1
|
Johnson CN, Evans MR, Blankenship AE, John CS, Rekowski MJ, Washburn MP, Phan A, Gouvion CM, Haeri M, Swerdlow RH, Geiger PC, Morris JK. Human skeletal muscle mitochondrial pathways are impacted by a neuropathologic diagnosis of Alzheimer's disease. Neurobiol Dis 2025; 210:106916. [PMID: 40250718 DOI: 10.1016/j.nbd.2025.106916] [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: 02/12/2025] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Alzheimer's disease (AD) is associated with reduced lean mass and impaired skeletal muscle mitochondrial and motor function. Although primary mitochondrial defects in AD may underlie these findings, molecular alterations in AD have not been thoroughly examined in human skeletal muscle. Here, we used two human skeletal muscle types, quadriceps (n = 81) and temporalis (n = 66), to compare the proteome of individuals with a neuropathologic AD diagnosis based on AD Neuropathologic Change (ADNPC+: n = 54 temporalis, 44 quadriceps) to controls (ADNPC-: n = 27 temporalis, 22 quadriceps). We determined the effects of ADNPC status within each muscle and within apolipoprotein E4 (APOE4) carriers and APOE4 non-carriers. Pathways that support mitochondrial metabolism, including oxidative phosphorylation, were downregulated in skeletal muscle of ADNPC+ versus ADNPC- individuals. Similar mitochondrial effects were observed across muscle types and APOE4 carrier groups, but nearly four times as many proteins were altered in temporalis versus quadriceps tissue and mitochondrial effects were most pronounced in APOE4 carriers compared to APOE4 non-carriers. Of all detected oxidative phosphorylation proteins, the expression of ∼29-61 % (dependent on muscle/APOE4 carrier group) significantly correlated with AD progression, ranked by Clinical Dementia Rating and ADNPC scores. Of these, 23 proteins decreased in expression with greater AD progression in all skeletal muscle type and APOE4 carrier groups. This is the first study to assess differences in the human skeletal muscle proteome in the context of AD. Our work shows that systemic mitochondrial alterations in AD extend to skeletal muscle and these effects are amplified by APOE4 and correlate with AD progression.
Collapse
Affiliation(s)
- Chelsea N Johnson
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA; University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mara R Evans
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Anneka E Blankenship
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA
| | - Casey S John
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Michaella J Rekowski
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Michael P Washburn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Andy Phan
- Bruker Daltonics, Inc, Billerica, MA 01821, USA.
| | - Cynthia M Gouvion
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA.
| | - Mohammad Haeri
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Paige C Geiger
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, KS 66205, USA; Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
2
|
Zhang Z, Mao C, Wu Y, Wang Y, Cong H. Application of non‑coding RNAs in tumors (Review). Mol Med Rep 2025; 31:164. [PMID: 40211701 PMCID: PMC12015154 DOI: 10.3892/mmr.2025.13529] [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: 10/20/2024] [Accepted: 01/31/2025] [Indexed: 04/25/2025] Open
Abstract
Tumors are associated with the highest mortality rates worldwide. For more than a decade, research has focused on the genetic involvement of proteins in cancer; however, a complete class of molecular non‑coding (nc)RNAs have been discovered in recent years, and these are considered to be associated with cancer. Notably, ncRNAs are highly conserved and multifunctional. These interact with multiple signaling pathways, influencing cell cycle progression and various physiological processes. Therefore, the present review aimed to investigate ncRNA, microRNA, transfer RNA‑derived small RNA, PIWI‑interacting RNA and long non‑coding RNA to further understand the associated generation processes, functional mechanisms and therapeutic roles in tumors. The present review demonstrated the critical role of ncRNAs in tumors, and may provide a novel theoretical basis for the role of ncRNAs as biomarkers or therapeutic tools in the treatment of cancer.
Collapse
Affiliation(s)
- Zhihan Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chunyan Mao
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yi Wu
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yin Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hui Cong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
- Department of Blood Transfusion, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
3
|
Gaston-Breton R, Disdier C, Hagberg H, Mabondzo A. Hypoxia-ischemia and sexual dimorphism: modeling mitochondrial dysfunction using brain organoids. Cell Biosci 2025; 15:67. [PMID: 40413513 PMCID: PMC12103005 DOI: 10.1186/s13578-025-01402-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/27/2025] [Indexed: 05/27/2025] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a leading cause of neurodevelopmental morbidities in full-term infants. There is strong evidence of sexual differences in hypoxic-ischemic (HI) injury where male neonates are at higher risk as they are subject to more pronounced neurological deficits and death than females. The cellular and molecular mechanisms underlying these sexual discrepancies in HI injury are poorly understood. Mitochondrial dysregulation has been increasingly explored in brain diseases and represents a major target during HI events. In this review, we discuss (1) different mitochondrial functions in the central nervous system (2), mitochondrial dysregulation in the context of HI injury (3), sex-dependent mitochondrial pathways in HIE and (4) modeling of mitochondrial dysfunction using human brain organoids. Gaining insight into these novel aspects of mitochondrial function will offer valuable understanding of brain development and neurological disorders such as HI injury, paving the way for the discovery and creation of new treatment approaches.
Collapse
Affiliation(s)
- Romane Gaston-Breton
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France
| | - Clémence Disdier
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France
| | | | - Aloïse Mabondzo
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Etude de l'Unité Neurovasculaire & Innovation Thérapeutique (LENIT), Gif-sur-Yvette cedex, 91191, France.
| |
Collapse
|
4
|
Yu L, Wang X, Lei Q, Liu Y, Li Z, Dai X, Song Z, He Y, Gao S, Yu C, Li L. Tongmai Yangxin pill alleviates myocardial ischemia/reperfusion injury by regulating mitochondrial fusion and fission through the estrogen receptor alpha/peroxisome proliferator-activated receptor gamma coactivator-1 alpha signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119639. [PMID: 40096900 DOI: 10.1016/j.jep.2025.119639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Tongmai Yangxin Pill (TMYX) is derived from the Zhigancao decoction recorded in Treatise on Cold Damage Disorders (Shang Han Lun) by Zhang Zhongjing during the Han dynasty. The prescription of TMYX reflects a therapeutic rationale and efficacy unique to traditional Chinese medicine. TMYX is clinically effective in alleviating myocardial ischemia-reperfusion injury (MI/RI). However, the precise active ingredients and underlying mechanisms remain unclear. AIM OF THE STUDY The primary objective of this study was to investigate the potential of TMYX in addressing MI/RI by activating the estrogen receptor ERα. We hypothesized that this action upregulates PGC-1α activity, subsequently promoting a balanced regulation of mitochondrial fusion and fission. MATERIALS AND METHODS UPLC-Q-TOF-MS/MS was used to identify the active components of TMYX. Subsequently, a network pharmacology approach was used to uncover the therapeutic targets and underlying pharmacological mechanisms through which TMYX mitigates MI/RI. Lastly, the anticipated outcomes were confirmed through in vivo and in vitro experimental validations. RESULTS Using UPLC-Q-TOF-MS/MS, we successfully identified 53 distinct compounds in TMYX. Network pharmacology analysis revealed 20 key TMYX targets associated with MI/RI. Enrichment studies using GO and KEGG analyses revealed that these targets were mainly associated with mitochondrial processes and estrogen signaling pathways. Both in vivo and in vitro studies confirmed that TMYX markedly improved mitochondrial function through the ERα/PGC-1α signaling cascade, leading to a reduction in the size of myocardial infarctions and the incidence of apoptosis. Notably, combining TMYX with siERα abolished the protective effect of TMYX on the mitochondria. CONCLUSION TMYX therapy can improve cardiac function in MI/RI. This effect is likely mediated by the ERα/PGC-1α signaling pathway. However, given the complex multi-component composition of traditional Chinese medicine formulas, additional studies are necessary to confirm the findings of this research.
Collapse
Affiliation(s)
- Lu Yu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Xu Wang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Qina Lei
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yutong Liu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Zhu Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Xiangdong Dai
- Pharmaron Beijing Co., Ltd. (China), BDA, 6 Taihe Road, Beijing, 100176, China
| | - Zhihui Song
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yuanyuan He
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Shan Gao
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
5
|
Zhang T, Li Z, Li J, Peng Y. Small open reading frame-encoded microproteins in cancer: identification, biological functions and clinical significance. Mol Cancer 2025; 24:105. [PMID: 40170020 PMCID: PMC11963466 DOI: 10.1186/s12943-025-02278-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
The human genome harbors approximately twenty thousand protein-coding genes, and a significant portion of life science research focuses on elucidating their functions and the underlying mechanisms. Recent studies have revealed that small open reading frame (sORF), originating from non-coding RNAs or the 5' leader sequences of messenger RNAs, can be translated into small peptides called microproteins through cap-dependent or cap-independent mechanisms. These microproteins interact with diverse molecular partners to modulate gene expression at multiple regulatory levels, thereby playing critical roles in various biological processes. Notably, sORF-encoded microproteins exhibit aberrant expression patterns in cancer and are implicated in tumor initiation and progression, expanding our understanding of cancer biology. In this review, we introduce the translational mechanisms and identification methods of microproteins, summarize their dysregulation in cancer and their biological functions in regulating gene expression, and emphasize their roles in driving hallmark events of cancer. Furthermore, we discuss their clinical significance as diagnostic and prognostic biomarkers, as well as therapeutic targets.
Collapse
Affiliation(s)
- Tingting Zhang
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhang Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Brzezniakiewicz-Janus K, Jarczak J, Konopko A, Ratajczak J, Kucia M, Ratajczak MZ. Mitochondria Express Functional Signaling Ligand-Binding Receptors that Regulate their Biological Responses - the Novel Role of Mitochondria as Stress-Response Sentinels. Stem Cell Rev Rep 2025; 21:597-604. [PMID: 39888573 PMCID: PMC11965210 DOI: 10.1007/s12015-025-10847-2] [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] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Evidence accumulated mitochondria, as the "powerplants of the cell," express several functional receptors for external ligands that modify their function and regulate cell biology. This review sheds new light on the role of these organelles in sensing external stimuli to facilitate energy production for cellular needs. This is possible because mitochondria express some receptors on their membranes that are responsible for their autonomous responses. This is not surprising given the widely accepted hypothesis that these intracellular organelles originated from prokaryotic ancestors that fused with eukaryotic cells during early evolution. It has been reported that mitochondria express functional estrogen, androgen, glucocorticoid, 5-hydroxytryptamine, melatonin, and cannabinoid receptors. What is intriguing is recent evidence showing that mitochondria could also be directly regulated by active mediators of intracellular complement (complosome) and intrinsic mediators of purinergic signaling. Accordingly, they express receptors for intracellular complement cleavage fragments (C5a and C3a) as well as for adenosine triphosphate (ATP), which, besides its crucial role in transferring energy in the cells, is also an important signaling molecule interacting with P2X7 receptor expressed not only on the cell surface but also on the mitochondria membrane. Based on this, intrinsic complosome and purinergic signaling mediators emerge as important cooperating regulators of reactive oxygen species (ROS) release from mitochondria and activators of intracellular pattern recognition receptor Nlrp3 inflammasome. This activation within the beneficial "hormetic zone response" regulates cell metabolism, proliferation, migration, and adaptation to the surrounding challenges of the microenvironment in a favorable way.
Collapse
Affiliation(s)
| | - Justyna Jarczak
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
- Center for Preclinical Studies and Technology, Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Adrian Konopko
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
- Center for Preclinical Studies and Technology, Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Janina Ratajczak
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, Kentucky, 40202, USA
| | - Magdalena Kucia
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
- Center for Preclinical Studies and Technology, Laboratory of Regenerative Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Z Ratajczak
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland.
- Stem Cell Institute at Graham Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, Kentucky, 40202, USA.
| |
Collapse
|
7
|
Tao X, Wang Y, Xiang B, Hu D, Xiong W, Liao W, Zhang S, Liu C, Wang X, Zhao Y. Sex bias in tumor immunity: insights from immune cells. Theranostics 2025; 15:5045-5072. [PMID: 40303343 PMCID: PMC12036885 DOI: 10.7150/thno.106465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/08/2025] [Indexed: 05/02/2025] Open
Abstract
Significant sex disparities have been observed in cancer incidence, treatment response to immunotherapy, and susceptibility to adverse effects, affecting both reproductive and non-reproductive organ cancers. While lifestyle factors, carcinogenic exposure, and healthcare access contribute to these disparities, they do not fully explain the observed male-female variation in anti-tumor immunity. Despite the preferential expression of sex hormone receptors in immune cells, X chromosome also contains numerous genes involved in immune function, and its incomplete inactivation may enhance anti-tumor immune responses in females. In contrast, loss or downregulation of Y-linked genes in males has been associated with an increased cancer risk. Additionally, estrogen, progesterone and androgen signaling pathways influence both innate and adaptive immune responses, contributing to sex-specific outcomes in cancer progression and therapy. Sex-biased differences are also evident in the epigenetic regulation of gene expression, cellular senescence, microbiota composition, metabolism, and DNA damage response, all of which impact anti-tumor immunity and immunotherapy treatment efficacy. In general, the combination of sex chromosomes, sex hormones, and hormone receptors orchestrates the phenotype and function of various immune cells involved in tumor immunity. However, sex disparity in each specific immune cell are context and environment dependent, considering the preferential expression of hormone receptor in immune cell and sex hormone levels fluctuate significantly across different life stages. This review aims to outline the molecular, cellular, and epigenetic changes in T cells, B cells, NK cells, DCs, neutrophils, and macrophages driven by sex chromosomes and sex hormone signaling. These insights may inform the design of sex-specific targeted therapies and leading to more individualized cancer treatment strategies.
Collapse
Affiliation(s)
- Xuerui Tao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yiling Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Binghua Xiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dongmei Hu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Xiong
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjun Liao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shichuan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiao Wang
- Department of Organ Transplantation, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Zhao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
8
|
Wang Y, Wang Y, Zhang W. Dysregulation of Mitochondrial in Pulmonary Hypertension-Related Right Ventricular Remodeling: Pathophysiological Features and Targeting Drugs. Rev Cardiovasc Med 2025; 26:25781. [PMID: 40160582 PMCID: PMC11951289 DOI: 10.31083/rcm25781] [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: 07/21/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 04/02/2025] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening condition characterized by right ventricular (RV) remodeling, which is a major determinant of patient survival. The progression of right ventricular remodeling is significantly influenced by mitochondrial dysfunction, providing profound insights into vascular health and cardiovascular risk. In this review, we discuss the molecular targets, pathophysiological characteristics, and potential mechanisms underlying mitochondrial dysfunction in PH, encompassing disturbances in mitochondrial dynamics, inflammation, and dysregulation of mitochondrial energy metabolism. Finally, we review the primary therapeutic targets currently utilized to address cardiac dysfunction resulting from mitochondrial damage. Hopefully, this might inspire novel approaches to the management of cardiovascular disorders.
Collapse
Affiliation(s)
- Yuehan Wang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Yingzhuo Wang
- The First Clinical Medical College, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Weifang Zhang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Signorile PG, Baldi A, Viceconte R, Boccellino M. The Role of Adenogenesis Factors in the Pathogenesis of Endometriosis. Int J Mol Sci 2025; 26:2076. [PMID: 40076699 PMCID: PMC11899868 DOI: 10.3390/ijms26052076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Endometriosis is a pathological condition characterized by the presence of the endometrial tissue, outside the uterine cavity. It affects nearly 10% of women of reproductive age and is responsible for infertility, chronic pain, and the weakening of the quality of life. Various pathogenetic mechanisms have been suggested; however, the essential pathogenesis of endometriosis remains insufficiently comprehended. A comprehensive literature search was conducted in databases such as PubMed, Scopus, and Web of Science up to December 2024. Inclusion criteria encompassed studies investigating the pathogenetic mechanisms of endometriosis, while exclusion criteria included reviews, case reports, and studies lacking primary data. The analyzed studies explored multiple pathogenetic mechanisms, including retrograde menstruation, coelomic metaplasia, embryological defects, stem cell involvement, and epigenetic modifications. Special emphasis was placed on the role of uterine adenogenesis factors in the development and progression of endometriosis. A deeper understanding of the various pathogenetic mechanisms underlying endometriosis is crucial for advancing targeted therapeutic strategies. Further research into uterine adenogenesis factors may provide new insights into the disease's pathophysiology and pave the way for novel treatment approaches.
Collapse
Affiliation(s)
- Pietro G. Signorile
- Italian Endometriosis Foundation, Formello (RM), 00060 Rome, Italy; (R.V.); (M.B.)
| | - Alfonso Baldi
- Italian Endometriosis Foundation, Formello (RM), 00060 Rome, Italy; (R.V.); (M.B.)
- Department of Life Science, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| | - Rosa Viceconte
- Italian Endometriosis Foundation, Formello (RM), 00060 Rome, Italy; (R.V.); (M.B.)
| | - Mariarosaria Boccellino
- Italian Endometriosis Foundation, Formello (RM), 00060 Rome, Italy; (R.V.); (M.B.)
- Department of Life Science, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
10
|
Zelco A, Joshi A. Single-Cell Analysis of Sex and Gender Differences in the Human Brain During Development and Disease. Cell Mol Neurobiol 2025; 45:20. [PMID: 40016536 PMCID: PMC11868228 DOI: 10.1007/s10571-025-01536-2] [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: 08/25/2024] [Accepted: 02/07/2025] [Indexed: 03/01/2025]
Abstract
Sex and gender (SG) differences in the human brain are of interest to society and science as numerous processes are impacted by them, including brain development, behavior, and diseases. By collecting publicly available single-cell data from the in-utero to elderly age in healthy, Alzheimer's disease and multiple sclerosis samples, we identified and characterized SG-biased genes in ten brain cell types across 9 age and disease groups. Sex and gender differences in the transcriptome were present throughout the lifespan and across all cell types. Although there was limited overlap among SG-biased genes across different age and disease groups, we observed significant functional overlap. Female-biased genes are consistently enriched for brain-related processes, while male-biased genes are enriched for metabolic pathways. Additionally, mitochondrial genes showed a consistent female bias across cell types. We also found that androgen response elements (not estrogen) were significantly enriched in both male- and female-biased genes, and thymosin hormone targets being consistently enriched only in male-biased genes. We systematically characterised SG differences in brain development and brain-related disorders at a single-cell level, by analysing a total of publicly available 419,885 single nuclei from 161 human brain samples (72 females, 89 males). The significant enrichment of androgen (not estrogen) response elements in both male- and female-biased genes suggests that androgens are important regulators likely establishing these SG differences. Finally, we provide full characterization of SG-biased genes at different thresholds for the scientific community as a web resource.
Collapse
Affiliation(s)
- Aura Zelco
- Department of Clinical Science, Computational Biology Unit, University of Bergen, Bergen, Norway.
| | - Anagha Joshi
- Department of Clinical Science, Computational Biology Unit, University of Bergen, Bergen, Norway.
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, IIT Madras, Chennai, India.
| |
Collapse
|
11
|
Shirvani P, Shirvani A, Holick MF. Mitochondrial Dysfunction and Its Potential Molecular Interplay in Hypermobile Ehlers-Danlos Syndrome: A Scoping Review Bridging Cellular Energetics and Genetic Pathways. Curr Issues Mol Biol 2025; 47:134. [PMID: 39996855 PMCID: PMC11854588 DOI: 10.3390/cimb47020134] [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: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Hypermobile Ehlers-Danlos Syndrome (hEDS) is a hereditary connective tissue disorder characterized by joint hypermobility, skin hyperextensibility, and systemic manifestations such as chronic fatigue, gastrointestinal dysfunction, and neurological symptoms. Unlike other EDS subtypes with known genetic mutations, hEDS lacks definitive markers, suggesting a multifactorial etiology involving both mitochondrial dysfunction and non-mitochondrial pathways. This scoping review, conducted in accordance with the PRISMA-ScR guidelines, highlights mitochondrial dysfunction as a potential unifying mechanism in hEDS pathophysiology. Impaired oxidative phosphorylation (OXPHOS), elevated reactive oxygen species (ROS) levels, and calcium dysregulation disrupt cellular energetics and extracellular matrix (ECM) homeostasis, contributing to the hallmark features of hEDS. We reviewed candidate genes associated with ECM remodeling, signaling pathways, and immune regulation. Protein-protein interaction (PPI) network analyses revealed interconnected pathways linking mitochondrial dysfunction with these candidate genes. Comparative insights from Fabry disease and fragile X premutation carriers underscore shared mechanisms such as RNA toxicity, matrix metalloproteinases (MMP) activation, and ECM degradation. These findings may suggest that mitochondrial dysfunction amplifies systemic manifestations through its interplay with non-mitochondrial molecular pathways. By integrating these perspectives, this review provides a potential framework for understanding hEDS pathogenesis while highlighting latent avenues for future research into its molecular basis. Understanding the potential role of mitochondrial dysfunction in hEDS not only sheds light on its complex molecular etiology but also opens new paths for targeted interventions.
Collapse
Affiliation(s)
| | - Arash Shirvani
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Michael F. Holick
- Ehlers-Danlos Syndrome Clinical Research Program, Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| |
Collapse
|
12
|
O'Mahony C, Hidalgo-Lanussa O, Barreto GE. Unveiling FOXO3's metabolic contribution to menopause and Alzheimer's disease. Exp Gerontol 2025; 200:112679. [PMID: 39778695 DOI: 10.1016/j.exger.2025.112679] [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: 07/22/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
The increasing prevalence of Alzheimer's disease (AD) calls for a comprehensive exploration of its complex etiology, with a focus on sex-specific vulnerability, particularly the heightened susceptibility observed in postmenopausal women. Neurometabolic alterations during the endocrine transition emerge as early indicators of AD pathology, including reduced glucose metabolism and increased amyloid-beta (Aβ) deposition. The fluctuating endocrine environment, marked by declining estradiol levels and reduced estrogen receptor beta (ERβ) activity, further exacerbates this process. In this context, here we explore the potential of forkhead box O3 (FOXO3) as a critical mediator linking metabolic disturbances to hormonal decline. We propose that FOXO3 plays a key role in the intersection of menopause and AD, given its dysregulation in both AD patients and postmenopausal women, modulating cellular metabolism through interactions with the AMPK/AKT/PI3K pathways. This relationship highlights the intersection between hormonal changes and increased AD susceptibility. This review aims to open a discussion on FOXO3's contribution to the metabolic dysregulation seen in menopause and its impact on the progression of AD. Understanding the functional role of FOXO3 in menopause-associated metabolic changes could lead to targeted therapeutic strategies, offering novel insights for managing for this condition.
Collapse
Affiliation(s)
- Christopher O'Mahony
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland
| | - Oscar Hidalgo-Lanussa
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland.
| |
Collapse
|
13
|
Olajide OJ, Batallán Burrowes AA, da Silva IF, Bergdahl A, Chapman CA. Reduced 17β-estradiol following ovariectomy induces mitochondrial dysfunction and degradation of synaptic proteins in the entorhinal cortex. Neuroscience 2025; 565:479-486. [PMID: 39617168 DOI: 10.1016/j.neuroscience.2024.11.071] [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: 07/19/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/19/2024]
Abstract
Reductions in circulating estrogens can contribute to cognitive decline, in part by impairing mitochondrial function within the hippocampal region. The entorhinal cortex provides the hippocampus with its main cortical inputs. To assess the impact of estrogen deficiency on mitochondrial respiration and synaptic proteins in the entorhinal cortex, female wildtype rats received either sham surgery, bilateral ovariectomy, or ovariectomy with implantation of a subdermal capsule to maintain low levels of circulating 17β-estradiol (E2). Mitochondrial respiration in the entorhinal cortex was not significantly affected two weeks following ovariectomy, but there was a reduction in oxygen consumption four weeks after ovariectomy that was prevented by E2 supplementation. The expression of mitochondrial membrane integrity element voltage-dependent anion channel protein (VDAC1) was also reduced four weeks after ovariectomy, suggesting that respiration was reduced due to a decline in mitochondrial density. Ovariectomy also increased mitochondrial and cytoplasmic cytochrome c and upregulated superoxide dismutase 2 (SOD2) both two and four weeks after ovariectomy, reflecting mitochondrial electron leakage and oxidative redox imbalance. Further, the ovariectomy-induced changes in mitochondrial proteins were associated with reductions in postsynaptic density protein 95 (PSD95) and the presynaptic protein synaptophysin. There were no changes in mitochondrial or synaptic proteins in ovariectomized animals that received E2 supplementation. Our findings indicate that reductions in circulating 17β-estradiol induced by ovariectomy disrupt mitochondrial functions in the entorhinal cortex, and suggest that a resulting increase in oxidative stress contributes to the degradation in synaptic proteins that may affect cognitive functions mediated by the hippocampal region.
Collapse
Affiliation(s)
- Olayemi Joseph Olajide
- Department of Psychology, Concordia University, Montreal, Canada; Division of Neurobiology, Department of Anatomy, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | | | - Igor Ferraz da Silva
- Department of Psychology, Concordia University, Montreal, Canada; Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - Andreas Bergdahl
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
| | - C Andrew Chapman
- Department of Psychology, Concordia University, Montreal, Canada.
| |
Collapse
|
14
|
Fila M, Przyslo L, Derwich M, Pawlowska E, Blasiak J. Sexual Dimorphism in Migraine. Focus on Mitochondria. Curr Pain Headache Rep 2025; 29:11. [PMID: 39760955 DOI: 10.1007/s11916-024-01317-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE OF REVIEW Migraine prevalence in females is up to 3 times higher than in males and females show higher frequency, longer duration, and increased severity of headache attacks, but the reason for that difference is not known. This narrative review presents the main aspects of sex dimorphism in migraine prevalence and discusses the role of sex-related differences in mitochondrial homeostasis in that dimorphism. The gender dimension is also shortly addressed. RECENT FINDINGS The imbalance between energy production and demand in the brain susceptible to migraine is an important element of migraine pathogenesis. Mitochondria are the main energy source in the brain and mitochondrial impairment is reported in both migraine patients and animal models of human migraine. However, it is not known whether the observed changes are consequences of primary disturbance of mitochondrial homeostasis or are secondary to the migraine-affected hyperexcitable brain. Sex hormones regulate mitochondrial homeostasis, and several reports suggest that the female hormones may act protectively against mitochondrial impairment, contributing to more effective energy production in females, which may be utilized in the mechanisms responsible for migraine progression. Migraine is characterized by several comorbidities that are characterized by sex dimorphism in their prevalence and impairments in mitochondrial functions. Mitochondria may play a major role in sexual dimorphism in migraine through the involvement in energy production, the dependence on sex hormones, and the involvement in sex-dependent comorbidities. Studies on the role of mitochondria in sex dimorphism in migraine may contribute to precise personal therapeutic strategies.
Collapse
Affiliation(s)
- Michal Fila
- Department of Developmental Neurology and Epileptology, Polish Mother's Memorial Hospital Research Institute, 93-338, Lodz, Poland
| | - Lukasz Przyslo
- Department of Developmental Neurology and Epileptology, Polish Mother's Memorial Hospital Research Institute, 93-338, Lodz, Poland
| | - Marcin Derwich
- Department of Developmental Dentistry, Medical University of Lodz, 90-647, Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Developmental Dentistry, Medical University of Lodz, 90-647, Lodz, Poland
| | - Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, 09-420, Plock, Poland.
| |
Collapse
|
15
|
Zhou HL, Wang BB, Fan XL, Zhang XM, Song Y. Carvacrol acetate activated Nrf2 modulates mitophagy for the treatment of neurocyte oxidative stress induced by chlorpyrifos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117484. [PMID: 39644575 DOI: 10.1016/j.ecoenv.2024.117484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
This study explored the protective effect and potential mechanism of carvacrol acetate (CAA) on the oxidation of chlorpyrifos (CPF). A model of oxidative stimulus damage was established in Sprague-Dawley rats by subcutaneous injection of the CPF poison. PC12 cells were used to construct an oxidative injury model using CPF, and the protective effects and mechanism of action of CAA against CPF-induced oxidative damage were explored in vitro. The key role of Nuclear factor erythroid-2-related factor 2 (Nrf2) in alleviating CPF-induced damage via CAA was further confirmed by administering Nrf2 inhibitors to PC12 cells. Administration of CAA significantly enhanced the locomotor ability of the rats, alleviated neuronal pathological alterations, and increased the number of Nissl bodies, while increasing autophagic bodies. In vitro, CAA promoted cell survival and augmented the mitochondrial membrane potential. It decreased both intra- and extracellular levels of reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), while markedly elevating mitochondrial DNA (mtDNA) copy number. Moreover, PC12 cells treated with Nrf2 inhibitors failed to exhibit any improvement in survival rate when treated with CAA after a toxic insult. Furthermore, ROS and MDA levels were not significantly reduced, SOD enzyme activity did not increase, and mitochondrial membrane potential and mtDNA copy number did not improve. Western blot analysis showed that the expression of Tfam, Beclin1, and LC3II/LC3I proteins in the CAA group decreased significantly after Nrf2 inhibition. These findings suggest that CAA modulates mitochondrial function and autophagy by regulating the Nrf2 signalling pathway to mitigate the toxic damage. Finally, the effect of the autophagy inhibitor, 3-MA, on PC12 cells suggests that CAA promotes mitophagy by participating in the Nrf2 pathway, thereby preventing CPF-induced oxidative stress damage.
Collapse
Affiliation(s)
- Hong-Ling Zhou
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Bei-Bei Wang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| | - Xu-Li Fan
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; Jinhua People's Hospital, Jinhua, Zhejiang 2321000, China.
| | - Xiao-Min Zhang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; First People's Hospital of Linping District, Hangzhou, Zhejiang 311103, China.
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; Hangzhou King's Bio-pharmaceutical Technology Co., Ltd., Hangzhou, Zhejiang 310007, China.
| |
Collapse
|
16
|
Queiroz ALF, Garcia CB, Silva JPMO, Cavalini DFA, Alexandrino AV, Cunha AF, Vercesi AE, Castilho RF, Shiguemoto GE. Preventive Effects of Resistance Training on Hemodynamics and Kidney Mitochondrial Bioenergetic Function in Ovariectomized Rats. Int J Mol Sci 2024; 26:266. [PMID: 39796122 PMCID: PMC11720031 DOI: 10.3390/ijms26010266] [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: 10/16/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Menopause occurs due to the depletion of the ovarian reserve, leading to a progressive decline in estrogen (E2) levels. This decrease in E2 levels increases the risk of developing several diseases and can coexist with chronic kidney disease (CKD). Arterial hypertension (AH) is another condition associated with menopause and may either contribute to or result from CKD. Ovariectomy (OVX) induces hypoestrogenism, which can lead to mitochondrial bioenergetic dysfunction in the kidneys. Previous studies have suggested that exercise training has beneficial effects on adults with CKD and AH. To investigate the effects of OVX and resistance training (RT) on hemodynamic parameters and mitochondrial bioenergetic function of the kidney, female Wistar rats were divided into ovariectomized (OVX) and intact (INT) groups. These rats were either kept sedentary (SED) or subjected to RT for thirteen weeks. The RT involved climbing a vertical ladder with a workload apparatus. Hemodynamic parameters were assessed via tail plethysmography. Mitochondrial respiratory function was evaluated with high-resolution respirometry. Gene expression related to the electron transport chain (ETC) and oxidative phosphorylation (OXPHOS) was evaluated by real-time qPCR. At week 13, key hemodynamic parameters (systolic blood pressure and mean arterial pressure) were significantly elevated in the OVX-SED group. Compared with those in the other groups, mitochondrial bioenergetics were impaired in the OVX-SED group. In contrast, the trained groups presented improved mitochondrial bioenergetic function compared with the sedentary groups. OVX led to reduced gene expression related to the mitochondrial ETC and OXPHOS, whereas RT both prevented this reduction and increased gene expression in the trained groups. Our results indicate that hypoestrogenism significantly decreases OXPHOS and ETC capacity in the kidneys of sedentary animals. However, RT effectively increased the expression of genes related to mitochondrial ETC and OXPHOS, thereby counteracting the effects of OVX.
Collapse
Affiliation(s)
- Anne L. F. Queiroz
- Department of Physiological Sciences, Interinstitutional Post-Graduate Program of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (A.L.F.Q.); (C.B.G.); (D.F.A.C.); (A.V.A.)
- Post-Graduate Program of Physiotherapy, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil
| | - Christopher B. Garcia
- Department of Physiological Sciences, Interinstitutional Post-Graduate Program of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (A.L.F.Q.); (C.B.G.); (D.F.A.C.); (A.V.A.)
| | - João P. M. O. Silva
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (J.P.M.O.S.); (A.F.C.)
| | - Diego F. A. Cavalini
- Department of Physiological Sciences, Interinstitutional Post-Graduate Program of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (A.L.F.Q.); (C.B.G.); (D.F.A.C.); (A.V.A.)
| | - André V. Alexandrino
- Department of Physiological Sciences, Interinstitutional Post-Graduate Program of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (A.L.F.Q.); (C.B.G.); (D.F.A.C.); (A.V.A.)
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (J.P.M.O.S.); (A.F.C.)
- Department of Biological Sicences, Central Paulista University Center (UNICEP), Campus São Carlos, São Carlos 13.570-300, SP, Brazil
| | - Anderson F. Cunha
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (J.P.M.O.S.); (A.F.C.)
| | - Anibal E. Vercesi
- Department of Pathology, University of Campinas (UNICAMP), Campinas 13.083-970, SP, Brazil; (A.E.V.); (R.F.C.)
| | - Roger F. Castilho
- Department of Pathology, University of Campinas (UNICAMP), Campinas 13.083-970, SP, Brazil; (A.E.V.); (R.F.C.)
| | - Gilberto E. Shiguemoto
- Department of Physiological Sciences, Interinstitutional Post-Graduate Program of Physiological Sciences, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil; (A.L.F.Q.); (C.B.G.); (D.F.A.C.); (A.V.A.)
- Post-Graduate Program of Physiotherapy, Federal University of São Carlos (UFSCar), São Carlos 13.566-490, SP, Brazil
| |
Collapse
|
17
|
Lu YJ, Wang HK, Lin YH. The interplay between sex hormones, mitochondrial dysfunction, and intervertebral disc degeneration: insights beyond Qiu et al. J Transl Med 2024; 22:1162. [PMID: 39741256 DOI: 10.1186/s12967-024-06026-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/02/2025] Open
Affiliation(s)
- Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, 5 Fu-Shing Street, Kweishan, Taoyuan, 333, Taiwan
- School of Medicine, Chang Gung University, Kwei-shan, Tao-Yuan, Taiwan
| | - Hong Kai Wang
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, 5 Fu-Shing Street, Kweishan, Taoyuan, 333, Taiwan
- School of Medicine, Chang Gung University, Kwei-shan, Tao-Yuan, Taiwan
| | - Yu-Hsiang Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, 5 Fu-Shing Street, Kweishan, Taoyuan, 333, Taiwan.
- Department of Urology, Chang Gung Memorial Hospital at Linkou, 5 Fu-Shing Street, Kweishan, Taoyuan, 333, Taiwan.
| |
Collapse
|
18
|
Lei J, Zhu Q, Guo J, Chen J, Qi L, Cui M, Jiang Z, Fan C, Wang L, Lai T, Jin Y, Si L, Liu Y, Yang Q, Bao D, Guo R. TEFM facilitates uterine corpus endometrial carcinoma progression by activating ROS-NFκB pathway. J Transl Med 2024; 22:1151. [PMID: 39731053 DOI: 10.1186/s12967-024-05833-0] [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: 09/09/2024] [Accepted: 10/31/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Mitochondrial transcription elongation factor (TEFM) is a recently discovered factor involved in mitochondrial DNA replication and transcription. Previous studies have reported that abnormal TEFM expression can disrupt the assembly of mitochondrial respiratory chain and thus mitochondrial function. However, the role of TEFM on Uterine corpus endometrial carcinoma (UCEC) progression remains unclear. The present study aims to investigate the expression of TEFM in tumor tissue of UCEC and the effect of abnormal TEFM expression on malignant phenotype of UCEC cells. METHODS The expressions of TEFM were measured in tumor tissues and cell lines of UCEC by immunohistochemistry, Western blotting, and real-time quantitative PCR assays. Besides, the effects of TEFM knockdown or overexpression on UCEC cell growth, metastasis, apoptosis, and autophagy were also determined using EdU, colony formation, flow cytometry, TUNEL, and transmission electron microscopy assays. Xenograft model was used to confirm the role of TEFM on proliferative potential of UECE cells in vivo. RESULTS Our bioinformatics analysis of CPTAC data showed that TEFM is abnormally overexpressed in UCEC and its upregulation was significantly associated with poor survival of patients with UCEC. We found that TEFM upregulation significantly promoted the growth and metastasis of UCEC cells. Mechanically, TEFM upregulation impaired the function of mitochondria, decreased their membrane potential and activated the AKT-NFκB pathway by promoting reactive oxygen species (ROS) production, leading to enhanced intracellular autophagy and thus UCEC growth and metastasis. CONCLUSION This study demonstrates that TEFM positively regulates autophagy to promote the growth and metastasis of UCEC cells, which provides a potential prognostic biomarker and therapeutic target for the treatment of UCEC.
Collapse
Affiliation(s)
- Jia Lei
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Radiotheraphy Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Qingguo Zhu
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Jianghao Guo
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Jiaxing Chen
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Lixia Qi
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Mengmeng Cui
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Zhixiong Jiang
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Chunhui Fan
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Lin Wang
- Radiotheraphy Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Tianjiao Lai
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Yuxi Jin
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Lulu Si
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Yana Liu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China
| | - Qi Yang
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China.
- School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Dengke Bao
- Laboratory of Cancer Biomarkers and Liquid Biopsy, School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China.
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, 475004, China.
| | - Ruixia Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Henan Key Medical Laboratory for the Prevention and Treatment of Gynecological Malignant Tumors, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
19
|
Sarver DC, Saqib M, Chen F, Wong GW. Mitochondrial respiration atlas reveals differential changes in mitochondrial function across sex and age. eLife 2024; 13:RP96926. [PMID: 39704485 PMCID: PMC11661797 DOI: 10.7554/elife.96926] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Organ function declines with age, and large-scale transcriptomic analyses have highlighted differential aging trajectories across tissues. The mechanism underlying shared and organ-selective functional changes across the lifespan, however, still remains poorly understood. Given the central role of mitochondria in powering cellular processes needed to maintain tissue health, we therefore undertook a systematic assessment of respiratory activity across 33 different tissues in young (2.5 months) and old (20 months) mice of both sexes. Our high-resolution mitochondrial respiration atlas reveals: (1) within any group of mice, mitochondrial activity varies widely across tissues, with the highest values consistently seen in heart, brown fat, and kidney; (2) biological sex is a significant but minor contributor to mitochondrial respiration, and its contributions are tissue-specific, with major differences seen in the pancreas, stomach, and white adipose tissue; (3) age is a dominant factor affecting mitochondrial activity, especially across most brain regions, different fat depots, skeletal muscle groups, eyes, and different regions of the gastrointestinal tract; (4) age effects can be sex- and tissue-specific, with some of the largest effects seen in pancreas, heart, adipose tissue, and skeletal muscle; and (5) while aging alters the functional trajectories of mitochondria in a majority of tissues, some are remarkably resilient to age-induced changes. Altogether, our data provide the most comprehensive compendium of mitochondrial respiration and illuminate functional signatures of aging across diverse tissues and organ systems.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Fangluo Chen
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Center for Metabolism and Obesity Research, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
20
|
Gowayed MA, Zakaraya ZZ, Abu-Samra N, Elhamammy RH, Abdel Moneim LM, Hafez HA, Moneam IA, Oriquat GA, Kamel MA. Crosstalk between mitochondrial homeostasis and AMPK pathway mediate the receptor-mediated cardioprotective effects of estradiol in ovariectomized female rats. PLoS One 2024; 19:e0312397. [PMID: 39693325 DOI: 10.1371/journal.pone.0312397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/04/2024] [Indexed: 12/20/2024] Open
Abstract
Estrogen (E2) deficiency is a risk factor for cardiovascular disease (CVD), however, the exact mechanism for the E2 protective effect on CVD remains unclear. This study aimed to investigate the estrogen receptor (ER) and non-receptor mediated effects of E2 treatment on the cardiac expression of adenosine monophosphate-dependent protein kinase (AMPK), autophagic, mitophagy and mitochondrial homeostasis-regulating genes in ovariectomized (OVX) rats. Female rats were divided into two main groups; sham and bilaterally OVX rats, then each group was subdivided into four subgroups according to treatment; untreated, subcutaneously treated with E2 (30 μg/kg), or Fulvestrant (F) (5 mg/Kg), or a combination of both drugs for 28 days. The OVX rats or F-treated sham rats showed dyslipidemia, and marked disturbances in parameters of AMPK signaling, autophagy, mitophagy, mitochondrial fission, fusion and biogenesis. E2 administration to OVX or F-treated sham rats has corrected the disturbed lipid and cardiac profiles, increased AMPK, and restored the balance of cardiac autophagy, mitophagy, and mitochondrial dynamics and homeostasis. Most of these effects in OVX rats were blocked by the ER antagonist (F). Estrogen treatment has cardioprotective effects in OVX females through modulating cardiac mitochondrial homeostasis, mitophagy and autophagy and restoring the AMPK signaling pathway. As witnessed by Fulvestrant, these effects suggest the main role of ER-mediated signaling in regulating mitophagy and plasma and cardiac lipids along with the existence of a post-translational control mechanism or the involvement of estrogenic non-receptor pathway controlling the postmenopausal cardiac mitochondrial energy production machinery that needs further investigation.
Collapse
Affiliation(s)
- Mennatallah A Gowayed
- Department of Pharmacology &Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Zainab Zaki Zakaraya
- Biopharmaceutics and Clinical Pharmacy Department, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Nehal Abu-Samra
- Department of Basic Sciences, Faculty of Physical Therapy, Pharos University in Alexandria, Alexandria, Egypt
| | - Reem H Elhamammy
- Department of Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Lobna M Abdel Moneim
- Department of Pharmacology &Therapeutics, Faculty of Pharmacy and Drug Manufacturing, Pharos University in Alexandria, Alexandria, Egypt
| | - Hala A Hafez
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ihab A Moneam
- Clinical Laboratory Sciences Department, College of Pharmacy, Almaaqal University, Basra, Iraq
- Supplementary General Sciences Department, Faculty of Dentistry, Future University, New Cairo, Egypt
| | - Ghaleb A Oriquat
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
21
|
Dos Santos BG, Brisnovali NF, Goedeke L. Biochemical basis and therapeutic potential of mitochondrial uncoupling in cardiometabolic syndrome. Biochem J 2024; 481:1831-1854. [PMID: 39630236 DOI: 10.1042/bcj20240005] [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: 08/05/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
Mild uncoupling of oxidative phosphorylation is an intrinsic property of all mitochondria, allowing for adjustments in cellular energy metabolism to maintain metabolic homeostasis. Small molecule uncouplers have been extensively studied for their potential to increase metabolic rate, and recent research has focused on developing safe and effective mitochondrial uncoupling agents for the treatment of obesity and cardiometabolic syndrome (CMS). Here, we provide a brief overview of CMS and cover the recent mechanisms by which chemical uncouplers regulate CMS-associated risk-factors and comorbidities, including dyslipidemia, insulin resistance, steatotic liver disease, type 2 diabetes, and atherosclerosis. Additionally, we review the current landscape of uncoupling agents, focusing on repurposed FDA-approved drugs and compounds in advanced preclinical or early-stage clinical development. Lastly, we discuss recent molecular insights by which chemical uncouplers enhance cellular energy expenditure, highlighting their potential as a new addition to the current CMS drug landscape, and outline several limitations that need to be addressed before these agents can successfully be introduced into clinical practice.
Collapse
Affiliation(s)
- Bernardo Gindri Dos Santos
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| | - Niki F Brisnovali
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| | - Leigh Goedeke
- Department of Medicine (Cardiology), The Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
- Department of Medicine (Endocrinology), The Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, U.S.A
| |
Collapse
|
22
|
Hinton A, Neikirk K, Le H, Harris C, Oliver A, Martin P, Gaye A. Estrogen receptors in mitochondrial metabolism: age-related changes and implications for pregnancy complications. AGING ADVANCES 2024; 1:154-171. [PMID: 39839811 PMCID: PMC11748122 DOI: 10.4103/agingadv.agingadv-d-24-00012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/24/2024] [Indexed: 01/23/2025]
Abstract
Estrogen hormones are primarily associated with their role as female sex hormones responsible for primary and secondary sexual development. Estrogen receptors are known to undergo age-dependent decreases due to age-related changes in hormone production. In the mitochondria, estrogen functions by reducing the production of reactive oxygen species in the electron transport chain, inhibiting apoptosis, and regulating mitochondrial DNA content. Moreover, estrogen receptors may be the key components in maintaining mitochondrial membrane potential and structure. Although estrogen plays a crucial role in the development of pregnancy, our understanding of how estrogen receptors change with aging during pregnancy remains limited. During pregnancy, estrogen levels are significantly elevated, with a corresponding upregulation of estrogen receptors, which play various roles in pregnancy. However, the exact role of estrogen receptors in pregnancy complications remains to be further investigated. The paper reviews the role of estrogen receptors in the regulation of mitochondrial metabolism and in pregnancy complications, with a special focus on the effect of age-related changes on estrogen levels and estrogen receptors function. We also address how estrogen maintains mitochondrial function, including reducing the production of reactive oxygen species in the electron transport chain, inhibiting apoptosis, regulating mitochondrial DNA content, and maintaining mitochondrial membrane potential and structure. However, the effects of estrogen on mitochondria-endoplasmic reticulum contacts have not been well studied. Based on these emergent roles in mitochondria, the differential roles of estrogen receptors in pregnancy complications are of great relevance. The paper emphasizes the association between maternal health and estrogen receptors and indicates the need for future research to elucidate the interdependence of estrogen receptor-regulated maternal health with mitochondrial function and their relationship with the gut microbiome. Overall, we summarize the important role of estrogen receptors during pregnancy and highlight the need for further research to better understand the role of estrogen receptors in aging and pregnancy complications. This not only helps to reveal the mechanism underlying the role of estrogen in maternal health but also has potential clinical implications for the development of new therapies targeting age-related diseases and pregnancy complications.
Collapse
Affiliation(s)
- Antentor Hinton
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Han Le
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Chanel Harris
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Ashton Oliver
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Pamela Martin
- Department of Biomedical Sciences, Meharry Medical College, Nashville, TN, USA
| | - Amadou Gaye
- Department of Integrative Genomics and Epidemiology, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
23
|
Mu E, Gurvich C, Kulkarni J. Estrogen and psychosis - a review and future directions. Arch Womens Ment Health 2024; 27:877-885. [PMID: 38221595 PMCID: PMC11579214 DOI: 10.1007/s00737-023-01409-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/02/2023] [Indexed: 01/16/2024]
Abstract
The link between sex hormones and schizophrenia has been suspected for over a century; however, scientific evidence supporting the pharmacotherapeutic effects of exogenous estrogen has only started to emerge during the past three decades. Accumulating evidence from epidemiological and basic research suggests that estrogen has a protective effect in women vulnerable to schizophrenia. Such evidence has led multiple researchers to investigate the role of estrogen in schizophrenia and its use in treatment. This narrative review provides an overview of the effects of estrogen as well as summarizes the recent work regarding estrogen as a treatment for schizophrenia, particularly the use of new-generation selective estrogen receptor modulators.
Collapse
Affiliation(s)
- Eveline Mu
- HER Centre Australia, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Caroline Gurvich
- HER Centre Australia, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jayashri Kulkarni
- HER Centre Australia, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Lin YH, Lin KJ, Chen JY. The interplay between aging, testosterone, and mitochondrial dysfunction: Revisiting the role of hormonal regulation in urological health. Biomed Pharmacother 2024; 181:117722. [PMID: 39615167 DOI: 10.1016/j.biopha.2024.117722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Affiliation(s)
- Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital, Linkou, Taiwan; School of Medicine, Chang Gung University, TaoYuan 333, Taiwan.
| | - Kou-Jen Lin
- Department of Urology, Chang Gung Memorial Hospital, Linkou, Taiwan; School of Medicine, Chang Gung University, TaoYuan 333, Taiwan
| | - Jau-Yuan Chen
- School of Medicine, Chang Gung University, TaoYuan 333, Taiwan; Department of Family Medicine, Chang-Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
25
|
Zheng Q, Zhang D, Xing J. NRF1-induced mmu_circ_0001388/hsa_circ_0029470 confers ferroptosis resistance in ischemic acute kidney injury via the miR-193b-3p/TCF4/GPX4 axis. Life Sci 2024; 358:123190. [PMID: 39481837 DOI: 10.1016/j.lfs.2024.123190] [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: 08/06/2024] [Revised: 10/16/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
AIMS Circular RNAs (circRNAs) are critical in the progression of ischemic acute kidney injury (AKI). Nevertheless, the specific functions and regulatory pathways of mmu_circ_0001388 and hsa_circ_0029470 remain elusive. METHODS Real-time quantitative polymerase chain reaction (RT-qPCR) was utilized to assess the expression patterns of mmu_circ_0001388, hsa_circ_0029470, and miR-139b-3p. Protein expressions of nuclear respiratory factor 1 (NRF1), transcription factor 4 (TCF4), glutathione peroxidase 4 (GPX4), and Acyl-CoA synthetase long-chain family member 4 (ACSL4) were identified via immunoblotting. Furthermore, the functions and control mechanisms of mmu_circ_003062 and hsa_circ_0075663 were examined via diverse cell and animal studies, encompassing bioinformatics prediction, dual-luciferase reporter (DLR), chromatin immunoprecipitation (ChIP), fluorescence in situ hybridization (FISH), flow cytometry (FCM), hematoxylin and eosin (H&E) staining, dihydroethidium (DHE), TUNEL, immunohistochemistry, and transmission electron microscopy (TEM), and Fe2+ assay. KEY FINDINGS Initially, the induction of mmu_circ_0001388 by NRF1 was observed in vitro and in vivo following ischemia/reperfusion (I/R) injury. Subsequently, knockdown or overexpression of mmu_circ_0001388 was found to either promote or inhibit ferroptosis caused by I/R in Boston University mouse proximal tubule (BUMPT) cells, respectively. From a mechanistic standpoint, mmu_circ_0001388 was found to function as a sponge for miR-193b-3p, which promoted TCF4 and subsequently enhanced GPX4, thereby suppressing ferroptosis. Finally, the overexpression of mmu_circ_0001388 was shown to ameliorate I/R-induced AKI in mice. In parallel, hsa_circ_0029470, homologous to mmu_circ_0001388, demonstrated an identical control pathway in human renal tubular epithelial (HK-2) cells. SIGNIFICANCE The NRF1/mmu_circ_0001388, hsa_circ_0029470/miR-193b-3p/TCF4/GPX4 axis is pivotal in regulating ferroptosis induced by ischemic AKI and holds potential as a therapeutic target.
Collapse
Affiliation(s)
- Qiang Zheng
- Department of Emergency, The First Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Dongshan Zhang
- Department of Emergency, The Second Xiangya Hospital of Central South University, Changsha 410000, Hunan, China; Emergency Medicine and Difficult Diseases Institute, The Second Xiangya Hospital of Central South University, Changsha 410000, Hunan, China; Department of Nephrology, The Second Xiangyi Hospital of Central South University, Changsha 410000, Hunan, China
| | - Jihong Xing
- Department of Emergency, The First Hospital of Jilin University, Changchun 130000, Jilin, China.
| |
Collapse
|
26
|
Zhang C, Feng X, Zhang X, Chen Y, Kong J, Lou Y. Research progress on the correlation between estrogen and estrogen receptor on postmenopausal sarcopenia. Front Endocrinol (Lausanne) 2024; 15:1494972. [PMID: 39640884 PMCID: PMC11617174 DOI: 10.3389/fendo.2024.1494972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Estrogen is a necessary sex steroid and potent neuroprotective hormone. It plays a multifaceted role beyond the reproductive system, extending its influence to the brain, skeletal muscle, and other organs. Estrogen's role in cognition, mood, autonomic regulation, and neuroprotection involves interactions with neurotransmitters, neuromodulators in a distributed manner. Notably, the impact of estrogen on mitochondrial metabolism in skeletal muscle is particularly significant due to a unique modulated bioenergetic profiles, synaptic plasticity, and neuronal health. The deficiency of estrogen in menopause has been linked to changes in brain structure, connectivity, energy metabolism. Therewith, these are crucial factors in cognitive function and the risk of Alzheimer's diseases. Besides, it leads to endocrine and metabolic dysfunction, resulting in osteoporosis, metabolic syndrome, and a tendency toward decreased muscle mass and strength. Estrogen's influence on mitochondrial function is particularly relevant to aging, as it affects the production of ATP and the overall metabolic health of the brain. Estrogen decline in women skeletal muscle mass is usually related to sarcopenia, a prevalent disease observed in vulnerable elderly individuals. Therefore, estrogen is considered to play a crucial role in skeletal muscle homeostasis and motor ability, although the exact mechanism remains unclear. This paper reviews the literature on the impact of estrogen on postmenopausal skeletal muscle diseases and the underlying molecular mechanisms, especially in terms of mitochondrial metabolism. In summary, estrogen plays an important role in the health of skeletal muscle in postmenopausal women, and its impact on mitochondrial function and homeostasis offers potential targets for the development of new strategies to treat sarcopenia.
Collapse
Affiliation(s)
- Chengmei Zhang
- Department of Clinical Nutrition, Shengjing Hospital of China Medical Universty, Shenyang, Liaoning, China
| | - Xin Feng
- Department of Nephrology, Liaoning Electric Power Central Hospital, Shenyang, China
| | - Xue Zhang
- Sheng Jing Hospital Affiliated, China Medical University, Shenyang, Liaoning, China
| | - Yu Chen
- The Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Juan Kong
- Department of Clinical Nutrition, Shengjing Hospital of China Medical Universty, Shenyang, Liaoning, China
| | - Yan Lou
- School of Intelligent Medicine, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
27
|
Wallings RL, McFarland K, Staley HA, Neighbarger N, Schaake S, Brüggemann N, Zittel S, Usnich T, Klein C, Sammler EM, Tansey MG. The R1441C-Lrrk2 mutation induces myeloid immune cell exhaustion in an age- and sex-dependent manner in mice. Sci Transl Med 2024; 16:eadl1535. [PMID: 39504353 DOI: 10.1126/scitranslmed.adl1535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/19/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024]
Abstract
Age is the greatest risk factor for many neurodegenerative diseases, yet immune system aging, a contributor to neurodegeneration, is understudied. Genetic variation in the LRRK2 gene affects risk for both familial and sporadic Parkinson's disease (PD). The leucine-rich repeat kinase 2 (LRRK2) protein is implicated in peripheral immune cell signaling, but the effects of an aging immune system on LRRK2 function remain unclear. We analyzed peritoneal macrophages from R1441C-Lrrk2 knock-in mice and observed a biphasic, age-dependent effect of the R1441C-Lrrk2 mutation on peritoneal macrophage function. We report increases in antigen presentation, anti-inflammatory cytokine production, lysosomal activity, and pathogen uptake in peritoneal macrophages from young (2- to 3-month-old) female R1441C-Lrrk2 mice. Conversely, macrophages from aged (18- to 21-month-old) female R1441C-Lrrk2 mice exhibited decreased antigen presentation after inflammatory insult, decreased lysosomal function, and pathogen uptake, with a concomitant increase in DNA fragmentation in the presence of pathogens. This immune cell exhaustion phenotype was not observed in male R1441C-Lrrk2 mice and was driven by increased LRRK2 protein kinase activity. This phenotype was also observed in human peripheral myeloid cells, with monocyte-derived macrophages from patients with PD who had either the R1441C- or Y1699C-LRRK2 mutation exhibiting decreased pathogen uptake and increased PDL1 expression, consistent with immune cell exhaustion. Our findings that LRRK2 mutations conferred an immunological advantage at a young age but could predispose the carrier to age-acquired immune cell exhaustion have implications for the therapeutic development of LRRK2 inhibitors.
Collapse
Affiliation(s)
- Rebecca L Wallings
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Karen McFarland
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Department of Neurology and Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
- Department of Pharmacology, Emory University, Atlanta, GA, USA
| | - Hannah A Staley
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
| | - Noelle Neighbarger
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
| | - Susen Schaake
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Zittel
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatiana Usnich
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Klein
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Esther M Sammler
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
- Department of Neurology, School of Medicine, Ninewells Hospital, Ninewells Drive, Dundee DD1 9SY, UK
| | - Malú Gámez Tansey
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Neurology and Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| |
Collapse
|
28
|
Lasisi-Sholola AS, Hammed SO, Ajike RA, Akhigbe RE, Afolabi OA. Estrogen replacement therapy reverses spatial memory loss and pyramidal cell neurodegeneration in the prefrontal cortex of lead-exposed ovariectomized Wistar rats. Curr Res Toxicol 2024; 7:100200. [PMID: 39583742 PMCID: PMC11582547 DOI: 10.1016/j.crtox.2024.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/16/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Although menopause is a component of chronological aging, it may be induced by exposure to heavy metals like lead. Interestingly, lead exposure, just like the postmenopausal state, has been associated with spatial memory loss and neurodegeneration; however, the impact of hormone replacement therapy (HRT) on menopause and lead-induced spatial memory loss and neurodegeneration is yet to be reported. AIM The present study investigated the effect and associated mechanism of HRT on ovariectomized-driven menopausal state and lead exposure-induced spatial memory loss and neurodegeneration. MATERIALS AND METHODS Thirty adult female Wistar rats were randomized into 6 groups (n = 5 rats/group); the sham-operated vehicle-treated, ovariectomized (OVX), OVX + HRT, lead-exposed, OVX + lead, and OVX + Lead + HRT groups. Treatment was daily via gavage and lasted for 28 days. RESULTS Ovariectomy and lead exposure impaired spatial memory deficit evidenced by a significant reduction in novel arm entry, time spent in the novel arm, alternation, time exploring novel and familiar objects, and discrimination index. These findings were accompanied by a marked distortion in the histology of the prefrontal cortex, and a decline in serum dopamine level and pyramidal neurons. In addition, ovariectomy and lead exposure induced metabolic disruption (as depicted by a marked rise in lactate level and lactate dehydrogenase and creatinine kinase activities), oxidative stress (evidenced by a significant increase in MDA level, and decrease in GSH level, and SOD and catalase activities), inflammation (as shown by significant upregulation of myeloperoxidase activity, and TNF-α and IL-1β), and apoptosis (evidenced by a rise in caspase 3 activity) of the prefrontal cortex. The observed biochemical and histological perturbations were attenuated by HRT. CONCLUSIONS This study revealed that HRT attenuated ovariectomy and lead-exposure-induced spatial memory deficit and pyramidal neurodegeneration by suppressing oxidative stress, inflammation, and apoptosis of the prefrontal cortex.
Collapse
Affiliation(s)
- Abiodun Shukrat Lasisi-Sholola
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - Sodiq Opeyemi Hammed
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Richard Adedamola Ajike
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Roland Eghoghosoa Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | - Oladele Ayobami Afolabi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
29
|
Guo D, Liu H, Zhao S, Lu X, Wan H, Zhao Y, Liang X, Zhang A, Wu M, Xiao Z, Hu N, Li Z, Xie D. Synergistic rheumatoid arthritis therapy by interrupting the detrimental feedback loop to orchestrate hypoxia M1 macrophage polarization using an enzyme-catalyzed nanoplatform. Bioact Mater 2024; 41:221-238. [PMID: 39149592 PMCID: PMC11324459 DOI: 10.1016/j.bioactmat.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
A detrimental feedback loop between hypoxia and oxidative stress consistently drives macrophage polarization toward a pro-inflammatory M1 phenotype, thus persistently aggravating rheumatoid arthritis (RA) progression. Herein, an enzyme-catalyzed nanoplatform with synergistic hypoxia-relieving and reactive oxygen species (ROS)-scavenging properties was developed using bovine serum albumin-bilirubin-platinum nanoparticles (BSA-BR-Pt NPs). Bilirubin was employed to eliminate ROS, while platinum exhibited a synergistic effect in scavenging ROS and simultaneously generated oxygen. In mice RA model, BSA-BR-Pt NPs treatment exhibited superior effects, resulting in significant improvements in joint inflammation, cartilage damage, and bone erosion, compared to methotrexate, the most widely used antirheumatic drug. Mechanistically, RNA-sequencing data and experimental results elucidated that BSA-BR-Pt NPs induced a re-polarization of hypoxic M1 macrophages to M2 macrophages via switching glycolysis to oxidative phosphorylation through the inhibition of HIF-1α pathway. Collectively, this research for the first time elaborated the underlying mechanism of enzyme-catalyzed nanoplatform in orchestrating macrophage polarization, and identified a novel therapeutic strategy for RA and other inflammatory disorders.
Collapse
Affiliation(s)
- Dong Guo
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Hui Liu
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Sheng Zhao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Xinya Lu
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Haoyu Wan
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Yitao Zhao
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Xinzhi Liang
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Anbiao Zhang
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Mengyuan Wu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China
| | - Zhisheng Xiao
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Zhong Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, NT, Hong Kong, PR China
| | - Denghui Xie
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, PR China
| |
Collapse
|
30
|
Nokoff NJ, Nemkov T, Bothwell S, Cree MG, Fuller KNZ, Keller AC, Kelsey MM, Nadeau KJ, Moreau KL. Differences in cardiorespiratory fitness by gonadotropin-releasing hormone agonist treatment before and after testosterone in transgender adolescents. J Appl Physiol (1985) 2024; 137:1470-1483. [PMID: 39417821 PMCID: PMC11573275 DOI: 10.1152/japplphysiol.00629.2024] [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: 08/14/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
There are known sex differences in cardiorespiratory fitness (CRF). Little is known about the impact of pubertal blockade with a gonadotropin-releasing hormone agonist (GnRHa) followed by hormone therapy on CRF for transgender adolescents. We aimed to 1) determine the effect of GnRHa monotherapy on CRF and mitochondrial function and associations with metabolomic profiles and 2) evaluate changes after 1 and 12 mo of testosterone therapy among transgender adolescents. Participants assigned female at birth (n = 19, baseline age of 15.0 ± 1.0 yr) from two groups: GnRHa+ (n = 8) and GnRHa- (n = 11) were examined at baseline and 1- and 12-mo post-testosterone therapy in a longitudinal observational study to assess cardiorespiratory fitness, mitochondrial respiration, and metabolic profile. Fasted morning labs included assessment of metabolomics and peripheral blood mononuclear cell mitochondrial respiration and degree of mitochondrial coupling (respiratory control ratio, RCR). A graded cycle ergometer test was performed. Baseline differences were evaluated between groups. Changes were compared with mixed linear regression models evaluating time (baseline, 1 mo, and 12 mo), group (GnRHa treatment yes/no), and their interaction. At baseline GnRHa+ individuals had higher relative V̇o2peak (30.1 ± 4.83 vs. 25.24 ± 4.47 mL/kg/min, P = 0.042) than GnRHa- individuals. In regression models, GnRHa+ individuals had a significant increase in peak watts (P = 0.011) and total exercise time (P = 0.005) after 12 mo of testosterone (P = 0.012) but not GnRHa- individuals. GnRHa+ individuals have significantly higher RCR under carbohydrate (P = 0.0007) and lipid (P = 0.0002) conditions than GnRHa+ individuals. Pretreatment with GnRHa positively influences peak CRF and mitochondrial respiration in adolescent transgender males undergoing testosterone therapy.NEW & NOTEWORTHY This study demonstrates differences in exercise capacity and mitochondrial respiration at baseline based on whether or not individuals had feminizing puberty blocked. Individuals who had puberty blocked had greater improvements in cardiopulmonary exercise testing parameters after 12 mo of testosterone than those who went through feminizing puberty.
Collapse
Affiliation(s)
- Natalie J Nokoff
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Ludeman Family Center for Women's Health, Aurora, Colorado, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Samantha Bothwell
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Melanie G Cree
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Ludeman Family Center for Women's Health, Aurora, Colorado, United States
| | - Kelly N Z Fuller
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Amy C Keller
- Division of Endocrinology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Geriatric Research Education and Clinical Center, Veterans Affairs Eastern Colorado, Aurora, Colorado, United States
| | - Megan M Kelsey
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Ludeman Family Center for Women's Health, Aurora, Colorado, United States
| | - Kristen J Nadeau
- Division of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Ludeman Family Center for Women's Health, Aurora, Colorado, United States
| | - Kerrie L Moreau
- Ludeman Family Center for Women's Health, Aurora, Colorado, United States
- Division of Geriatrics, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
31
|
Guo Z, Tian Y, Gao J, Zhou B, Zhou X, Chang X, Zhou H. Enhancement of Mitochondrial Homeostasis: A Novel Approach to Attenuate Hypoxic Myocardial Injury. Int J Med Sci 2024; 21:2897-2911. [PMID: 39628681 PMCID: PMC11610329 DOI: 10.7150/ijms.103986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/18/2024] [Indexed: 12/06/2024] Open
Abstract
Cardiomyocytes are highly oxygen-dependent cells, relying on oxygen-driven oxidative phosphorylation to maintain their function. During hypoxia, mitochondrial ATP production decreases, leading to calcium overload, acidosis, and oxidative stress, which collectively trigger myocardial injury. Ischemic heart disease, caused by coronary atherosclerosis, results in myocardial ischemia and hypoxia, leading to ischemia-reperfusion (I/R) injury. Early myocardial injury is attributed to ischemia and hypoxia, but even after thrombolytic therapy, interventional surgery, or coronary artery bypass grafting (CABG) restores local blood flow and oxygen supply, myocardial reperfusion injury (I/R) may still occur. Mitochondria, often referred to as the "powerhouses" of the cell, play a crucial role in cellular energy production. In the early stages of ischemia and hypoxia, mitochondrial dysfunction disrupts mitochondrial homeostasis, causing the accumulation of unfolded or misfolded proteins in the mitochondria. This activates the mitochondrial unfolded protein response (mtUPR) and mitophagy, which work to clear damaged proteins and mitochondria, playing a key role during this period. This review focuses on mitochondrial mechanisms during the ischemic phase of ischemia-reperfusion injury, aiming to provide new theoretical foundations and potential therapeutic strategies to reduce myocardial damage.
Collapse
Affiliation(s)
- Zhijiang Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yingjie Tian
- Beijing University of Chinese Medicine, Beijing, 100028, China
| | - Jing Gao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Bei Zhou
- Center for Drug Evaluation, National Medical Products Administration, Beijing, China
| | - XiuTeng Zhou
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences/State Key Laboratory for Quality Assurance and Sustainable Use of Dao-di Herbs, Beijing, 100700, China
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
32
|
Antelo-Cea DA, Martínez-Rojas L, Cabrerizo-Ibáñez I, Roudi Rashtabady A, Hernández-Alvarez MI. Regulation of Mitochondrial and Peroxisomal Metabolism in Female Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:11237. [PMID: 39457018 PMCID: PMC11508381 DOI: 10.3390/ijms252011237] [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: 10/03/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Obesity and type 2 diabetes (T2D) are widespread metabolic disorders that significantly impact global health today, affecting approximately 17% of adults worldwide with obesity and 9.3% with T2D. Both conditions are closely linked to disruptions in lipid metabolism, where peroxisomes play a pivotal role. Mitochondria and peroxisomes are vital organelles responsible for lipid and energy regulation, including the β-oxidation and oxidation of very long-chain fatty acids (VLCFAs), cholesterol biosynthesis, and bile acid metabolism. These processes are significantly influenced by estrogens, highlighting the interplay between these organelles' function and hormonal regulation in the development and progression of metabolic diseases, such as obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and T2D. Estrogens modulate lipid metabolism through interactions with nuclear receptors, like peroxisome proliferator-activated receptors (PPARs), which are crucial for maintaining metabolic balance. Estrogen deficiency, such as in postmenopausal women, impairs PPAR regulation, leading to lipid accumulation and increased risk of metabolic disorders. The disruption of peroxisomal-mitochondrial function and estrogen regulation exacerbates lipid imbalances, contributing to insulin resistance and ROS accumulation. This review emphasizes the critical role of these organelles and estrogens in lipid metabolism and their implications for metabolic health, suggesting that therapeutic strategies, including hormone replacement therapy, may offer potential benefits in treating and preventing metabolic diseases.
Collapse
Affiliation(s)
- Damián A. Antelo-Cea
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - Laura Martínez-Rojas
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Izan Cabrerizo-Ibáñez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
| | - Ayda Roudi Rashtabady
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - María Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; (D.A.A.-C.); (L.M.-R.); (I.C.-I.); (A.R.R.)
- IBUB Universitat de Barcelona—Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
33
|
Corti G, Kim J, Enguita FJ, Guarnieri JW, Grossman LI, Costes SV, Fuentealba M, Scott RT, Magrini A, Sanders LM, Singh K, Sen CK, Juran CM, Paul AM, Furman D, Calleja-Agius J, Mason CE, Galeano D, Bottini M, Beheshti A. To boldly go where no microRNAs have gone before: spaceflight impact on risk for small-for-gestational-age infants. Commun Biol 2024; 7:1268. [PMID: 39369042 PMCID: PMC11455966 DOI: 10.1038/s42003-024-06944-6] [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: 04/03/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024] Open
Abstract
In the era of renewed space exploration, comprehending the effects of the space environment on human health, particularly for deep space missions, is crucial. While extensive research exists on the impacts of spaceflight, there is a gap regarding female reproductive risks. We hypothesize that space stressors could have enduring effects on female health, potentially increasing risks for future pregnancies upon return to Earth, particularly related to small-for-gestational-age (SGA) fetuses. To address this, we identify a shared microRNA (miRNA) signature between SGA and the space environment, conserved across humans and mice. These miRNAs target genes and pathways relevant to diseases and development. Employing a machine learning approach, we identify potential FDA-approved drugs to mitigate these risks, including estrogen and progesterone receptor antagonists, vitamin D receptor antagonists, and DNA polymerase inhibitors. This study underscores potential pregnancy-related health risks for female astronauts and proposes pharmaceutical interventions to counteract the impact of space travel on female health.
Collapse
Affiliation(s)
- Giada Corti
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - JangKeun Kim
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Francisco J Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joseph W Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lawrence I Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | | | - Ryan T Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Andrea Magrini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Lauren M Sanders
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Kanhaiya Singh
- McGowan Institute for Regenerative Medicine and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chandan K Sen
- McGowan Institute for Regenerative Medicine and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cassandra M Juran
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, USA
| | - Amber M Paul
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA
| | - Jean Calleja-Agius
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Diego Galeano
- Facultad de Ingeniería, Universidad Nacional de Asunción, MF9M + 958, San Lorenzo, Paraguay
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- Sanford Children's Health Research Center, Sanford Burnham Prebys, La Jolla, CA, USA
| | - Afshin Beheshti
- McGowan Institute for Regenerative Medicine and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Space Biomedicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
34
|
Rubtsova MP, Nikishin DA, Vyssokikh MY, Koriagina MS, Vasiliev AV, Dontsova OA. Telomere Reprogramming and Cellular Metabolism: Is There a Link? Int J Mol Sci 2024; 25:10500. [PMID: 39408829 PMCID: PMC11476947 DOI: 10.3390/ijms251910500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Telomeres-special DNA-protein structures at the ends of linear eukaryotic chromosomes-define the proliferation potential of cells. Extremely short telomeres promote a DNA damage response and cell death to eliminate cells that may have accumulated mutations after multiple divisions. However, telomere elongation is associated with the increased proliferative potential of specific cell types, such as stem and germ cells. This elongation can be permanent in these cells and is activated temporally during immune response activation and regeneration processes. The activation of telomere lengthening mechanisms is coupled with increased proliferation and the cells' need for energy and building resources. To obtain the necessary nutrients, cells are capable of finely regulating energy production and consumption, switching between catabolic and anabolic processes. In this review, we focused on the interconnection between metabolism programs and telomere lengthening mechanisms during programmed activation of proliferation, such as in germ cell maturation, early embryonic development, neoplastic lesion growth, and immune response activation. It is generally accepted that telomere disturbance influences biological processes and promotes dysfunctionality. Here, we propose that metabolic conditions within proliferating cells should be involved in regulating telomere lengthening mechanisms, and telomere length may serve as a marker of defects in cellular functionality. We propose that it is possible to reprogram metabolism in order to regulate the telomere length and proliferative activity of cells, which may be important for the development of approaches to regeneration, immune response modulation, and cancer therapy. However, further investigations in this area are necessary to improve the understanding and manipulation of the molecular mechanisms involved in the regulation of proliferation, metabolism, and aging.
Collapse
Affiliation(s)
- Maria P. Rubtsova
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
| | - Denis A. Nikishin
- Department of Embryology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (D.A.N.); (A.V.V.)
| | - Mikhail Y. Vyssokikh
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia;
| | - Maria S. Koriagina
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
| | - Andrey V. Vasiliev
- Department of Embryology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (D.A.N.); (A.V.V.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Olga A. Dontsova
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia;
- Skolkovo Institute of Science and Technology, Center for Molecular and Cellular Biology, Moscow 121205, Russia
| |
Collapse
|
35
|
Mauvais-Jarvis F, Lindsey SH. Metabolic benefits afforded by estradiol and testosterone in both sexes: clinical considerations. J Clin Invest 2024; 134:e180073. [PMID: 39225098 PMCID: PMC11364390 DOI: 10.1172/jci180073] [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] [Indexed: 09/04/2024] Open
Abstract
Testosterone (T) and 17β-estradiol (E2) are produced in male and female humans and are potent metabolic regulators in both sexes. When E2 and T production stops or decreases during aging, metabolic dysfunction develops and promotes degenerative metabolic and vascular disease. Here, we discuss the shared benefits afforded by E2 and T for metabolic function human females and males. In females, E2 is central to bone and vascular health, subcutaneous adipose tissue distribution, skeletal muscle insulin sensitivity, antiinflammatory immune function, and mitochondrial health. However, T also plays a role in female skeletal, vascular, and metabolic health. In males, T's conversion to E2 is fundamental to bone and vascular health, as well as prevention of excess visceral adiposity and the promotion of insulin sensitivity via activation of the estrogen receptors. However, T and its metabolite dihydrotestosterone also prevent excess visceral adiposity and promote skeletal muscle growth and insulin sensitivity via activation of the androgen receptor. In conclusion, T and E2 are produced in both sexes at sex-specific concentrations and provide similar and potent metabolic benefits. Optimizing levels of both hormones may be beneficial to protect patients from cardiometabolic disease and frailty during aging, which requires further study.
Collapse
Affiliation(s)
- Franck Mauvais-Jarvis
- Medicine Service, Section of Endocrinology, Hormone Therapy Clinic, Southeast Louisiana VA Medical Center, New Orleans, Louisiana, USA
- Deming Department of Medicine, Section of Endocrinology and Metabolism, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, Louisiana, USA
| | - Sarah H. Lindsey
- Tulane Center of Excellence in Sex-Based Biology & Medicine, New Orleans, Louisiana, USA
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
36
|
Smith LC, Abramova E, Vayas K, Rodriguez J, Gelfand-Titiyevksiy B, Roepke TA, Laskin JD, Gow AJ, Laskin DL. Transcriptional profiling of lung macrophages following ozone exposure in mice identifies signaling pathways regulating immunometabolic activation. Toxicol Sci 2024; 201:103-117. [PMID: 38897669 PMCID: PMC11347782 DOI: 10.1093/toxsci/kfae081] [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] [Indexed: 06/21/2024] Open
Abstract
Macrophages play a key role in ozone-induced lung injury by regulating both the initiation and resolution of inflammation. These distinct activities are mediated by pro-inflammatory and anti-inflammatory/proresolution macrophages which sequentially accumulate in injured tissues. Macrophage activation is dependent, in part, on intracellular metabolism. Herein, we used RNA-sequencing (seq) to identify signaling pathways regulating macrophage immunometabolic activity following exposure of mice to ozone (0.8 ppm, 3 h) or air control. Analysis of lung macrophages using an Agilent Seahorse showed that inhalation of ozone increased macrophage glycolytic activity and oxidative phosphorylation at 24 and 72 h post-exposure. An increase in the percentage of macrophages in S phase of the cell cycle was observed 24 h post ozone. RNA-seq revealed significant enrichment of pathways involved in innate immune signaling and cytokine production among differentially expressed genes at both 24 and 72 h after ozone, whereas pathways involved in cell cycle regulation were upregulated at 24 h and intracellular metabolism at 72 h. An interaction network analysis identified tumor suppressor 53 (TP53), E2F family of transcription factors (E2Fs), cyclin-dependent kinase inhibitor 1A (CDKN1a/p21), and cyclin D1 (CCND1) as upstream regulators of cell cycle pathways at 24 h and TP53, nuclear receptor subfamily 4 group a member 1 (NR4A1/Nur77), and estrogen receptor alpha (ESR1/ERα) as central upstream regulators of mitochondrial respiration pathways at 72 h. To assess whether ERα regulates metabolic activity, we used ERα-/- mice. In both air and ozone-exposed mice, loss of ERα resulted in increases in glycolytic capacity and glycolytic reserve in lung macrophages with no effect on mitochondrial oxidative phosphorylation. Taken together, these results highlight the complex interaction between cell cycle, intracellular metabolism, and macrophage activation which may be important in the initiation and resolution of inflammation following ozone exposure.
Collapse
Affiliation(s)
- Ley Cody Smith
- Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT 06269, United States
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Elena Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Kinal Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Jessica Rodriguez
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Benjamin Gelfand-Titiyevksiy
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ 08854, United States
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| |
Collapse
|
37
|
Ihalainen JK, Mikkonen RS, Ackerman KE, Heikura IA, Mjøsund K, Valtonen M, Hackney AC. Beyond Menstrual Dysfunction: Does Altered Endocrine Function Caused by Problematic Low Energy Availability Impair Health and Sports Performance in Female Athletes? Sports Med 2024; 54:2267-2289. [PMID: 38995599 PMCID: PMC11393114 DOI: 10.1007/s40279-024-02065-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/13/2024]
Abstract
Low energy availability, particularly when problematic (i.e., prolonged and/or severe), has numerous negative consequences for health and sports performance as characterized in relative energy deficiency in sport. These consequences may be driven by disturbances in endocrine function, although scientific evidence clearly linking endocrine dysfunction to decreased sports performance and blunted or diminished training adaptations is limited. We describe how low energy availability-induced changes in sex hormones manifest as menstrual dysfunction and accompanying hormonal dysfunction in other endocrine axes that lead to adverse health outcomes, including negative bone health, impaired metabolic activity, undesired outcomes for body composition, altered immune response, problematic cardiovascular outcomes, iron deficiency, as well as impaired endurance performance and force production, all of which ultimately may influence athlete health and performance. Where identifiable menstrual dysfunction indicates hypothalamic-pituitary-ovarian axis dysfunction, concomitant disturbances in other hormonal axes and their impact on the athlete's health and sports performance must be recognized as well. Given that the margin between podium positions and "losing" in competitive sports can be very small, several important questions regarding low energy availability, endocrinology, and the mechanisms behind impaired training adaptations and sports performance have yet to be explored.
Collapse
Affiliation(s)
- Johanna K Ihalainen
- Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, PO Box 35, 40014, Jyväskylä, Finland.
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland.
| | - Ritva S Mikkonen
- Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, PO Box 35, 40014, Jyväskylä, Finland
- Sports Technology Unit, Faculty of Sport and Health Sciences, University of Jyväskylä, Vuokatti, Finland
| | - Kathryn E Ackerman
- Wu Tsai Female Athlete Program, Division of Sports Medicine, Boston Children's Hospital, Boston, MA, USA
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ida A Heikura
- Canadian Sport Institute-Pacific, Victoria, BC, Canada
- Exercise Science, Physical and Health Education, University of Victoria, Victoria, BC, Canada
| | - Katja Mjøsund
- Paavo Nurmi Centre and Unit for Health and Physical Activity, University of Turku, Turku, Finland
- National Olympic Training Centre Helsinki, Helsinki, Finland
| | - Maarit Valtonen
- Finnish Institute of High Performance Sport KIHU, Jyväskylä, Finland
| | - Anthony C Hackney
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
38
|
De Vos K, Mavrogiannis A, Wolters JC, Schlenner S, Wierda K, Cortés Calabuig Á, Chinnaraj R, Dermesrobian V, Armoudjian Y, Jacquemyn M, Corthout N, Daelemans D, Annaert P. Tankyrase1/2 inhibitor XAV-939 reverts EMT and suggests that PARylation partially regulates aerobic activities in human hepatocytes and HepG2 cells. Biochem Pharmacol 2024; 227:116445. [PMID: 39053638 DOI: 10.1016/j.bcp.2024.116445] [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: 12/06/2023] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
The maintenance of a highly functional metabolic epithelium in vitro is challenging. Metabolic impairments in primary human hepatocytes (PHHs) over time is primarily due to epithelial-to-mesenchymal transitioning (EMT). The immature hepatoma cell line HepG2 was used as an in vitro model to explore strategies for enhancing the hepatic phenotype. The phenotypic characterization includes measuring the urea cycle, lipid storage, tricarboxylic acid-related metabolites, reactive oxygen species, endoplasmic reticulum calcium efflux, mitochondrial membrane potentials, oxygen consumptions rate, and CYP450 biotransformation capacity. Expression studies were performed with transcriptomics, co-immunoprecipitation and proteomics. CRISPR/Cas9 was also employed to genetically engineer HepG2 cells. After confirming that PHHs develop an EMT phenotype, expression of tankyrase1/2 was found to increase over time. EMT was reverted when blocking tankyrases1/2-dependent poly-ADP-ribosylation (PARylation) activity, by biochemical and genetic perturbation. Wnt/β-catenin inhibitor XAV-939 blocks tankyrase1/2 and treatment elevated several oxygen-consuming reactions (electron-transport chain, OXHPOS, CYP450 mono-oxidase activity, phase I/II xenobiotic biotransformation, and prandial turnover), suggesting that cell metabolism was enhanced. Glutathione-dependent redox homeostasis was also significantly improved in the XAV-939 condition. Oxygen consumption rate and proteomics experiments in tankyrase1/2 double knockout HepG2 cells then uncovered PARylation as master regulator of aerobic-dependent cell respiration. Furthermore, novel tankyrase1/2-dependent PARylation targets, including mitochondrial DLST, and OGDH, were revealed. This work exposed a new mechanistic framework by linking PARylation to respiration and metabolism, thereby broadening the current understanding that underlies these vital processes. XAV-939 poses an immediate and straightforward strategy to improve aerobic activities, and metabolism, in (immature) cell cultures.
Collapse
Affiliation(s)
- Kristof De Vos
- Laboratory of Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Adamantios Mavrogiannis
- Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Justina Clarinda Wolters
- Section Systems Medicine of Metabolism and Signaling, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, the Netherlands
| | - Susan Schlenner
- Adaptive Immunology, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Electrophysiology Unit, VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium
| | | | - Reena Chinnaraj
- KU Leuven Flow and Mass Cytometry Facility, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Vera Dermesrobian
- KU Leuven Flow and Mass Cytometry Facility, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | | | - Maarten Jacquemyn
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute, 3000 Leuven, Belgium
| | - Nikky Corthout
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; VIB Bio Imaging Core, 3000 Leuven, Belgium
| | - Dirk Daelemans
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute, 3000 Leuven, Belgium
| | - Pieter Annaert
- Laboratory of Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; BioNotus GCV, 2845 Niel, Belgium.
| |
Collapse
|
39
|
Tang H, Kong Q, Zhang Z, Wu W, Yuan L, Liu X. Regulation of transcription factor function by purinergic signalling in cardiovascular diseases. Purinergic Signal 2024:10.1007/s11302-024-10045-8. [PMID: 39215950 DOI: 10.1007/s11302-024-10045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular diseases (CVDs), including hypertension, atherosclerosis, myocardial ischemia, and myocardial infarction, constitute the primary cause of mortality worldwide. Transcription factors play critical roles in the development of CVDs and contribute to the pathophysiology of these diseases by coordinating the transcription of many genes involved in inflammation, oxidative stress, angiogenesis, and glycolytic metabolism. One important regulator of hemostasis in both healthy and pathological settings has been identified as a purinergic signalling pathway. Research has demonstrated that several signalling networks implicated in the pathophysiology of CVDs are formed by transcription factors that are regulated by purinergic substances. Here, we briefly summarize the roles and mechanisms of the transcription factors regulated by purinergic pathways in various types of CVD. This information will be essential for discovering novel approaches for CVD treatment and prevention.
Collapse
Affiliation(s)
- Hao Tang
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qihang Kong
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhewei Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wenchao Wu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Lixing Yuan
- Public Laboratory of West China Second University Hospital and Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Sichuan University, Chengdu, 610041, China.
| | - Xiaojing Liu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
40
|
Rieder AS, Ramires Júnior OV, Prauchner GRK, Wyse ATS. Effects of methylphenidate on mitochondrial dynamics and bioenergetics in the prefrontal cortex of juvenile rats are sex-dependent. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111057. [PMID: 38880464 DOI: 10.1016/j.pnpbp.2024.111057] [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: 12/07/2023] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
Methylphenidate (MPH) is a central nervous system stimulant drug and a first order prescription in the treatment of Attention Deficit Hyperactivity Disorder (ADHD). Although MPH biochemistry in neurodevelopment is not completely understood, studies showed it alters energy metabolism in rat brains. ADHD prevalence during neurodevelopment is related to males and the investigation has been mainly done in these subjects, therefore, little is known about MPH action in females and, consequently, about sexual dimorphism. In the present study we evaluated markers of mitochondrial dynamics (DRP1 and MFN2, fission and fusion, respectively), biogenesis (mtTFA) and bioenergetics (respiratory chain complexes) in prefrontal cortex of male and female juvenile rats submitted to exposure to MPH to better understand MPH effect during postnatal neurodevelopment. ATP and oxidative stress levels were also evaluated. Wistar rats received intraperitoneal injection of MPH (2.0 mg/kg) or control (saline), once a day, from 15th to 45th day of age. Results showed that MPH increased DRP1 and decreased MFN2, as well as increased mtTFA in prefrontal cortex of male rats. In female, MPH decreased NRF1 and increased Parkin, which are mitochondrial regulatory proteins. Respiratory chain complexes (complex I, SDH, complexes III and IV), ATP production and oxidative stress parameters were altered and shown to be sex-dependent. Taken together, results suggest that chronic MPH exposure at an early age in healthy animals changes mitochondrial dynamics, biogenesis and bioenergetics differently depending on the sex of the subjects.
Collapse
Affiliation(s)
- Alessandra Schmitt Rieder
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Osmar Vieira Ramires Júnior
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Gustavo Ricardo Krupp Prauchner
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Neurometabolic Diseases, Department of Biochemistry, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
41
|
Barcena ML, Christiansen-Mensch C, Aslam M, Haritonow N, Ladilov Y, Regitz-Zagrosek V. Upregulation of Mitochondrial Sirt3 and Alleviation of the Inflammatory Phenotype in Macrophages by Estrogen. Cells 2024; 13:1420. [PMID: 39272992 PMCID: PMC11393879 DOI: 10.3390/cells13171420] [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: 06/27/2024] [Revised: 08/16/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Aging and comorbidities like type 2 diabetes and obesity contribute to the development of chronic systemic inflammation, which impacts the development of heart failure and vascular disease. Increasing evidence suggests a role of pro-inflammatory M1 macrophages in chronic inflammation. A shift of metabolism from mitochondrial oxidation to glycolysis is essential for the activation of the pro-inflammatory M1 phenotype. Thus, reprogramming the macrophage metabolism may alleviate the pro-inflammatory phenotype and protect against cardiovascular diseases. In the present study, we hypothesized that the activation of estrogen receptors leads to the elevation of the mitochondrial deacetylase Sirt3, which supports mitochondrial function and mitigates the pro-inflammatory phenotype in macrophages. MATERIALS AND METHODS Experiments were performed using the mouse macrophage cell line RAW264.7, as well as primary male or female murine bone marrow macrophages (BMMs). Macrophages were treated for 24 h with estradiol (E2) or vehicle (dextrin). The effect of E2 on Sirt3 expression was investigated in pro-inflammatory M1, anti-inflammatory/immunoregulatory M2, and naïve M0 macrophages. Mitochondrial respiration was measured by Seahorse assay, and protein expression and acetylation were determined by western blotting. RESULTS E2 treatment upregulated mitochondrial Sirt3, reduced mitochondrial protein acetylation, and increased basal mitochondrial respiration in naïve RAW264.7 macrophages. Similar effects on Sirt3 expression and mitochondrial protein acetylation were observed in primary female but not in male murine BMMs. Although E2 upregulated Sirt3 in naïve M0, pro-inflammatory M1, and anti-inflammatory/immunoregulatory M2 macrophages, it reduced superoxide dismutase 2 acetylation and suppressed mitochondrial reactive oxygen species formation only in pro-inflammatory M1 macrophages. E2 alleviated the pro-inflammatory phenotype in M1 RAW264.7 cells. CONCLUSIONS The study suggests that E2 treatment upregulates Sirt3 expression in macrophages. In primary BMMs, female-specific Sirt3 upregulation was observed. The Sirt3 upregulation was accompanied by mitochondrial protein deacetylation and the alleviation of the oxidative and pro-inflammatory phenotype in M1 macrophages. Thus, the E2-Sirt3 axis might be used in a therapeutic strategy to fight chronic systemic inflammation and prevent the development of inflammation-linked diseases.
Collapse
Affiliation(s)
- Maria Luisa Barcena
- Department of Urology, Eberhard Karl University of Tuebingen, 72076 Tuebingen, Germany
- German Center for Cardiovascular Research (DZHK), Berlin Partner Site, 10115 Berlin, Germany
| | | | - Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine I, Justus Liebig University, 35392 Giessen, Germany;
- German Center for Cardiovascular Research (DZHK), RheinMain Partner Site, 61231 Bad Nauheim, Germany
| | - Natalie Haritonow
- Department of Geriatrics and Medical Gerontology, Charité–Universitätsmedizin Berlin, 12203 Berlin, Germany;
| | - Yury Ladilov
- Department of Cardiovascular Surgery, Heart Center Brandenburg, Brandenburg Medical School, Bernau bei Berlin, 16321 Brandenburg, Germany;
| | - Vera Regitz-Zagrosek
- Institute for Gender in Medicine, Charité–Universitätsmedizin Berlin, 10115 Berlin, Germany; (C.C.-M.)
- Department of Cardiology, University Hospital Zürich, University of Zürich, 8091 Zürich, Switzerland
| |
Collapse
|
42
|
Wang J, Liang Y, Liang X, Peng H, Wang Y, Xu M, Liang X, Yao H, Liu X, Zeng L, Yao P, Xiang D. Evodiamine suppresses endometriosis development induced by early EBV exposure through inhibition of ERβ. Front Pharmacol 2024; 15:1426660. [PMID: 39148548 PMCID: PMC11324466 DOI: 10.3389/fphar.2024.1426660] [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: 05/01/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction: Endometriosis (EMS) is characterized as a prevalent gynecological inflammatory disorder marked by the existence of endometrial tissues situated beyond the uterus. This condition leads to persistent pelvic pain and may contribute to infertility. In this investigation, we explored the potential mechanism underlying the development of endometriosis (EMS) triggered by transient exposure to either latent membrane protein 1 (LMP1) or Epstein-Barr virus (EBV) in a mouse model. Additionally, we examined the potential inhibitory effect of evodiamine (EDM) on EMS. Methods: Immortalized human endometrial stromal cells (HESC) or epithelial cells (HEEC) were transiently exposed to either EBV or LMP1. The presence of evodiamine (EDM) was assessed for its impact on estrogen receptor β (ERβ) expression, as well as on cell metabolism parameters such as redox balance, mitochondrial function, inflammation, and proliferation. Additionally, a mixture of LMP1-treated HESC and HEEC was administered intraperitoneally to generate an EMS mouse model. Different dosages of EDM were employed for treatment to evaluate its potential suppressive effect on EMS development. Results: Transient exposure to either EBV or LMP1 triggers persistent ERβ expression through epigenetic modifications, subsequently modulating related cell metabolism for EMS development. Furthermore, 4.0 µM of EDM can efficiently reverse this effect in in vitro cell culture studies. Additionally, 20 mg/kg body weight of EDM treatment can partly suppress EMS development in the in vivo EMS mouse model. Conclusion: Transient EBV/LMP1 exposure triggers permanent ERβ expression, favoring later EMS development, EDM inhibits EMS development through ERβ suppression. This presents a novel mechanism for the development of endometriosis (EMS) in adulthood stemming from early Epstein-Barr virus (EBV) exposure during childhood. Moreover, evodiamine (EDM) stands out as a prospective candidate for treating EMS.
Collapse
Affiliation(s)
- Junling Wang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanqi Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoru Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huijuan Peng
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongxia Wang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Mingtao Xu
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuefang Liang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Helen Yao
- University of California at Riverside, Riverside, CA, United States
| | - Xiaohan Liu
- Department of Gynecology, Sun Yat-Sen University Affiliated No. 8 Hospital, Shenzhen, China
| | - Liqin Zeng
- Department of Gynecology, Sun Yat-Sen University Affiliated No. 8 Hospital, Shenzhen, China
| | - Paul Yao
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongfang Xiang
- Department of Gynecology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
43
|
Zachos KA, Gamboa JA, Dewji AS, Lee J, Brijbassi S, Andreazza AC. The interplay between mitochondria, the gut microbiome and metabolites and their therapeutic potential in primary mitochondrial disease. Front Pharmacol 2024; 15:1428242. [PMID: 39119601 PMCID: PMC11306032 DOI: 10.3389/fphar.2024.1428242] [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: 05/06/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
The various roles of the mitochondria and the microbiome in health and disease have been thoroughly investigated, though they are often examined independently and in the context of chronic disease. However, the mitochondria and microbiome are closely connected, namely, through their evolution, maternal inheritance patterns, overlapping role in many diseases and their importance in the maintenance of human health. The concept known as the "mitochondria-microbiome crosstalk" is the ongoing bidirectional crosstalk between these two entities and warrants further exploration and consideration, especially in the context of primary mitochondrial disease, where mitochondrial dysfunction can be detrimental for clinical manifestation of disease, and the role and composition of the microbiome is rarely investigated. A potential mechanism underlying this crosstalk is the role of metabolites from both the mitochondria and the microbiome. During digestion, gut microbes modulate compounds found in food, which can produce metabolites with various bioactive effects. Similarly, mitochondrial metabolites are produced from substrates that undergo biochemical processes during cellular respiration. This review aims to provide an overview of current literature examining the mitochondria-microbiome crosstalk, the role of commonly studied metabolites serve in signaling and mediating these biochemical pathways, and the impact diet has on both the mitochondria and the microbiome. As a final point, this review highlights the up-to-date implications of the mitochondria-microbiome crosstalk in mitochondrial disease and its potential as a therapeutic tool or target.
Collapse
Affiliation(s)
- Kassandra A. Zachos
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Mitochondrial Innovation Initiative, MITO2i, Toronto, ON, Canada
| | - Jann Aldrin Gamboa
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Aleena S. Dewji
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jocelyn Lee
- Mitochondrial Innovation Initiative, MITO2i, Toronto, ON, Canada
| | - Sonya Brijbassi
- Mitochondrial Innovation Initiative, MITO2i, Toronto, ON, Canada
| | - Ana C. Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Mitochondrial Innovation Initiative, MITO2i, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
Schneider Gasser EM, Schaer R, Mueller FS, Bernhardt AC, Lin HY, Arias-Reyes C, Weber-Stadlbauer U. Prenatal immune activation in mice induces long-term alterations in brain mitochondrial function. Transl Psychiatry 2024; 14:289. [PMID: 39009558 PMCID: PMC11251165 DOI: 10.1038/s41398-024-03010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Prenatal exposure to infections is a risk factor for neurodevelopmental disorders in offspring, and alterations in mitochondrial function are discussed as a potential underlying factor. Here, using a mouse model of viral-like maternal immune activation (MIA) based on poly(I:C) (POL) treatment at gestational day (GD) 12, we show that adult offspring exhibit behavioral deficits, such as reduced levels of social interaction. In addition, we found increased nicotinamidadenindinucleotid (NADH)- and succinate-linked mitochondrial respiration and maximal electron transfer capacity in the prefrontal cortex (PFC) and in the amygdala (AMY) of males and females. The increase in respiratory capacity resulted from an increase in mitochondrial mass in neurons (as measured by complex IV activity and transcript expression), presumably to compensate for a reduction in mitochondrion-specific respiration. Moreover, in the PFC of control (CON) male offspring a higher excess capacity compared to females was observed, which was significantly reduced in the POL-exposed male offspring, and, along with a higher leak respiration, resulted in a lower mitochondrial coupling efficiency. Transcript expression of the uncoupling proteins (UCP4 and UCP5) showed a reduction in the PFC of POL male mice, suggesting mitochondrial dysfunction. In addition, in the PFC of CON females, a higher expression of the antioxidant enzyme superoxide dismutase (SOD1) was observed, suggesting a higher antioxidant capacity as compared to males. Finally, transcripts analysis of genes involved in mitochondrial biogenesis and dynamics showed reduced expression of fission/fusion transcripts in PFC of POL offspring of both sexes. In conclusion, we show that MIA causes alterations in neuronal mitochondrial function and mass in the PFC and AMY of adult offspring with some effects differing between males and females.
Collapse
Affiliation(s)
- Edith M Schneider Gasser
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland.
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland.
- Department of Pediatrics, Faculty of Medicine, Université Laval, Québec, QC, Canada.
- Neuroscience Center Zurich, University of Zurich, and ETH, Zurich, 8057, Switzerland.
| | - Ron Schaer
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | - Flavia S Mueller
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | - Alexandra C Bernhardt
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | - Han-Yu Lin
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | | | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
- Neuroscience Center Zurich, University of Zurich, and ETH, Zurich, 8057, Switzerland
| |
Collapse
|
45
|
Liang C, Li X, Song G, Schmidt SF, Sun L, Chen J, Pan X, Zhao H, Yan Y. Adipose Kiss1 controls aerobic exercise-related adaptive responses in adipose tissue energy homeostasis. FASEB J 2024; 38:e23743. [PMID: 38877852 DOI: 10.1096/fj.202302598rr] [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: 12/15/2023] [Revised: 05/13/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024]
Abstract
Kisspeptin signaling regulates energy homeostasis. Adiposity is the principal source and receiver of peripheral Kisspeptin, and adipose Kiss1 metastasis suppressor (Kiss1) gene expression is stimulated by exercise. However, whether the adipose Kiss1 gene regulates energy homeostasis and plays a role in adaptive alterations during prolonged exercise remains unknown. Here, we investigated the role of Kiss1 role in mice and adipose tissues and the adaptive changes it induces after exercise, using adipose-specific Kiss1 knockout (Kiss1adipoq-/-) and adeno-associated virus-induced adipose tissue Kiss1-overexpressing (Kiss1adipoq over) mice. We found that adipose-derived kisspeptin signal regulates lipid and glucose homeostasis to maintain systemic energy homeostasis, but in a sex-dependent manner, with more pronounced metabolic changes in female mice. Kiss1 regulated adaptive alterations of genes and proteins in tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OxPhos) pathways in female gWAT following prolonged aerobic exercise. We could further show that adipose Kiss1 deficiency leads to reduced peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) protein content of soleus muscle and maximum oxygen uptake (VO2 max) of female mice after prolonged exercise. Therefore, adipose Kisspeptin may be a novel adipokine that increases organ sensitivity to glucose, lipids, and oxygen following exercise.
Collapse
Affiliation(s)
- Chunyu Liang
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
- Department of Biochemistry and Molecular Biology, Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark (SDU), Odense, Denmark
- School of Physical Education, Guangxi University (GXU), Nanning, China
| | - Xuehan Li
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Ge Song
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Søren Fisker Schmidt
- Department of Biochemistry and Molecular Biology, Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark (SDU), Odense, Denmark
| | - Lingyu Sun
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Jianhao Chen
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Xinliang Pan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Haotian Zhao
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| | - Yi Yan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing, China
- Laboratory of Sports Stress and Adaptation, General Administration of Sport of China, Beijing, China
| |
Collapse
|
46
|
Zhang G, Zeng C, Sun X, Zhang Q, Wang Y, Xia R, Mai Q, Xue G, Huang H, Wang F. Zearalenone modulates the function of goat endometrial cells via the mitochondrial quality control system. FASEB J 2024; 38:e23701. [PMID: 38941193 DOI: 10.1096/fj.202302198rr] [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: 10/26/2023] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/30/2024]
Abstract
Zearalenone (ZEN) is a mycotoxin known for its estrogen-like effects, which can disrupt the normal physiological function of endometrial cells and potentially lead to abortion in female animals. However, the precise mechanism by which ZEN regulates endometrial function remains unclear. In this study, we found that the binding receptor estrogen receptors for ZEN is extensively expressed across various segments of the uterus and within endometrial cells, and a certain concentration of ZEN treatment reduced the proliferation capacity of goat endometrial epithelial cells (EECs) and endometrial stromal cells (ESCs). Meanwhile, cell cycle analysis revealed that ZEN treatment leaded to cell cycle arrest in goat EECs and ESCs. To explore the underlying mechanism, we investigated the mitochondrial quality control systems and observed that ZEN triggered excessive mitochondrial fission and disturbed the balance of mitochondrial fusion-fission dynamics, impaired mitochondrial biogenesis, increased mitochondrial unfolded protein response and mitophagy in goat EECs and ESCs. Additionally, ZEN treatment reduced the activities of mitochondrial respiratory chain complexes, heightened the production of hydrogen peroxide and reactive oxygen species, and caused cellular oxidative stress and mitochondrial dysfunction. These results suggest that ZEN has adverse effects on goat endometrium cells by disrupting the mitochondrial quality control system and affecting cell cycle and proliferation. Understanding the underlying molecular pathways involved in ZEN-induced mitochondrial dysfunction and its consequences on cell function will provide critical insights into the reproductive toxicity of ZEN and contribute to safeguarding the health and wellbeing of animals and humans exposed to this mycotoxin.
Collapse
Affiliation(s)
- Guomin Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Cheng Zeng
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Xuan Sun
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Qi Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Yifei Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Rongxin Xia
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Qingyang Mai
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Gang Xue
- Yangtze River Delta White Goat Breeding Research Institute, Nantong, China
| | - Han Huang
- Mashan County Centre for Animal Disease Control and Prevention, Nanning, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
47
|
Ejikeme C, Safdar Z. Exploring the pathogenesis of pulmonary vascular disease. Front Med (Lausanne) 2024; 11:1402639. [PMID: 39050536 PMCID: PMC11267418 DOI: 10.3389/fmed.2024.1402639] [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: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a complex cardiopulmonary disorder impacting the lung vasculature, resulting in increased pulmonary vascular resistance that leads to right ventricular dysfunction. Pulmonary hypertension comprises of 5 groups (PH group 1 to 5) where group 1 pulmonary arterial hypertension (PAH), results from alterations that directly affect the pulmonary arteries. Although PAH has a complex pathophysiology that is not completely understood, it is known to be a multifactorial disease that results from a combination of genetic, epigenetic and environmental factors, leading to a varied range of symptoms in PAH patients. PAH does not have a cure, its incidence and prevalence continue to increase every year, resulting in higher morbidity and mortality rates. In this review, we discuss the different pathologic mechanisms with a focus on epigenetic modifications and their roles in the development and progression of PAH. These modifications include DNA methylation, histone modifications, and microRNA dysregulation. Understanding these epigenetic modifications will improve our understanding of PAH and unveil novel therapeutic targets, thus steering research toward innovative treatment strategies.
Collapse
Affiliation(s)
| | - Zeenat Safdar
- Department of Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
48
|
Szczepanska-Sadowska E, Czarzasta K, Bogacki-Rychlik W, Kowara M. The Interaction of Vasopressin with Hormones of the Hypothalamo-Pituitary-Adrenal Axis: The Significance for Therapeutic Strategies in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2024; 25:7394. [PMID: 39000501 PMCID: PMC11242374 DOI: 10.3390/ijms25137394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
A large body of evidence indicates that vasopressin (AVP) and steroid hormones are frequently secreted together and closely cooperate in the regulation of blood pressure, metabolism, water-electrolyte balance, and behavior, thereby securing survival and the comfort of life. Vasopressin cooperates with hormones of the hypothalamo-pituitary-adrenal axis (HPA) at several levels through regulation of the release of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and multiple steroid hormones, as well as through interactions with steroids in the target organs. These interactions are facilitated by positive and negative feedback between specific components of the HPA. Altogether, AVP and the HPA cooperate closely as a coordinated functional AVP-HPA system. It has been shown that cooperation between AVP and steroid hormones may be affected by cellular stress combined with hypoxia, and by metabolic, cardiovascular, and respiratory disorders; neurogenic stress; and inflammation. Growing evidence indicates that central and peripheral interactions between AVP and steroid hormones are reprogrammed in cardiovascular and metabolic diseases and that these rearrangements exert either beneficial or harmful effects. The present review highlights specific mechanisms of the interactions between AVP and steroids at cellular and systemic levels and analyses the consequences of the inappropriate cooperation of various components of the AVP-HPA system for the pathogenesis of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | | | | | | |
Collapse
|
49
|
Kim SO, Albrecht ED, Pepe GJ. Estrogen promotes fetal skeletal muscle mitochondrial distribution and ATP synthase activity important for insulin sensitivity in offspring. Endocrine 2024; 85:417-427. [PMID: 38478198 PMCID: PMC11246263 DOI: 10.1007/s12020-024-03764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/25/2024] [Indexed: 07/14/2024]
Abstract
PURPOSE We previously showed that offspring delivered to baboons in which levels of estradiol (E2) were suppressed during the second half of gestation exhibit insulin resistance. Mitochondria are essential for the production of ATP as the main source of energy for intracellular metabolic pathways, and skeletal muscle of type 2 diabetics exhibit mitochondrial abnormalities. Mitochondria express estrogen receptor β and E2 enhances mitochondrial function in adults. Therefore, the current study ascertained whether exposure of the fetus to E2 is essential for mitochondrial development. METHODS Levels of ATP synthase and citrate synthase and the morphology of mitochondria were determined in fetal skeletal muscle obtained near term from baboons untreated or treated daily with the aromatase inhibitor letrozole or letrozole plus E2. RESULTS Specific activity and amount of ATP synthase were 2-fold lower (P < 0.05) in mitochondria from skeletal muscle of E2 suppressed letrozole-treated fetuses and restored to normal by treatment with letrozole plus E2. Immunocytochemistry showed that in contrast to the punctate formation of mitochondria in myocytes of untreated and letrozole plus E2 treated animals, mitochondria appeared to be diffuse in myocytes of estrogen-suppressed fetuses. However, citrate synthase activity and levels of proteins that control mitochondrial fission/fusion were similar in estrogen replete and suppressed animals. CONCLUSION We suggest that estrogen is essential for fetal skeletal muscle mitochondrial development and thus glucose homeostasis in adulthood.
Collapse
Affiliation(s)
- Soon Ok Kim
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Eugene D Albrecht
- Departments of Obstetrics/Gynecology/Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
50
|
Kapic A, Zaman K, Nguyen V, Neagu GC, Sumien N, Prokai L, Prokai-Tatrai K. The Prodrug DHED Delivers 17β-Estradiol into the Retina for Protection of Retinal Ganglion Cells and Preservation of Visual Function in an Animal Model of Glaucoma. Cells 2024; 13:1126. [PMID: 38994978 PMCID: PMC11240555 DOI: 10.3390/cells13131126] [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: 06/04/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
We report a three-pronged phenotypic evaluation of the bioprecursor prodrug 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED) that selectively produces 17β-estradiol (E2) in the retina after topical administration and halts glaucomatous neurodegeneration in a male rat model of the disease. Ocular hypertension (OHT) was induced by hyperosmotic saline injection into an episcleral vein of the eye. Animals received daily DHED eye drops for 12 weeks. Deterioration of visual acuity and contrast sensitivity by OHT in these animals were markedly prevented by the DHED-derived E2 with concomitant preservation of retinal ganglion cells and their axons. In addition, we utilized targeted retina proteomics and a previously established panel of proteins as preclinical biomarkers in the context of OHT-induced neurodegeneration as a characteristic process of the disease. The prodrug treatment provided retina-targeted remediation against the glaucomatous dysregulations of these surrogate endpoints without increasing circulating E2 levels. Collectively, the demonstrated significant neuroprotective effect by the DHED-derived E2 in the selected animal model of glaucoma supports the translational potential of our presented ocular neuroprotective approach owing to its inherent therapeutic safety and efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Katalin Prokai-Tatrai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.K.); (K.Z.); (V.N.); (G.C.N.); (N.S.); (L.P.)
| |
Collapse
|