C Weiner
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Nick Ceglia
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Tyler Funnell
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Tricia Park
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Sonia Boscenco
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ciara H O'Flanagan
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Hui Jiang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Diljot Grewal
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Cerise Tang
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Nicole Rusk
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Payam A Gammage
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- CRUK Beatson Institute, Glasgow, UK
| | - Andrew McPherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Sam Aparicio
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Sohrab P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| | - Ed Reznik
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York City, NY, USA.
| |
Collapse
|
11
|
He S, Li Z, Wang L, Yao N, Wen H, Yuan H, Zhang J, Li Z, Shen C. A nanoenzyme-modified hydrogel targets macrophage reprogramming-angiogenesis crosstalk to boost diabetic wound repair. Bioact Mater 2024; 35:17-30. [PMID: 38304915 PMCID: PMC10831190 DOI: 10.1016/j.bioactmat.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
Diabetic wounds has a gradually increasing incidence and morbidity. Excessive inflammation due to immune imbalance leads to delayed wound healing. Here, we reveal the interconnection between activation of the NLRP3 inflammatory pathway in endotheliocyte and polarization of macrophages via the cGAS-STING pathway in the oxidative microenvironment. To enhance the immune-regulation based on repairing mitochondrial oxidative damage, a zeolitic imidazolate framework-8 coated with cerium dioxide that carries Rhoassociated protein kinase inhibition Y-27632 (CeO2-Y@ZIF-8) is developed. It is encapsulated in a photocross-linkable hydrogel (GelMA) with cationic quaternary ammonium salt groups modified to endow the antibacterial properties (CeO2-Y@ZIF-8@Gel). CeO2 with superoxide dismutase and catalase activities can remove excess reactive oxygen species to limit mitochondrial damage and Y-27632 can repair damaged mitochondrial DNA, thus improving the proliferation of endotheliocyte. After endotheliocyte uptakes CeO2-Y@ZIF-8 NPs to degrade peroxides into water and oxygen in cells and mitochondria, NLRP3 inflammatory pathway is inhibited and the leakage of oxidatively damaged mitochondrial DNA (Ox-mtDNA, a damage-associated molecular pattern) through mPTP decreases. Futhermore, as the cGAS-STING pathway activated by Ox-mtDNA down-regulated, the M2 phenotype polarization and anti-inflammatory factors increase. Collectively, CeO2-Y@ZIF-8@Gel, through remodulating the crosstalk between macrophage reprogramming and angiogenesis to alleviate inflammation in the microenvironment and accelerates wound healing.
Collapse
Affiliation(s)
- Shan He
- Department of Burns and Plastic Surgery, Fourth Medical Center of PLA General Hospital, No. 51, Fucheng Road, Haidian District, Beijing, 100048, China
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhenhao Li
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Lu Wang
- Department of Burns and Plastic Surgery, Fourth Medical Center of PLA General Hospital, No. 51, Fucheng Road, Haidian District, Beijing, 100048, China
| | - Nannan Yao
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Huangding Wen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huageng Yuan
- Department of Burns and Plastic Surgery, Fourth Medical Center of PLA General Hospital, No. 51, Fucheng Road, Haidian District, Beijing, 100048, China
| | - Jiatao Zhang
- Jiatao Zhang, Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Ministry of Industry and Information Technology, Beijing Key Laboratory of Construction Tailorable Advanced Functional Materials and Green Applications, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhiqing Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chuanan Shen
- Department of Burns and Plastic Surgery, Fourth Medical Center of PLA General Hospital, No. 51, Fucheng Road, Haidian District, Beijing, 100048, China
| |
Collapse
|
12
|
Sousa AD, Costa AL, Costa V, Pereira C. Prediction and biological analysis of yeast VDAC1 phosphorylation. Arch Biochem Biophys 2024; 753:109914. [PMID: 38290597 DOI: 10.1016/j.abb.2024.109914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/02/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
The mitochondrial outer membrane protein porin 1 (Por1), the yeast orthologue of mammalian voltage-dependent anion channel (VDAC), is the major permeability pathway for the flux of metabolites and ions between cytosol and mitochondria. In yeast, several Por1 phosphorylation sites have been identified. Protein phosphorylation is a major modification regulating a variety of biological activities, but the potential biological roles of Por1 phosphorylation remains unaddressed. In this work, we analysed 10 experimentally observed phosphorylation sites in yeast Por1 using bioinformatics tools. Two of the residues, T100 and S133, predicted to reduce and increase pore permeability, respectively, were validated using biological assays. In accordance, Por1T100D reduced mitochondrial respiration, while Por1S133E phosphomimetic mutant increased it. Por1T100A expression also improved respiratory growth, while Por1S133A caused defects in all growth conditions tested, notably in fermenting media. In conclusion, we found phosphorylation has the potential to modulate Por1, causing a marked effect on mitochondrial function. It can also impact on cell morphology and growth both in respiratory and, unpredictably, also in fermenting conditions, expanding our knowledge on the role of Por1 in cell physiology.
Collapse
Affiliation(s)
- André D Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Celular e Molecular, Universidade do Porto, Portugal
| | - Ana Luisa Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Celular e Molecular, Universidade do Porto, Portugal
| | - Vítor Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Celular e Molecular, Universidade do Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Clara Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Celular e Molecular, Universidade do Porto, Portugal.
| |
Collapse
|
13
|
Agarwala S, Dhabal S, Mitra K. Significance of quantitative analyses of the impact of heterogeneity in mitochondrial content and shape on cell differentiation. Open Biol 2024; 14:230279. [PMID: 38228170 PMCID: PMC10791538 DOI: 10.1098/rsob.230279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/15/2023] [Indexed: 01/18/2024] Open
Abstract
Mitochondria, classically known as the powerhouse of cells, are unique double membrane-bound multifaceted organelles carrying a genome. Mitochondrial content varies between cell types and precisely doubles within cells during each proliferating cycle. Mitochondrial content also increases to a variable degree during cell differentiation triggered after exit from the proliferating cycle. The mitochondrial content is primarily maintained by the regulation of mitochondrial biogenesis, while damaged mitochondria are eliminated from the cells by mitophagy. In any cell with a given mitochondrial content, the steady-state mitochondrial number and shape are determined by a balance between mitochondrial fission and fusion processes. The increase in mitochondrial content and alteration in mitochondrial fission and fusion are causatively linked with the process of differentiation. Here, we critically review the quantitative aspects in the detection methods of mitochondrial content and shape. Thereafter, we quantitatively link these mitochondrial properties in differentiating cells and highlight the implications of such quantitative link on stem cell functionality. Finally, we discuss an example of cell size regulation predicted from quantitative analysis of mitochondrial shape and content. To highlight the significance of quantitative analyses of these mitochondrial properties, we propose three independent rationale based hypotheses and the relevant experimental designs to test them.
Collapse
Affiliation(s)
- Swati Agarwala
- Department of Biology, Ashoka University, Delhi (NCR), India
| | - Sukhamoy Dhabal
- Department of Biology, Ashoka University, Delhi (NCR), India
| | - Kasturi Mitra
- Department of Biology, Ashoka University, Delhi (NCR), India
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
14
|
Fahrenthold BK, Cavanaugh MR, Tamhankar M, Lam BL, Feldon SE, Johnson BA, Huxlin KR. Training in cortically-blind fields confers patient-specific benefit against retinal thinning after occipital stroke. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.19.23298260. [PMID: 38196617 PMCID: PMC10775322 DOI: 10.1101/2023.12.19.23298260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Purpose Damage to the adult primary visual cortex (V1) causes vision loss in the contralateral hemifield, initiating a process of trans-synaptic retrograde degeneration (TRD). Here, we examined retinal correlates of TRD using a new metric to account for global changes in inner retinal thickness, and asked if perceptual training in the intact or blind field impacts its progression. Methods We performed a meta-analysis of optical coherence tomography (OCT) data in 48 participants with unilateral V1 stroke and homonymous visual defects, who completed clinical trial NCT03350919. After measuring the thickness of the macular ganglion cell and inner plexiform layers (GCL-IPL), and the peripapillary retinal nerve fiber layer (RNFL), we computed individual laterality indices (LI) at baseline and after ~6 months of daily motion discrimination training in the intact- or blind-field. Increasingly positive LI denoted greater layer thinning in retinal regions affected versus unaffected by the cortical damage. Results Pre-training, the affected GCL-IPL and RNFL were thinner than their unaffected counterparts, generating LI values positively correlated with time since stroke. Participants trained in their intact-field exhibited increased LIGCL-IPL. Those trained in their blind-field had no significant change in LIGCL-IPL. LIRNFL did not change in either group. Conclusions Relative shrinkage of the affected versus unaffected macular GCL-IPL can be reliably measured at an individual level and increases with time post-V1 stroke. Relative thinning progressed during intact-field training, but appeared to be halted by training within the blind field, suggesting a potentially neuroprotective effect of this simple behavioral intervention.
Collapse
Affiliation(s)
- Berkeley K. Fahrenthold
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Matthew R. Cavanaugh
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Madhura Tamhankar
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Byron L. Lam
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA
| | - Steven E. Feldon
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Brent A. Johnson
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, USA
| | - Krystel R. Huxlin
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| |
Collapse
|
15
|
Chevalier RL. Why is chronic kidney disease progressive? Evolutionary adaptations and maladaptations. Am J Physiol Renal Physiol 2023; 325:F595-F617. [PMID: 37675460 DOI: 10.1152/ajprenal.00134.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/08/2023] [Accepted: 08/27/2023] [Indexed: 09/08/2023] Open
Abstract
Despite significant advances in renal physiology, the global prevalence of chronic kidney disease (CKD) continues to increase. The emergence of multicellular organisms gave rise to increasing complexity of life resulting in trade-offs reflecting ancestral adaptations to changing environments. Three evolutionary traits shape CKD over the lifespan: 1) variation in nephron number at birth, 2) progressive nephron loss with aging, and 3) adaptive kidney growth in response to decreased nephron number. Although providing plasticity in adaptation to changing environments, the cell cycle must function within constraints dictated by available energy. Prioritized allocation of energy available through the placenta can restrict fetal nephrogenesis, a risk factor for CKD. Moreover, nephron loss with aging is a consequence of cell senescence, a pathway accelerated by adaptive nephron hypertrophy that maintains metabolic homeostasis at the expense of increased vulnerability to stressors. Driven by reproductive fitness, natural selection operates in early life but diminishes thereafter, leading to an exponential increase in CKD with aging, a product of antagonistic pleiotropy. A deeper understanding of the evolutionary constraints on the cell cycle may lead to manipulation of the balance between progenitor cell renewal and differentiation, regulation of cell senescence, and modulation of the balance between cell proliferation and hypertrophy. Application of an evolutionary perspective may enhance understanding of adaptation and maladaptation by nephrons in the progression of CKD, leading to new therapeutic advances.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics, The University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
16
|
Lanz MC, Zhang S, Swaffer MP, Hernández Götz L, McCarty F, Ziv I, Jarosz DF, Elias JE, Skotheim JM. Genome dilution by cell growth drives starvation-like proteome remodeling in mammalian and yeast cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562558. [PMID: 37905015 PMCID: PMC10614910 DOI: 10.1101/2023.10.16.562558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cell size is tightly controlled in healthy tissues and single-celled organisms, but it remains unclear how size influences cell physiology. Increasing cell size was recently shown to remodel the proteomes of cultured human cells, demonstrating that large and small cells of the same type can be biochemically different. Here, we corroborate these results in mouse hepatocytes and extend our analysis using yeast. We find that size-dependent proteome changes are highly conserved and mostly independent of metabolic state. As eukaryotic cells grow larger, the dilution of the genome elicits a starvation-like proteome phenotype, suggesting that growth in large cells is limited by the genome in a manner analogous to a limiting nutrient. We also demonstrate that the proteomes of replicatively-aged yeast are primarily determined by their large size. Overall, our data suggest that genome concentration is a universal determinant of proteome content in growing cells.
Collapse
|
17
|
Lavecchia AM, Mantzouratou P, Cerullo D, Locatelli M, Conti S, Tironi M, Sangalli F, Corna D, Zoja C, Remuzzi G, Xinaris C. Thyroid hormone treatment counteracts cellular phenotypical remodeling in diabetic organs. iScience 2023; 26:107826. [PMID: 37752946 PMCID: PMC10518716 DOI: 10.1016/j.isci.2023.107826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/24/2023] [Accepted: 09/01/2023] [Indexed: 09/28/2023] Open
Abstract
Diabetes mellitus and alterations in thyroid hormone (TH) signaling are closely linked. Though the role of TH signaling in cell differentiation and growth is well known, it remains unclear whether its alterations contribute to the pathobiology of diabetic cells. Here, we aim to investigate whether the administration of exogenous T3 can counteract the cellular remodeling that occurs in diabetic cardiomyocytes, podocytes, and pancreatic beta cells. Treating diabetic rats with T3 prevents dedifferentiation, pathological growth, and ultrastructural alterations in podocytes and cardiomyocytes. In vitro, T3 reverses glucose-induced growth in human podocytes and cardiomyocytes, restores cardiomyocyte cytoarchitecture, and reverses pathological alterations in kidney and cardiac organoids. Finally, T3 treatment counteracts glucose-induced transdifferentiation, cell growth, and loss in pancreatic beta cells through TH receptor alpha1 activation. Our studies indicate that TH signaling activation substantially counteracts diabetes-induced pathological remodeling, and provide a potential therapeutic approach for the treatment of diabetes and its complications.
Collapse
Affiliation(s)
- Angelo M. Lavecchia
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Polyxeni Mantzouratou
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Domenico Cerullo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Monica Locatelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Sara Conti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Matteo Tironi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Fabio Sangalli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Daniela Corna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Carlamaria Zoja
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| | - Christodoulos Xinaris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87 -, 24126 Bergamo, Italy
| |
Collapse
|
18
|
Chaudhary MR, Chaudhary S, Sharma Y, Singh TA, Mishra AK, Sharma S, Mehdi MM. Aging, oxidative stress and degenerative diseases: mechanisms, complications and emerging therapeutic strategies. Biogerontology 2023; 24:609-662. [PMID: 37516673 DOI: 10.1007/s10522-023-10050-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/28/2023] [Indexed: 07/31/2023]
Abstract
Aging accompanied by several age-related complications, is a multifaceted inevitable biological progression involving various genetic, environmental, and lifestyle factors. The major factor in this process is oxidative stress, caused by an abundance of reactive oxygen species (ROS) generated in the mitochondria and endoplasmic reticulum (ER). ROS and RNS pose a threat by disrupting signaling mechanisms and causing oxidative damage to cellular components. This oxidative stress affects both the ER and mitochondria, causing proteopathies (abnormal protein aggregation), initiation of unfolded protein response, mitochondrial dysfunction, abnormal cellular senescence, ultimately leading to inflammaging (chronic inflammation associated with aging) and, in rare cases, metastasis. RONS during oxidative stress dysregulate multiple metabolic pathways like NF-κB, MAPK, Nrf-2/Keap-1/ARE and PI3K/Akt which may lead to inappropriate cell death through apoptosis and necrosis. Inflammaging contributes to the development of inflammatory and degenerative diseases such as neurodegenerative diseases, diabetes, cardiovascular disease, chronic kidney disease, and retinopathy. The body's antioxidant systems, sirtuins, autophagy, apoptosis, and biogenesis play a role in maintaining homeostasis, but they have limitations and cannot achieve an ideal state of balance. Certain interventions, such as calorie restriction, intermittent fasting, dietary habits, and regular exercise, have shown beneficial effects in counteracting the aging process. In addition, interventions like senotherapy (targeting senescent cells) and sirtuin-activating compounds (STACs) enhance autophagy and apoptosis for efficient removal of damaged oxidative products and organelles. Further, STACs enhance biogenesis for the regeneration of required organelles to maintain homeostasis. This review article explores the various aspects of oxidative damage, the associated complications, and potential strategies to mitigate these effects.
Collapse
Affiliation(s)
- Mani Raj Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Yogita Sharma
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Thokchom Arjun Singh
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Shweta Sharma
- Chitkara School of Health Sciences, Chitkara University, Chandigarh, Punjab, 140401, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
19
|
Fahimi P, Matta CF, Okie JG. Are size and mitochondrial power of cells inter-determined? J Theor Biol 2023; 572:111565. [PMID: 37369290 DOI: 10.1016/j.jtbi.2023.111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/16/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023]
Abstract
Mitochondria are the central hub of ATP production in most eukaryotic cells. Cellular power (energy per unit time), which is primarily generated in these organelles, is crucial to our understanding of cell function in health and disease. We investigated the relation between a mitochondrion's power (metabolic rate) and host cell size by combining metabolic theory with the analysis of two recent databases, one covering 109 protists and the other 63 species including protists, metazoans, microalgae, and vascular plants. We uncovered an interesting statistical regularity: in well-fed protists, relatively elevated values of mitochondrion power cluster around the smallest cell sizes and the medium-large cell sizes. In contrast, in starved protists and metazoans, the relation between mitochondrion power and cell size is inconclusive, and in microalgae and plants, mitochondrion power seems to increase from smaller cells to larger ones (where this investigation includes plant cells of volume up to ca. 2.18 × 105 μm3). Using these results, estimates are provided of the number of active ATP synthase molecules and basal uncouplers.
Collapse
Affiliation(s)
- Peyman Fahimi
- Département de Chimie, Université Laval, Québec, QC G1V0A6, Canada
| | - Chérif F Matta
- Département de Chimie, Université Laval, Québec, QC G1V0A6, Canada; Department of Chemistry and Physics, Mount Saint Vincent University, Halifax, NS B3M2J6, Canada.
| | - Jordan G Okie
- School of Earth and Space Exploration, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
20
|
Le NT. Metabolic regulation of endothelial senescence. Front Cardiovasc Med 2023; 10:1232681. [PMID: 37649668 PMCID: PMC10464912 DOI: 10.3389/fcvm.2023.1232681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/18/2023] [Indexed: 09/01/2023] Open
Abstract
Endothelial cell (EC) senescence is increasingly recognized as a significant contributor to the development of vascular dysfunction and age-related disorders and diseases, including cancer and cardiovascular diseases (CVD). The regulation of cellular senescence is known to be influenced by cellular metabolism. While extensive research has been conducted on the metabolic regulation of senescence in other cells such as cancer cells and fibroblasts, our understanding of the metabolic regulation of EC senescence remains limited. The specific metabolic changes that drive EC senescence are yet to be fully elucidated. The objective of this review is to provide an overview of the intricate interplay between cellular metabolism and senescence, with a particular emphasis on recent advancements in understanding the metabolic changes preceding cellular senescence. I will summarize the current knowledge on the metabolic regulation of EC senescence, aiming to offer insights into the underlying mechanisms and future research directions.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
21
|
Triplett E, Hayes C, Emendack Y, Longing S, Monclova C, Simpson C, Laza HE. Leaf structural traits mediating pre-existing physical innate resistance to sorghum aphid in sorghum under uninfested conditions. PLANTA 2023; 258:46. [PMID: 37468707 DOI: 10.1007/s00425-023-04194-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/23/2023] [Indexed: 07/21/2023]
Abstract
KEY MESSAGE We found four indicative traits of innate immunity. Sorghum-resistant varieties had a greater trichome, stomatal and chloroplast density, and smaller mesophyll intercellular width than susceptible varieties. The sorghum aphid (SA), Melanaphis sorghi (Theobald), can severely reduce sorghum yield. The contribution of structural traits to SA resistance has not been extensively studied. Moreover, the current screening method for resistance is inherently subjective for resistance and requires infestation in plants. Quantifying the microanatomical basis of innate SA resistance is crucial for developing reliable screening tools requiring no infestation. The goal of this study was to identify structural traits linked to physical innate SA resistance in sorghum. We conducted controlled environment and field experiments under no SA infestation conditions, with two resistant (R. LBK1 and R. Tx2783) and two susceptible (R. Tx7000 and R. Tx430) varieties. Leaf tissues collected at the fifth leaf stage in the controlled environment experiment were analyzed for the epidermal and mesophyll traits using light and transmission electron microscopy. Leaf tissues collected at physiological maturity in the field experiment were analyzed for surface traits using scanning electron microscopy. Our results showed that stomatal density, trichome density, trichome length, and chloroplast density are key leaf structural traits indicative of physical innate SA resistance. We found that resistant varieties had a greater density of trichomes (39%), stomata (31%), and chloroplast (42%), and smaller mesophyll intercellular width (- 52%) than susceptible varieties. However, the chloroplast, mitochondria, and epidermal cell ultrastructural traits were ineffective indicators of SA resistance. Our findings provide the foundation for developing an objective high-throughput method for SA resistance screening. We suggest a follow-up validation experiment to confirm our outcomes under SA infestation conditions.
Collapse
Affiliation(s)
- Ethan Triplett
- Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, 79409, USA
| | - Chad Hayes
- Cropping Systems Research Laboratory, USDA-ARS, Lubbock, TX, 79415, USA
| | - Yves Emendack
- Cropping Systems Research Laboratory, USDA-ARS, Lubbock, TX, 79415, USA
| | - Scott Longing
- Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, 79409, USA
| | | | - Catherine Simpson
- Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, 79409, USA
| | - Haydee E Laza
- Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, 79409, USA.
| |
Collapse
|
22
|
Lucas V, Cavadas C, Aveleira CA. Cellular Senescence: From Mechanisms to Current Biomarkers and Senotherapies. Pharmacol Rev 2023; 75:675-713. [PMID: 36732079 DOI: 10.1124/pharmrev.122.000622] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/29/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
An increase in life expectancy in developed countries has led to a surge of chronic aging-related diseases. In the last few decades, several studies have provided evidence of the prominent role of cellular senescence in many of these pathologies. Key traits of senescent cells include cell cycle arrest, apoptosis resistance, and secretome shift to senescence-associated secretory phenotype resulting in increased secretion of various intermediate bioactive factors important for senescence pathophysiology. However, cellular senescence is a highly phenotypically heterogeneous process, hindering the discovery of totally specific and accurate biomarkers. Also, strategies to prevent the pathologic effect of senescent cell accumulation during aging by impairing senescence onset or promoting senescent cell clearance have shown great potential during in vivo studies, and some are already in early stages of clinical translation. The adaptability of these senotherapeutic approaches to human application has been questioned due to the lack of proper senescence targeting and senescence involvement in important physiologic functions. In this review, we explore the heterogeneous phenotype of senescent cells and its influence on the expression of biomarkers currently used for senescence detection. We also discuss the current evidence regarding the efficacy, reliability, development stage, and potential for human applicability of the main existing senotherapeutic strategies. SIGNIFICANCE STATEMENT: This paper is an extensive review of what is currently known about the complex process of cellular senescence and explores its most defining features. The main body of the discussion focuses on how the multifeature fluctuation of the senescence phenotype and the physiological role of cellular senescence have both caused a limitation in the search for truly reliable senescence biomarkers and the progression in the development of senotherapies.
Collapse
Affiliation(s)
- Vasco Lucas
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| | - Célia Alexandra Aveleira
- Centre for Neuroscience and Cell Biology (CNC) (V.L., C.C., C.A.A.), Centre for Innovation in Biomedicine and Biotechnology (CIBB) (V.L., C.C., C.A.A.), Faculty of Pharmacy (C.C.), and Multidisciplinary Institute of Ageing (MIA-Portugal) (C.A.A.), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
23
|
Khang A, Nguyen Q, Feng X, Howsmon DP, Sacks MS. Three-dimensional analysis of hydrogel-imbedded aortic valve interstitial cell shape and its relation to contractile behavior. Acta Biomater 2023; 163:194-209. [PMID: 35085795 PMCID: PMC9309197 DOI: 10.1016/j.actbio.2022.01.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/11/2022] [Accepted: 01/18/2022] [Indexed: 12/31/2022]
Abstract
Cell-shape is a conglomerate of mechanical, chemical, and biological mechanisms that reflects the cell biophysical state. In a specific application, we consider aortic valve interstitial cells (AVICs), which maintain the structure and function of aortic heart valve leaflets. Actomyosin stress fibers help determine AVIC shape and facilitate processes such as adhesion, contraction, and mechanosensing. However, detailed 3D assessment of stress fiber architecture and function is currently impractical. Herein, we assessed AVIC shape and contractile behaviors using hydrogel-based 3D traction force microscopy to intuit the orientation and behavior of AVIC stress fibers. We utilized spherical harmonics (SPHARM) to quantify AVIC geometries through three days of incubation, which demonstrated a shift from a spherical shape to forming substantial protrusions. Furthermore, we assessed changes in post-three day AVIC shape and contractile function within two testing regimes: (1) normal contractile level to relaxation (cytochalasin D), and (2) normal contractile level to hyper-contraction (endothelin-1). In both scenarios, AVICs underwent isovolumic shape changes and produced complex displacement fields within the hydrogel. AVICs were more elongated when relaxed and more spherical in hyper-contraction. Locally, AVIC protrusions contracted along their long axis and expanded in their circumferential direction, indicating predominately axially aligned stress fibers. Furthermore, the magnitude of protrusion displacements was correlated with protrusion length and approached a consistent displacement plateau at a similar critical length across all AVICs. This implied that stress fiber behavior is conserved, despite great variations in AVIC shapes. We anticipate our findings will bolster future investigations into AVIC stress fiber architecture and function. STATEMENT OF SIGNIFICANCE: Within the aortic valve there exists a population of aortic valve interstitial cells, which orchestrate the turnover, secretion, and remodeling of its extracellular matrix, maintaining tissue integrity and ultimately sustaining the proper mechanical function. Alterations in these processes are thought to underlie diseases of the aortic valve, which affect hundreds of thousands domestically and world-wide. Yet, to date, there are no non-surgical treatments for aortic heart valve disease, in part due to our limited understanding of the underlying disease processes. In the present study, we built upon our previous study to include a full 3D analysis of aortic valve interstitial cell shapes at differing contractile levels. The resulting detailed shape and deformation analysis provided insight into the underlying stress-fiber structures and mechanical behaviors.
Collapse
Affiliation(s)
- Alex Khang
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712-1229, USA
| | - Quan Nguyen
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712-1229, USA
| | - Xinzeng Feng
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712-1229, USA
| | - Daniel P Howsmon
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712-1229, USA
| | - Michael S Sacks
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences and the Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712-1229, USA.
| |
Collapse
|
24
|
Fung HF, Bergmann DC. Function follows form: How cell size is harnessed for developmental decisions. Eur J Cell Biol 2023; 102:151312. [PMID: 36989838 DOI: 10.1016/j.ejcb.2023.151312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Cell size has profound effects on biological function, influencing a wide range of processes, including biosynthetic capacity, metabolism, and nutrient uptake. As a result, size is typically maintained within a narrow, population-specific range through size control mechanisms, which are an active area of study. While the physiological consequences of cell size are relatively well-characterized, less is known about its developmental consequences, and specifically its effects on developmental transitions. In this review, we compare systems where cell size is linked to developmental transitions, paying particular attention to examples from plants. We conclude by proposing that size can offer a simple readout of complex inputs, enabling flexible decisions during plant development.
Collapse
|
25
|
Mori Sequeiros Garcia MM, Paz C, Castillo AF, Benzo Y, Belluno MA, Balcázar Martínez A, Maloberti PM, Cornejo Maciel F, Poderoso C. New insights into signal transduction pathways in adrenal steroidogenesis: role of mitochondrial fusion, lipid mediators, and MAPK phosphatases. Front Endocrinol (Lausanne) 2023; 14:1175677. [PMID: 37223023 PMCID: PMC10200866 DOI: 10.3389/fendo.2023.1175677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/17/2023] [Indexed: 05/25/2023] Open
Abstract
Hormone-receptor signal transduction has been extensively studied in adrenal gland. Zona glomerulosa and fasciculata cells are responsible for glucocorticoid and mineralocorticoid synthesis by adrenocorticotropin (ACTH) and angiotensin II (Ang II) stimulation, respectively. Since the rate-limiting step in steroidogenesis occurs in the mitochondria, these organelles are key players in the process. The maintenance of functional mitochondria depends on mitochondrial dynamics, which involves at least two opposite events, i.e., mitochondrial fusion and fission. This review presents state-of-the-art data on the role of mitochondrial fusion proteins, such as mitofusin 2 (Mfn2) and optic atrophy 1 (OPA1), in Ang II-stimulated steroidogenesis in adrenocortical cells. Both proteins are upregulated by Ang II, and Mfn2 is strictly necessary for adrenal steroid synthesis. The signaling cascades of steroidogenic hormones involve an increase in several lipidic metabolites such as arachidonic acid (AA). In turn, AA metabolization renders several eicosanoids released to the extracellular medium able to bind membrane receptors. This report discusses OXER1, an oxoeicosanoid receptor which has recently arisen as a novel participant in adrenocortical hormone-stimulated steroidogenesis through its activation by AA-derived 5-oxo-ETE. This work also intends to broaden knowledge of phospho/dephosphorylation relevance in adrenocortical cells, particularly MAP kinase phosphatases (MKPs) role in steroidogenesis. At least three MKPs participate in steroid production and processes such as the cellular cycle, either directly or by means of MAP kinase regulation. To sum up, this review discusses the emerging role of mitochondrial fusion proteins, OXER1 and MKPs in the regulation of steroid synthesis in adrenal cortex cells.
Collapse
Affiliation(s)
- María Mercedes Mori Sequeiros Garcia
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Cristina Paz
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Ana Fernanda Castillo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Yanina Benzo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Matías A. Belluno
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Ariana Balcázar Martínez
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Paula Mariana Maloberti
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Fabiana Cornejo Maciel
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Cecilia Poderoso
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| |
Collapse
|
26
|
McQuate A, Knecht S, Raible DW. Activity regulates a cell type-specific mitochondrial phenotype in zebrafish lateral line hair cells. eLife 2023; 12:e80468. [PMID: 36912880 PMCID: PMC10129330 DOI: 10.7554/elife.80468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
Hair cells of the inner ear are particularly sensitive to changes in mitochondria, the subcellular organelles necessary for energy production in all eukaryotic cells. There are over 30 mitochondrial deafness genes, and mitochondria are implicated in hair cell death following noise exposure, aminoglycoside antibiotic exposure, as well as in age-related hearing loss. However, little is known about the basic aspects of hair cell mitochondrial biology. Using hair cells from the zebrafish lateral line as a model and serial block-face scanning electron microscopy, we have quantifiably characterized a unique hair cell mitochondrial phenotype that includes (1) a high mitochondrial volume and (2) specific mitochondrial architecture: multiple small mitochondria apically, and a reticular mitochondrial network basally. This phenotype develops gradually over the lifetime of the hair cell. Disrupting this mitochondrial phenotype with a mutation in opa1 impacts mitochondrial health and function. While hair cell activity is not required for the high mitochondrial volume, it shapes the mitochondrial architecture, with mechanotransduction necessary for all patterning, and synaptic transmission necessary for the development of mitochondrial networks. These results demonstrate the high degree to which hair cells regulate their mitochondria for optimal physiology and provide new insights into mitochondrial deafness.
Collapse
Affiliation(s)
- Andrea McQuate
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Department of Otolaryngology-HNS, University of WashingtonSeattleUnited States
| | - Sharmon Knecht
- Department of Biological Structure, University of WashingtonSeattleUnited States
| | - David W Raible
- Department of Biological Structure, University of WashingtonSeattleUnited States
- Department of Otolaryngology-HNS, University of WashingtonSeattleUnited States
| |
Collapse
|
27
|
Chen P, Levy DL. Regulation of organelle size and organization during development. Semin Cell Dev Biol 2023; 133:53-64. [PMID: 35148938 PMCID: PMC9357868 DOI: 10.1016/j.semcdb.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/20/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
During early embryogenesis, as cells divide in the developing embryo, the size of intracellular organelles generally decreases to scale with the decrease in overall cell size. Organelle size scaling is thought to be important to establish and maintain proper cellular function, and defective scaling may lead to impaired development and disease. However, how the cell regulates organelle size and organization are largely unanswered questions. In this review, we summarize the process of size scaling at both the cell and organelle levels and discuss recently discovered mechanisms that regulate this process during early embryogenesis. In addition, we describe how some recently developed techniques and Xenopus as an animal model can be used to investigate the underlying mechanisms of size regulation and to uncover the significance of proper organelle size scaling and organization.
Collapse
Affiliation(s)
- Pan Chen
- Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Daniel L Levy
- Department of Molecular Biology, University of Wyoming, Laramie, WY 82071, USA.
| |
Collapse
|
28
|
Maciak S. Cell size, body size and Peto's paradox. BMC Ecol Evol 2022; 22:142. [PMID: 36513976 PMCID: PMC9746147 DOI: 10.1186/s12862-022-02096-5] [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: 09/29/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Carcinogenesis is one of the leading health concerns afflicting presumably every single animal species, including humans. Currently, cancer research expands considerably beyond medicine, becoming a focus in other branches of natural science. Accumulating evidence suggests that a proportional scale of tumor deaths involves domestic and wild animals and poses economical or conservation threats to many species. Therefore, understanding the genetic and physiological mechanisms of cancer initiation and its progression is essential for our future action and contingent prevention. From this perspective, I used an evolutionary-based approach to re-evaluate the baseline for debate around Peto's paradox. First, I review the background of information on which current understanding of Peto's paradox and evolutionary concept of carcinogenesis have been founded. The weak points and limitations of theoretical modeling or indirect reasoning in studies based on intraspecific, comparative studies of carcinogenesis are highlighted. This is then followed by detail discussion of an effect of the body mass in cancer research and the importance of cell size in consideration of body architecture; also, I note to the ambiguity around cell size invariance hypothesis and hard data for variability of cell size across species are provided. Finally, I point to the new research area that is driving concepts to identify exact molecular mechanisms promoting the process of tumorigenesis, which in turn may provide a proximate explanation of Peto's paradox. The novelty of the approach proposed therein lies in intraspecies testing of the effect of differentiation of cell size/number on the probability of carcinogenesis while controlling for the confounding effect of body mass/size.
Collapse
Affiliation(s)
- Sebastian Maciak
- grid.25588.320000 0004 0620 6106Department of Evolutionary and Physiological Ecology, Faculty of Biology, University of Białystok, K. Ciołkowskiego 1J, 15-245 Białystok, Poland
| |
Collapse
|
29
|
Balachandra S, Sarkar S, Amodeo AA. The Nuclear-to-Cytoplasmic Ratio: Coupling DNA Content to Cell Size, Cell Cycle, and Biosynthetic Capacity. Annu Rev Genet 2022; 56:165-185. [PMID: 35977407 PMCID: PMC10165727 DOI: 10.1146/annurev-genet-080320-030537] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Though cell size varies between different cells and across species, the nuclear-to-cytoplasmic (N/C) ratio is largely maintained across species and within cell types. A cell maintains a relatively constant N/C ratio by coupling DNA content, nuclear size, and cell size. We explore how cells couple cell division and growth to DNA content. In some cases, cells use DNA as a molecular yardstick to control the availability of cell cycle regulators. In other cases, DNA sets a limit for biosynthetic capacity. Developmentally programmed variations in the N/C ratio for a given cell type suggest that a specific N/C ratio is required to respond to given physiological demands. Recent observations connecting decreased N/C ratios with cellular senescence indicate that maintaining the proper N/C ratio is essential for proper cellular functioning. Together, these findings suggest a causative, not simply correlative, role for the N/C ratio in regulating cell growth and cell cycle progression.
Collapse
Affiliation(s)
- Shruthi Balachandra
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire, USA; ,
| | - Sharanya Sarkar
- Department of Microbiology and Immunology, Dartmouth College, Hanover, New Hampshire, USA;
| | - Amanda A Amodeo
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire, USA; ,
| |
Collapse
|
30
|
Liu S, Tan C, Tyers M, Zetterberg A, Kafri R. What programs the size of animal cells? Front Cell Dev Biol 2022; 10:949382. [PMID: 36393871 PMCID: PMC9665425 DOI: 10.3389/fcell.2022.949382] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/07/2022] [Indexed: 01/19/2023] Open
Abstract
The human body is programmed with definite quantities, magnitudes, and proportions. At the microscopic level, such definite sizes manifest in individual cells - different cell types are characterized by distinct cell sizes whereas cells of the same type are highly uniform in size. How do cells in a population maintain uniformity in cell size, and how are changes in target size programmed? A convergence of recent and historical studies suggest - just as a thermostat maintains room temperature - the size of proliferating animal cells is similarly maintained by homeostatic mechanisms. In this review, we first summarize old and new literature on the existence of cell size checkpoints, then discuss additional advances in the study of size homeostasis that involve feedback regulation of cellular growth rate. We further discuss recent progress on the molecules that underlie cell size checkpoints and mechanisms that specify target size setpoints. Lastly, we discuss a less-well explored teleological question: why does cell size matter and what is the functional importance of cell size control?
Collapse
Affiliation(s)
- Shixuan Liu
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, United States
| | - Ceryl Tan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, University of Montréal, Montréal, QC, Canada
| | - Anders Zetterberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ran Kafri
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
31
|
Thumfart KM, Lazzeri S, Manuella F, Mansuy IM. Long-term effects of early postnatal stress on Sertoli cells. Front Genet 2022; 13:1024805. [PMID: 36353105 PMCID: PMC9638847 DOI: 10.3389/fgene.2022.1024805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2023] Open
Abstract
Sertoli cells are somatic cells in testis essential for spermatogenesis, that support the development, maturation, and differentiation of germ cells. Sertoli cells are metabolically highly active and physiologically regulated by external signals, particularly factors in the blood stream. In disease conditions, circulating pathological signals may affect Sertoli cells and consequentially, alter germ cells and fertility. While the effects of stress on reproductive cells have been well studied, how Sertoli cells respond to stress remains poorly characterized. We used a mouse model of early postnatal stress to assess the effects of stress on Sertoli cells. We developed an improved strategy based on intracellular stainings and obtained enriched preparations of Sertoli cells from exposed males. We show that adult Sertoli cells have impaired electron transport chain (ETC) pathways and that several components of ETC complexes particularly complex I, III, and IV are persistently affected. We identify serum as potential mediator of the effects of stress on Sertoli cells by showing that it can recapitulate ETC alterations in primary cells. These results highlight Sertoli cells as cellular targets of stress in early life that can keep a trace of exposure until adulthood.
Collapse
Affiliation(s)
- Kristina M. Thumfart
- Laboratory of Neuroepigenetics, Neuroscience Center Zürich, Brain Research Institute, Medical Faculty of the University Zürich, and Institute of Neuroscience of the Department of Health Science and Technology, ETH Zürich, Zurich, Switzerland
| | - Samuel Lazzeri
- Laboratory of Neuroepigenetics, Neuroscience Center Zürich, Brain Research Institute, Medical Faculty of the University Zürich, and Institute of Neuroscience of the Department of Health Science and Technology, ETH Zürich, Zurich, Switzerland
- IFOM, FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Francesca Manuella
- Laboratory of Neuroepigenetics, Neuroscience Center Zürich, Brain Research Institute, Medical Faculty of the University Zürich, and Institute of Neuroscience of the Department of Health Science and Technology, ETH Zürich, Zurich, Switzerland
| | - Isabelle M. Mansuy
- Laboratory of Neuroepigenetics, Neuroscience Center Zürich, Brain Research Institute, Medical Faculty of the University Zürich, and Institute of Neuroscience of the Department of Health Science and Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
32
|
Metabolic reprogramming and alteration of the redox state in hyper-productive MDCK cells for influenza a virus production. Biologicals 2022; 80:35-42. [PMID: 36114098 DOI: 10.1016/j.biologicals.2022.08.004] [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: 04/27/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
Influenza is a global public health issue leading to widespread morbidity and mortality with devastating economic loss annually. Madin-Darby Canine Kidney (MDCK) cell line has been a major cell line for influenza vaccine applications. Though many details of the host metabolic responses upon influenza A virus (IAV) infection have been documented, little is known about the metabolic reprogramming features of a hyper-productive host for IAV vaccine production. In this study, a MDCK cell clone H1 was shown to have a particular high productivity of 30 × 103 virions/cell. The glucose and amino acid metabolism of H1 were evaluated, indicating that the high producer had a particular metabolic reprogramming phenotype compared to its parental cell line (P): elevated glucose uptake, superior tricarboxylic acid cycle flux, moderate amino acid consumption, and better regulation of reactive oxygen species. Combined with the stronger mitochondrial function and mild antiviral and inflammatory responses characterized previously, our results indicated that the high producer had a sufficient intracellular energy supply, and balanced substrate distribution for IAV and host protein synthesis as well as the intracellular redox status. Understanding of these metabolic alterations paves the way for the rational cell line development and reasonable process optimization for high-yield influenza vaccine production.
Collapse
|
33
|
Fumagalli S, Pende M. S6 kinase 1 at the central node of cell size and ageing. Front Cell Dev Biol 2022; 10:949196. [PMID: 36036012 PMCID: PMC9417411 DOI: 10.3389/fcell.2022.949196] [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: 05/20/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Genetic evidence in living organisms from yeast to plants and animals, including humans, unquestionably identifies the Target Of Rapamycin kinase (TOR or mTOR for mammalian/mechanistic) signal transduction pathway as a master regulator of growth through the control of cell size and cell number. Among the mTOR targets, the activation of p70 S6 kinase 1 (S6K1) is exquisitely sensitive to nutrient availability and rapamycin inhibition. Of note, in vivo analysis of mutant flies and mice reveals that S6K1 predominantly regulates cell size versus cell proliferation. Here we review the putative mechanisms of S6K1 action on cell size by considering the main functional categories of S6K1 targets: substrates involved in nucleic acid and protein synthesis, fat mass accumulation, retrograde control of insulin action, senescence program and cytoskeleton organization. We discuss how S6K1 may be involved in the observed interconnection between cell size, regenerative and ageing responses.
Collapse
Affiliation(s)
| | - Mario Pende
- *Correspondence: Stefano Fumagalli, ; Mario Pende,
| |
Collapse
|
34
|
Cadart C, Heald R. Scaling of biosynthesis and metabolism with cell size. Mol Biol Cell 2022; 33:pe5. [PMID: 35862496 PMCID: PMC9582640 DOI: 10.1091/mbc.e21-12-0627] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
Cells adopt a size that is optimal for their function, and pushing them beyond this limit can cause cell aging and death by senescence or reduce proliferative potential. However, by increasing their genome copy number (ploidy), cells can increase their size dramatically and homeostatically maintain physiological properties such as biosynthesis rate. Recent studies investigating the relationship between cell size and rates of biosynthesis and metabolism under normal, polyploid, and pathological conditions are revealing new insights into how cells attain the best function or fitness for their size by tuning processes including transcription, translation, and mitochondrial respiration. A new frontier is to connect single-cell scaling relationships with tissue and whole-organism physiology, which promises to reveal molecular and evolutionary principles underlying the astonishing diversity of size observed across the tree of life.
Collapse
Affiliation(s)
- Clotilde Cadart
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA 94720-3200
| | - Rebecca Heald
- Molecular and Cell Biology Department, University of California, Berkeley, Berkeley, CA 94720-3200
| |
Collapse
|
35
|
Glazier DS. How Metabolic Rate Relates to Cell Size. BIOLOGY 2022; 11:1106. [PMID: 35892962 PMCID: PMC9332559 DOI: 10.3390/biology11081106] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/19/2022]
Abstract
Metabolic rate and its covariation with body mass vary substantially within and among species in little understood ways. Here, I critically review explanations (and supporting data) concerning how cell size and number and their establishment by cell expansion and multiplication may affect metabolic rate and its scaling with body mass. Cell size and growth may affect size-specific metabolic rate, as well as the vertical elevation (metabolic level) and slope (exponent) of metabolic scaling relationships. Mechanistic causes of negative correlations between cell size and metabolic rate may involve reduced resource supply and/or demand in larger cells, related to decreased surface area per volume, larger intracellular resource-transport distances, lower metabolic costs of ionic regulation, slower cell multiplication and somatic growth, and larger intracellular deposits of metabolically inert materials in some tissues. A cell-size perspective helps to explain some (but not all) variation in metabolic rate and its body-mass scaling and thus should be included in any multi-mechanistic theory attempting to explain the full diversity of metabolic scaling. A cell-size approach may also help conceptually integrate studies of the biological regulation of cellular growth and metabolism with those concerning major transitions in ontogenetic development and associated shifts in metabolic scaling.
Collapse
|
36
|
Lu L, Shi M, Qiu J, Shi Z, Wang C, Fu Y, Lin C, Zhang L, Tao J, Liu C, Wei F, Liang S, Zheng J. ZBTB20 regulates cardiac allograft rejection through NFкB-mediated inflammation in mouse heart transplantation. Transpl Immunol 2022; 74:101676. [PMID: 35872083 DOI: 10.1016/j.trim.2022.101676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022]
Abstract
Allograft rejection is a major obstacle for the long-term survival of heart transplantation (Htx) patients. The cardiac allograft rejection requires the activation of macrophages and effector T cells. In this study, we explored the role of zinc-finger and BTB domain containing protein 20 (ZBTB20) in the regulation of heart allograft rejection. Flow cytometry analysis of the spleen cells from mice undergoing an acute cardiac rejection revealed that the ZBTB20 protein expression was upregulated in both T and B cells(n = 4,P < 0.01). In addition, ZBTB20 gene knockdown significantly prolonged the survival of heart allografts in mice(n = 4,P < 0.01). Lack of ZBTB20 increased the expression of Foxp3 and limited the response of T helper 1 (Th1) cells(n = 4,P < 0.01). The ZBTB20-related regulation occurred through the activation of the NFкB pathway. In conclusion, our data suggest that ZBTB20 is involved in the regulation of T cells involved in acute heart allograft rejection. Hence, downregulation of ZBTB20 expression may inhibit T cells to prolong heart transplant survival.
Collapse
Affiliation(s)
- Liuyi Lu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
| | - Maomao Shi
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Junxiong Qiu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhanyue Shi
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chao Wang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Thoracic and Cardiac Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Yuan Fu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Canxiang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou 510630, China
| | - Lisui Zhang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chang Liu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Feng Wei
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shi Liang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Junmeng Zheng
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Surgery, Kiang Wu Hospital, Macao SAR, China.
| |
Collapse
|
37
|
Ortolan D, Sharma R, Volkov A, Maminishkis A, Hotaling NA, Huryn LA, Cukras C, Di Marco S, Bisti S, Bharti K. Single-cell-resolution map of human retinal pigment epithelium helps discover subpopulations with differential disease sensitivity. Proc Natl Acad Sci U S A 2022; 119:e2117553119. [PMID: 35522714 PMCID: PMC9171647 DOI: 10.1073/pnas.2117553119] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/25/2022] [Indexed: 12/31/2022] Open
Abstract
Regional phenotypic and functional differences in the retinal pigment epithelium (RPE) monolayer have been suggested to account for regional susceptibility in ocular diseases such as age-related macular degeneration (AMD), late-onset retinal degeneration (L-ORD), and choroideremia (CHM). However, a comprehensive description of human topographical RPE diversity is not yet available, thus limiting the understanding of regional RPE diversity and degenerative disease sensitivity in the eye. To develop a complete morphometric RPE map of the human eye, artificial intelligence–based software was trained to recognize, segment, and analyze RPE borders. Five statistically different, concentric RPE subpopulations (P1 to P5) were identified using cell area as a parameter, including a subpopulation (P4) with cell area comparable to that of macular cells in the far periphery of the eye. This work provides a complete reference map of human RPE subpopulations and their location in the eye. In addition, the analysis of cadaver non-AMD and AMD eyes and ultra-widefield fundus images of patients revealed differential vulnerability of the five RPE subpopulations to different retinal diseases.
Collapse
Affiliation(s)
- Davide Ortolan
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD 20892
| | - Ruchi Sharma
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD 20892
| | - Andrei Volkov
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD 20892
| | - Arvydas Maminishkis
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD 20892
| | - Nathan A. Hotaling
- Information Resources Technology Branch, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892
| | - Laryssa A. Huryn
- Ophthalmic Clinical Genetics Section, National Eye Institute, NIH, Bethesda, MD 20892
| | - Catherine Cukras
- Unit on Clinical Investigation of Retinal Disease, National Eye Institute, NIH, Bethesda, MD 20892
| | - Stefano Di Marco
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
| | - Silvia Bisti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, 16132 Genova, Italy
- Biostructures and Biosystems National Institute, 00136 Roma, Italy
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, National Eye Institute, NIH, Bethesda, MD 20892
| |
Collapse
|
38
|
Lavecchia AM, Pelekanos K, Mavelli F, Xinaris C. Cell Hypertrophy: A “Biophysical Roadblock” to Reversing Kidney Injury. Front Cell Dev Biol 2022; 10:854998. [PMID: 35309910 PMCID: PMC8927721 DOI: 10.3389/fcell.2022.854998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
In anamniotes cell loss can typically be compensated for through proliferation, but in amniotes, this capacity has been significantly diminished to accommodate tissue complexity. In order to cope with the increased workload that results from cell death, instead of proliferation highly specialised post-mitotic cells undergo polyploidisation and hypertrophy. Although compensatory hypertrophy is the main strategy of repair/regeneration in various parenchymal tissues, the long-term benefits and its capacity to sustain complete recovery of the kidney has not been addressed sufficiently. In this perspective article we integrate basic principles from biophysics and biology to examine whether renal cell hypertrophy is a sustainable adaptation that can efficiently regenerate tissue mass and restore organ function, or a maladaptive detrimental response.
Collapse
Affiliation(s)
- Angelo Michele Lavecchia
- Laboratory of Organ Regeneration, Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| | | | - Fabio Mavelli
- Department of Chemistry, University of Bari Aldo Moro, Bari, Italy
| | - Christodoulos Xinaris
- Laboratory of Organ Regeneration, Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
- *Correspondence: Christodoulos Xinaris,
| |
Collapse
|
39
|
Ultrastructural Changes in Mitochondria in Patients with Dilated Cardiomyopathy and Parvovirus B19 Detected in Heart Tissue without Myocarditis. J Pers Med 2022; 12:jpm12020177. [PMID: 35207664 PMCID: PMC8880015 DOI: 10.3390/jpm12020177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/12/2022] [Accepted: 01/20/2022] [Indexed: 01/04/2023] Open
Abstract
Understanding the meaning of parvovirus B19 (PB19V) in an etiology of dilated cardiomyopathy (DCM) is difficult. Viruses change the dynamics of the mitochondria by interfering with the mitochondrial process/function, causing the alteration of mitochondrial morphology. In this study, the ultrastructural changes in the mitochondria in endomyocardial biopsy (EMB) samples from patients with DCM and PB19V were determined. Methods: The PB19V evaluation was performed in EMB specimens by real-time PCR in 20 patients (age: 28 ± 6 years). The biopsy specimens were examined by histo- and immunohistochemistry to detect the inflammatory response. The ultrastructural features of the mitochondria were evaluated by electron microscopy. Results: The presence of PB19V in the heart tissue without the presence of inflammatory process, defined according to Dallas and immunohistochemical criteria, was associated with ultrastructural changes in the mitochondria. Distinctive ultrastructural pathologies were indicated, such as the presence of mitochondria in the vicinity of the expanded sarcoplasmic reticulum with amorphous material, blurred structure of mitochondria, interrupted outer mitochondrial membrane and mitophagy. Conclusions: Extending diagnostics with ultrastructural analysis of biopsy samples provides new knowledge of the changes associated with the presence of PB19V in the heart tissue. The observed changes can be a basis for searching for the damage mechanisms, as well as for new therapeutic solutions.
Collapse
|
40
|
Fontana F, Limonta P. The multifaceted roles of mitochondria at the crossroads of cell life and death in cancer. Free Radic Biol Med 2021; 176:203-221. [PMID: 34597798 DOI: 10.1016/j.freeradbiomed.2021.09.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022]
Abstract
Mitochondria are the cytoplasmic organelles mostly known as the "electric engine" of the cells; however, they also play pivotal roles in different biological processes, such as cell growth/apoptosis, Ca2+ and redox homeostasis, and cell stemness. In cancer cells, mitochondria undergo peculiar functional and structural dynamics involved in the survival/death fate of the cell. Cancer cells use glycolysis to support macromolecular biosynthesis and energy production ("Warburg effect"); however, mitochondrial OXPHOS has been shown to be still active during carcinogenesis and even exacerbated in drug-resistant and stem cancer cells. This metabolic rewiring is associated with mutations in genes encoding mitochondrial metabolic enzymes ("oncometabolites"), alterations of ROS production and redox biology, and a fine-tuned balance between anti-/proapoptotic proteins. In cancer cells, mitochondria also experience dynamic alterations from the structural point of view undergoing coordinated cycles of biogenesis, fusion/fission and mitophagy, and physically communicating with the endoplasmic reticulum (ER), through the Ca2+ flux, at the MAM (mitochondria-associated membranes) levels. This review addresses the peculiar mitochondrial metabolic and structural dynamics occurring in cancer cells and their role in coordinating the balance between cell survival and death. The role of mitochondrial dynamics as effective biomarkers of tumor progression and promising targets for anticancer strategies is also discussed.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milano, Italy.
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Milano, Italy.
| |
Collapse
|
41
|
Fernandes Gyorfy M, Miller ER, Conover JL, Grover CE, Wendel JF, Sloan DB, Sharbrough J. Nuclear-cytoplasmic balance: whole genome duplications induce elevated organellar genome copy number. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2021; 108:219-230. [PMID: 34309123 DOI: 10.1111/tpj.15436] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 06/13/2023]
Abstract
The plant genome is partitioned across three distinct subcellular compartments: the nucleus, mitochondria, and plastids. Successful coordination of gene expression among these organellar genomes and the nuclear genome is critical for plant function and fitness. Whole genome duplication (WGD) events in the nucleus have played a major role in the diversification of land plants and are expected to perturb the relative copy number (stoichiometry) of nuclear, mitochondrial, and plastid genomes. Thus, elucidating the mechanisms whereby plant cells respond to the cytonuclear stoichiometric imbalance that follows WGDs represents an important yet underexplored question in understanding the evolutionary consequences of genome doubling. We used droplet digital PCR to investigate the relationship between nuclear and organellar genome copy numbers in allopolyploids and their diploid progenitors in both wheat and Arabidopsis. Polyploids exhibit elevated organellar genome copy numbers per cell, largely preserving the cytonuclear stoichiometry observed in diploids despite the change in nuclear genome copy number. To investigate the timescale over which cytonuclear stoichiometry may respond to WGD, we also estimated the organellar genome copy number in Arabidopsis synthetic autopolyploids and in a haploid-induced diploid line. We observed corresponding changes in organellar genome copy number in these laboratory-generated lines, indicating that at least some of the cellular response to cytonuclear stoichiometric imbalance is immediate following WGD. We conclude that increases in organellar genome copy numbers represent a common response to polyploidization, suggesting that maintenance of cytonuclear stoichiometry is an important component in establishing polyploid lineages.
Collapse
Affiliation(s)
| | - Emma R Miller
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, USA
| | - Justin L Conover
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, USA
| | - Corrinne E Grover
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, USA
| | - Jonathan F Wendel
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, USA
| | - Daniel B Sloan
- Biology Department, Colorado State University, Fort Collins, CO, USA
| | - Joel Sharbrough
- Biology Department, Colorado State University, Fort Collins, CO, USA
- Biology Department, New Mexico Institute of Mining and Technology, Socorro, NM, USA
| |
Collapse
|
42
|
Ajdary M, Keyhanfar F, Moosavi MA, Shabani R, Mehdizadeh M, Varma RS. Potential toxicity of nanoparticles on the reproductive system animal models: A review. J Reprod Immunol 2021; 148:103384. [PMID: 34583090 DOI: 10.1016/j.jri.2021.103384] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/06/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022]
Abstract
Over the past two decades, nanotechnology has been involved in an array of applications in various fields, including diagnostic kits, disease treatment, drug manufacturing, drug delivery, and gene therapy. But concerns about the toxicity of nanoparticles have greatly hindered their use; also, due to their increasing use in various industries, all members of society are exposed to the toxicity of these nanoparticles. Nanoparticles have a negative impact on various organs, including the reproductive system. They also can induce abortion in women, reduce fetal growth and development, and can damage the reproductive system and sperm morphology in men. In some cases, it has been observed that despite the modification of nanoparticles in composition, concentration, and method of administration, there is still damage to the reproductive organs. Therefore, understanding how nanoparticles affect the reproductive system is of very importance. In several studies, the nanoparticle toxicity effect on the genital organs has been investigated at the clinical and molecular levels using the in vivo and in vitro models. This study reviews these investigations and provides important data on the toxicity, hazards, and safety of nanoparticles in the reproductive system to facilitate the optimal use of nanoparticles in the industry.
Collapse
Affiliation(s)
- Marziyeh Ajdary
- Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fariborz Keyhanfar
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, 14965/161, Iran
| | - Ronak Shabani
- Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Department of Anatomy, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rajender S Varma
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute, Palacky University in Olomouc, Šlechtitelů 27, 783 71, Olomouc, Czech Republic.
| |
Collapse
|
43
|
Evolution of a remarkable intracellular polymer and extreme cell allometry in hagfishes. Curr Biol 2021; 31:5062-5068.e4. [PMID: 34547222 DOI: 10.1016/j.cub.2021.08.066] [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: 03/24/2021] [Revised: 06/28/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
The size of animal cells rarely scales with body size, likely due to biophysical and physiological constraints.1,2 In hagfishes, gland thread cells (GTCs) each produce a silk-like proteinaceous fiber called a slime thread.3,4 The slime threads impart strength to a hagfish's defensive slime and thus are potentially subject to selection on their function outside of the body.5-8 Body size is of fundamental importance in predator-prey interactions, which led us to hypothesize that larger hagfishes produce longer and stronger slime threads than smaller ones.9 Here, by sampling a range of sizes of hagfish from 19 species, we systematically examined the scaling of GTC and slime-thread dimensions with body size within both phylogenetic and ontogenetic contexts. We found that the length of GTCs varied between 40 and 250 μm and scaled positively with body size, exhibiting an allometric exponent greater than those in other animal cells. Correspondingly, larger hagfishes produce longer and thicker slime threads and thus are equipped to defend against larger predators. With diameter and length varying 4-fold (0.7-4 μm and 5-22 cm, respectively) over a body-size range of 10-128 cm, the slime threads characterize the largest intracellular polymers known in biology. Our results suggest selection for stronger defensive slime in larger hagfishes has driven the evolution of extreme size and allometry of GTCs.
Collapse
|
44
|
Osteocyte Dysfunction in Joint Homeostasis and Osteoarthritis. Int J Mol Sci 2021; 22:ijms22126522. [PMID: 34204587 PMCID: PMC8233862 DOI: 10.3390/ijms22126522] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 01/29/2023] Open
Abstract
Structural disturbances of the subchondral bone are a hallmark of osteoarthritis (OA), including sclerotic changes, cystic lesions, and osteophyte formation. Osteocytes act as mechanosensory units for the micro-cracks in response to mechanical loading. Once stimulated, osteocytes initiate the reparative process by recruiting bone-resorbing cells and bone-forming cells to maintain bone homeostasis. Osteocyte-expressed sclerostin is known as a negative regulator of bone formation through Wnt signaling and the RANKL pathway. In this review, we will summarize current understandings of osteocytes at the crossroad of allometry and mechanobiology to exploit the relationship between osteocyte morphology and function in the context of joint aging and osteoarthritis. We also aimed to summarize the osteocyte dysfunction and its link with structural and functional disturbances of the osteoarthritic subchondral bone at the molecular level. Compared with normal bones, the osteoarthritic subchondral bone is characterized by a higher bone volume fraction, a larger trabecular bone number in the load-bearing region, and an increase in thickness of pre-existing trabeculae. This may relate to the aberrant expressions of sclerostin, periostin, dentin matrix protein 1, matrix extracellular phosphoglycoprotein, insulin-like growth factor 1, and transforming growth factor-beta, among others. The number of osteocyte lacunae embedded in OA bone is also significantly higher, yet the volume of individual lacuna is relatively smaller, which could suggest abnormal metabolism in association with allometry. The remarkably lower percentage of sclerostin-positive osteocytes, together with clustering of Runx-2 positive pre-osteoblasts, may suggest altered regulation of osteoblast differentiation and osteoblast-osteocyte transformation affected by both signaling molecules and the extracellular matrix. Aberrant osteocyte morphology and function, along with anomalies in molecular signaling mechanisms, might explain in part, if not all, the pre-osteoblast clustering and the uncoupled bone remodeling in OA subchondral bone.
Collapse
|
45
|
Khan AUH, Almutairi SM, Ali AK, Salcedo R, Stewart CA, Wang L, Lee SH. Expression of Nutrient Transporters on NK Cells During Murine Cytomegalovirus Infection Is MyD88-Dependent. Front Immunol 2021; 12:654225. [PMID: 34093543 PMCID: PMC8177011 DOI: 10.3389/fimmu.2021.654225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/06/2021] [Indexed: 12/03/2022] Open
Abstract
Natural killer (NK) cells are the predominant innate lymphocytes that provide early defense against infections. In the inflammatory milieu, NK cells modify their metabolism to support high energy demands required for their proliferation, activation, and functional plasticity. This metabolic reprogramming is usually accompanied by the upregulation of nutrient transporter expression on the cell surface, leading to increased nutrient uptake required for intense proliferation. The interleukin-1 family members of inflammatory cytokines are critical in activating NK cells during infection; however, their underlying mechanism in NK cell metabolism is not fully elucidated. Previously, we have shown that IL-18 upregulates the expression of solute carrier transmembrane proteins and thereby induces a robust metabolic boost in NK cells. Unexpectedly, we found that IL-18 signaling is dispensable during viral infection in vivo, while the upregulation of nutrient transporters is primarily MyD88-dependent. NK cells from Myd88-/- mice displayed significantly reduced surface expression of nutrient receptors and mTOR activity during MCMV infection. We also identified that IL-33, another cytokine employing MyD88 signaling, induces the expression of nutrient transporters but requires a pre-exposure to IL-12. Moreover, signaling through the NK cell activating receptor, Ly49H, can also promote the expression of nutrient transporters. Collectively, our findings revealed multiple pathways that can induce the expression of nutrient transporters on NK cells while highlighting the imperative role of MyD88 in NK cell metabolism during infection.
Collapse
Affiliation(s)
- Abrar Ul Haq Khan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Saeedah Musaed Almutairi
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Botany and Microbiology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Alaa Kassim Ali
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rosalba Salcedo
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, United States
| | - C. Andrew Stewart
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, United States
| | - Lisheng Wang
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Ottawa, ON, Canada
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Ottawa, ON, Canada
| |
Collapse
|
46
|
Takazaki KAG, Quinaglia T, Venancio TD, Martinez ARM, Shah RV, Neilan TG, Jerosch-Herold M, Coelho-Filho OR, França MC. Pre-clinical left ventricular myocardial remodeling in patients with Friedreich's ataxia: A cardiac MRI study. PLoS One 2021; 16:e0246633. [PMID: 33770103 PMCID: PMC7996973 DOI: 10.1371/journal.pone.0246633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 01/22/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Heart Failure (HF) is the most common cause of death in Friedreich's ataxia (FRDA), an inherited mitochondrial disease. Myocardial fibrosis and myocardial hypertrophy are well-documented autopsy features among FRDA patients with HF. OBJECTIVES To leverage the unique tissue characterization features of cardiac magnetic resonance (CMR) for characterizing myocardial remodeling in patients with genetically confirmed FRDA without HF and preserved left ventricular ejection fraction (LVEF > 55%). METHODS Twenty-seven FRDA's patients (age 27.6 ± 9.7 years, 15 women) and 10 healthy controls (32.6±7.3 years, 5 women) underwent a CMR for assessment of LV function, myocardial T1, late gadolinium enhancement (LGE), extracellular volume fraction (ECV), and intracellular water-lifetime (τic), a marker of cardiomyocyte size. RESULTS As compared to controls, FRDA patients had a preserved LVEF (LVEF: 70.5±7.4% vs. 63.9±9.0%, P<0.058), larger LV mass index (LVMASSi: 61±21.7 vs. 45±4.2g/m2, P<0.02), and decreased LV end-diastolic volume index (LVEDVi 53.1±12.0 vs. 75.7±16.1ml/m2, P<0.001), compared with controls. Additionally, ECV and cardiomyocyte size (τic,) were larger in FRDA patients (ECV: 0.36 ±0.05 vs. 0.25±0.02, P<0.001; τic: 0.15±0.08 vs. 0.06±0.03 s, P = 0.02). ECV and τic were positively associated with LV mass-to-volume ratio (ECV: r = 0.57, P = 0.003; τic: r = 0.39; P = 0.05). LVMASSi and cardiomyocyte mass-index [(1-ECV)·LVMASSi] declined with age at the CMR exam, independent of the age at initial diagnosis. CONCLUSIONS LV hypertrophy and concentric LV remodeling in FRDA are associated at the tissue level with an expansion of the ECV and an increase in cardiomyocyte size. The adverse tissue remodeling assessed by ECV and τic is associated with more severe cardiomyopathy classification, suggesting a role for these markers in tracking disease progression.
Collapse
Affiliation(s)
- Karen A. G. Takazaki
- Division of Medicine, Section of Neurology, Department of Neurology, School of Medical Sciences, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| | - Thiago Quinaglia
- Division of Medicine, Section of Cardiology, Department of Internal Medicine, Division of Medicine, Section of Neurology, School of Medical Sciences, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| | - Thiago D. Venancio
- Division of Medicine, Section of Neurology, Department of Neurology, School of Medical Sciences, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| | - Alberto R. M. Martinez
- Division of Medicine, Section of Neurology, Department of Neurology, School of Medical Sciences, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| | - Ravi V. Shah
- Division of Medicine, Section of Cardiovascular Imaging Research Program and Cardiac MR PET CT Program, Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Tomas G. Neilan
- Division of Medicine, Section of Cardiovascular Imaging Research Program and Cardiac MR PET CT Program, Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael Jerosch-Herold
- Division of Medicine, Section of MRI Physics, Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Otávio R. Coelho-Filho
- Division of Medicine, Section of Cardiology, Department of Internal Medicine, Division of Medicine, Section of Neurology, School of Medical Sciences, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| | - Marcondes C. França
- Division of Medicine, Section of Neurology, Department of Neurology, School of Medical Sciences, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| |
Collapse
|
47
|
Praharaj PP, Patro BS, Bhutia SK. Dysregulation of mitophagy and mitochondrial homeostasis in cancer stem cells: Novel mechanism for anti-cancer stem cell-targeted cancer therapy. Br J Pharmacol 2021; 179:5015-5035. [PMID: 33527371 DOI: 10.1111/bph.15401] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the potential of cancer medicine, cancer stem cells (CSCs) associated with chemoresistance and disease recurrence are the significant challenges currently opposing the efficacy of available cancer treatment options. Mitochondrial dynamics involving the fission-fusion cycle and mitophagy are the major contributing factors to better adaptation, enabling CSCs to survive and grow better under tumour micro-environment-associated stress. Moreover, mitophagy is balanced with mitochondrial biogenesis to maintain mitochondrial homeostasis in CSCs, which are necessary for the growth and maintenance of CSCs and regulate metabolic switching from glycolysis to oxidative phosphorylation. In this review, we discuss different aspects of mitochondrial dynamics, mitophagy, and mitochondrial homeostasis and their effects on modulating CSCs behaviour during cancer development. Moreover, the efficacy of pharmacological targeting of these cellular processes using anti-CSC drugs in combination with currently available chemotherapeutic drugs improves the patient's survival of aggressive cancer types.
Collapse
Affiliation(s)
- Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| | | | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| |
Collapse
|
48
|
Mitochondrial osmoregulation in evolution, cation transport and metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148368. [PMID: 33422486 DOI: 10.1016/j.bbabio.2021.148368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 11/24/2022]
Abstract
This review provides a retrospective on the role of osmotic regulation in the process of eukaryogenesis. Specifically, it focuses on the adjustments which must have been made by the original colonizing α-proteobacteria that led to the evolution of modern mitochondria. We focus on the cations that are fundamentally involved in volume determination and cellular metabolism and define the transporter landscape in relation to these ions in mitochondria as we know today. We provide analysis on how the cations interplay and together maintain osmotic balance that allows for effective ATP synthesis in the organelle.
Collapse
|
49
|
Davis BM, Guo L, Ravindran N, Shamsher E, Baekelandt V, Mitchell H, Bharath AA, De Groef L, Cordeiro MF. Dynamic changes in cell size and corresponding cell fate after optic nerve injury. Sci Rep 2020; 10:21683. [PMID: 33303775 PMCID: PMC7730151 DOI: 10.1038/s41598-020-77760-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/10/2020] [Indexed: 01/21/2023] Open
Abstract
Identifying disease-specific patterns of retinal cell loss in pathological conditions has been highlighted by the emergence of techniques such as Detection of Apoptotic Retinal Cells and Adaptive Optics confocal Scanning Laser Ophthalmoscopy which have enabled single-cell visualisation in vivo. Cell size has previously been used to stratify Retinal Ganglion Cell (RGC) populations in histological samples of optic neuropathies, and early work in this field suggested that larger RGCs are more susceptible to early loss than smaller RGCs. More recently, however, it has been proposed that RGC soma and axon size may be dynamic and change in response to injury. To address this unresolved controversy, we applied recent advances in maximising information extraction from RGC populations in retinal whole mounts to evaluate the changes in RGC size distribution over time, using three well-established rodent models of optic nerve injury. In contrast to previous studies based on sampling approaches, we examined the whole Brn3a-positive RGC population at multiple time points over the natural history of these models. The morphology of over 4 million RGCs was thus assessed to glean novel insights from this dataset. RGC subpopulations were found to both increase and decrease in size over time, supporting the notion that RGC cell size is dynamic in response to injury. However, this study presents compelling evidence that smaller RGCs are lost more rapidly than larger RGCs despite the dynamism. Finally, using a bootstrap approach, the data strongly suggests that disease-associated changes in RGC spatial distribution and morphology could have potential as novel diagnostic indicators.
Collapse
Affiliation(s)
- Benjamin M Davis
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
- Western Eye Hospital, ICORG, Imperial College London, London, NW1 5QH, UK
| | - Li Guo
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Nivedita Ravindran
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Ehtesham Shamsher
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Kapucijnenvoer 33, 3000, Leuven, Belgium
| | - Hannah Mitchell
- School of Mathematics and Physics, Queens University Belfast, Belfast, Ireland
| | - Anil A Bharath
- Department of Bioengineering, Imperial College London, South Kensington Campus, London, UK
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Naamsestraat 61, 3000, Leuven, Belgium.
| | - M Francesca Cordeiro
- Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
- Western Eye Hospital, ICORG, Imperial College London, London, NW1 5QH, UK.
| |
Collapse
|
50
|
Verspagen N, Leiva FP, Janssen IM, Verberk WCEP. Effects of developmental plasticity on heat tolerance may be mediated by changes in cell size in Drosophila melanogaster. INSECT SCIENCE 2020; 27:1244-1256. [PMID: 31829515 PMCID: PMC7687148 DOI: 10.1111/1744-7917.12742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/07/2019] [Accepted: 11/27/2019] [Indexed: 05/25/2023]
Abstract
There is a growing interest in the physiology underpinning heat tolerance of ectotherms and their responses to the ongoing rise in temperature. However, there is no consensus about the underlying physiological mechanisms. According to "the maintain aerobic scope and regulate oxygen supply" hypothesis, responses to warming at different organizational levels contribute to the ability to safeguard energy metabolism via aerobic pathways. At the cellular level, a decrease in cell size increases the capacity for the uptake of resources (e.g., food and oxygen), but the maintenance of electrochemical gradients across cellular membranes implies greater energetic costs in small cells. In this study, we investigated how different rearing temperatures affected cell size and heat tolerance in the fruit fly Drosophila melanogaster. We tested the hypothesis that smaller-celled flies are more tolerant to acute, intense heat stress whereas larger-celled flies are more tolerant to chronic, mild heat stress. We used the thermal tolerance landscape framework, which incorporates the intensity and duration of thermal challenge. Rearing temperatures strongly affected both cell size and survival times. We found different effects of developmental plasticity on tolerance to either chronic or acute heat stress. Warm-reared flies had both smaller cells and exhibited higher survival times under acute, intense heat stress when compared to cold-reared flies. However, under chronic, mild heat stress, the situation was reversed and cold-reared flies, consisting of larger cells, showed better survival. These differences in heat tolerance could have resulted from direct effects of rearing temperature or they may be mediated by the correlated changes in cell size. Notably, our results are consistent with the idea that a smaller cell size may confer tolerance to acute temperatures via enhanced oxygen supply, while a larger cell may confer greater tolerance to chronic and less intense heat stress via more efficient use of resources.
Collapse
Affiliation(s)
- Nadja Verspagen
- Department of Animal Ecology and Physiology, Institute for Water and Wetland ResearchRadboud UniversityNijmegenThe Netherlands
| | - Félix P. Leiva
- Department of Animal Ecology and Physiology, Institute for Water and Wetland ResearchRadboud UniversityNijmegenThe Netherlands
| | - Irene M. Janssen
- Department of Human Genetics, Radboud University Medical CenterRadboud Institute for Molecular Life SciencesNijmegenThe Netherlands
| | - Wilco C. E. P. Verberk
- Department of Animal Ecology and Physiology, Institute for Water and Wetland ResearchRadboud UniversityNijmegenThe Netherlands
| |
Collapse
|