1
|
Maghfour J, Ozog DM, Mineroff J, Jagdeo J, Kohli I, Lim HW. Photobiomodulation CME part I: Overview and mechanism of action. J Am Acad Dermatol 2024; 91:793-802. [PMID: 38309304 DOI: 10.1016/j.jaad.2023.10.073] [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: 05/09/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 02/05/2024]
Abstract
Photobiomodulation (PBM), previously known as low-level laser light therapy, represents a noninvasive form of phototherapy that utilizes wavelengths in the red light (RL, 620-700 nm) portion of the visible light (VL, 400-700 nm) spectrum and the near-infrared (NIR, 700-1440 nm) spectrum. PBM is a promising and increasingly used therapy for the treatment of various dermatologic and nondermatologic conditions. Photons from RL and NIR are absorbed by endogenous photoreceptors including mitochondrial cytochrome C oxidase (COX). Activation of COX leads to the following changes: modulation of mitochondrial adenosine triphosphate (ATP), generation of reactive oxygen species (ROS), and alterations in intracellular calcium levels. The associated modulation of ATP, ROS and calcium levels promotes the activation of various signaling pathways (eg, insulin-like growth factors, phosphoinositide 3-kinase pathways), which contribute to downstream effects on cellular proliferation, migration, and differentiation. Effective PBM therapy is dependent on treatment parameters (eg, fluence, treatment duration and output power). PBM is generally well-tolerated and safe with erythema being the most common and self-limiting adverse cutaneous effect.
Collapse
Affiliation(s)
- Jalal Maghfour
- Department of Dermatology, Henry Ford Health, Detroit, Michigan
| | - David M Ozog
- Department of Dermatology, Henry Ford Health, Detroit, Michigan; The Henry W. Lim, MD, Division of Photobiology and Photomedicine, Department of Dermatology, Henry Ford Health, Detroit, Michigan; College of Human Medicine, Michigan State University, East Lansing, Michigan.
| | - Jessica Mineroff
- Department of Dermatology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Jared Jagdeo
- Department of Dermatology, State University of New York, Downstate Health Sciences University, Brooklyn, New York
| | - Indermeet Kohli
- The Henry W. Lim, MD, Division of Photobiology and Photomedicine, Department of Dermatology, Henry Ford Health, Detroit, Michigan; College of Human Medicine, Michigan State University, East Lansing, Michigan
| | - Henry W Lim
- Department of Dermatology, Henry Ford Health, Detroit, Michigan; The Henry W. Lim, MD, Division of Photobiology and Photomedicine, Department of Dermatology, Henry Ford Health, Detroit, Michigan; College of Human Medicine, Michigan State University, East Lansing, Michigan
| |
Collapse
|
2
|
Goyani S, Shukla S, Jadiya P, Tomar D. Calcium signaling in mitochondrial intermembrane space. Biochem Soc Trans 2024; 52:2215-2229. [PMID: 39392359 DOI: 10.1042/bst20240319] [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: 10/12/2024]
Abstract
The mitochondrial intermembrane space (IMS) is a highly protected compartment, second only to the matrix. It is a crucial bridge, coordinating mitochondrial activities with cellular processes such as metabolites, protein, lipid, and ion exchange. This regulation influences signaling pathways for metabolic activities and cellular homeostasis. The IMS harbors various proteins critical for initiating apoptotic cascades and regulating reactive oxygen species production by controlling the respiratory chain. Calcium (Ca2+), a key intracellular secondary messenger, enter the mitochondrial matrix via the IMS, regulating mitochondrial bioenergetics, ATP production, modulating cell death pathways. IMS acts as a regulatory site for Ca2+ entry due to the presence of different Ca2+ sensors such as MICUs, solute carriers (SLCs); ion exchangers (LETM1/SCaMCs); S100A1, mitochondrial glycerol-3-phosphate dehydrogenase, and EFHD1, each with unique Ca2+ binding motifs and spatial localizations. This review primarily emphasizes the role of these IMS-localized Ca2+ sensors concerning their spatial localization, mechanism, and molecular functions. Additionally, we discuss how these sensors contribute to the progression and pathogenesis of various human health conditions and diseases.
Collapse
Affiliation(s)
- Shanikumar Goyani
- Department of Internal Medicine, Section of Cardiovascular Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, U.S.A
| | - Shatakshi Shukla
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, U.S.A
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, U.S.A
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, U.S.A
| |
Collapse
|
3
|
Piamsiri C, Fefelova N, Pamarthi SH, Gwathmey JK, Chattipakorn SC, Chattipakorn N, Xie LH. Potential Roles of IP 3 Receptors and Calcium in Programmed Cell Death and Implications in Cardiovascular Diseases. Biomolecules 2024; 14:1334. [PMID: 39456267 PMCID: PMC11506173 DOI: 10.3390/biom14101334] [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: 09/23/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) play a crucial role in maintaining intracellular/cytosolic calcium ion (Ca2+i) homeostasis. The release of Ca2+ from IP3Rs serves as a second messenger and a modulatory factor influencing various intracellular and interorganelle communications during both physiological and pathological processes. Accumulating evidence from in vitro, in vivo, and clinical studies supports the notion that the overactivation of IP3Rs is linked to the pathogenesis of various cardiac conditions. The overactivation of IP3Rs results in the dysregulation of Ca2+ concentration ([Ca2+]) within cytosolic, mitochondrial, and nucleoplasmic cellular compartments. In cardiovascular pathologies, two isoforms of IP3Rs, i.e., IP3R1 and IP3R2, have been identified. Notably, IP3R1 plays a pivotal role in cardiac ischemia and diabetes-induced arrhythmias, while IP3R2 is implicated in sepsis-induced cardiomyopathy and cardiac hypertrophy. Furthermore, IP3Rs have been reported to be involved in various programmed cell death (PCD) pathways, such as apoptosis, pyroptosis, and ferroptosis underscoring their multifaceted roles in cardiac pathophysiology. Based on these findings, it is evident that exploring potential therapeutic avenues becomes crucial. Both genetic ablation and pharmacological intervention using IP3R antagonists have emerged as promising strategies against IP3R-related pathologies suggesting their potential therapeutic potency. This review summarizes the roles of IP3Rs in cardiac physiology and pathology and establishes a foundational understanding with a particular focus on their involvement in the various PCD pathways within the context of cardiovascular diseases.
Collapse
Affiliation(s)
- Chanon Piamsiri
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Sri Harika Pamarthi
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Judith K. Gwathmey
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers University-New Jersey Medical School, Newark, NJ 07103, USA; (C.P.); (N.F.)
| |
Collapse
|
4
|
Salis Torres A, Lee JE, Caporali A, Semple RK, Horrocks MH, MacRae VE. Mitochondrial Dysfunction as a Potential Mechanism Mediating Cardiac Comorbidities in Parkinson's Disease. Int J Mol Sci 2024; 25:10973. [PMID: 39456761 PMCID: PMC11507255 DOI: 10.3390/ijms252010973] [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: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Individuals diagnosed with Parkinson's disease (PD) often exhibit heightened susceptibility to cardiac dysfunction, reflecting a complex interaction between these conditions. The involvement of mitochondrial dysfunction in the development and progression of cardiac dysfunction and PD suggests a plausible commonality in some aspects of their molecular pathogenesis, potentially contributing to the prevalence of cardiac issues in PD. Mitochondria, crucial organelles responsible for energy production and cellular regulation, play important roles in tissues with high energetic demands, such as neurons and cardiac cells. Mitochondrial dysfunction can occur in different and non-mutually exclusive ways; however, some mechanisms include alterations in mitochondrial dynamics, compromised bioenergetics, biogenesis deficits, oxidative stress, impaired mitophagy, and disrupted calcium balance. It is plausible that these factors contribute to the increased prevalence of cardiac dysfunction in PD, suggesting mitochondrial health as a potential target for therapeutic intervention. This review provides an overview of the physiological mechanisms underlying mitochondrial quality control systems. It summarises the diverse roles of mitochondria in brain and heart function, highlighting shared pathways potentially exhibiting dysfunction and driving cardiac comorbidities in PD. By highlighting strategies to mitigate dysfunction associated with mitochondrial impairment in cardiac and neural tissues, our review aims to provide new perspectives on therapeutic approaches.
Collapse
Affiliation(s)
- Agustina Salis Torres
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
| | - Ji-Eun Lee
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Andrea Caporali
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Robert K. Semple
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Mathew H. Horrocks
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Vicky E. MacRae
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
| |
Collapse
|
5
|
Xie Y, Zhang W, Peng T, Wang X, Lian X, He J, Wang C, Xie N. TBC1D15-regulated mitochondria-lysosome membrane contact exerts neuroprotective effects by alleviating mitochondrial calcium overload in seizure. Sci Rep 2024; 14:23782. [PMID: 39390030 PMCID: PMC11467349 DOI: 10.1038/s41598-024-74388-3] [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: 03/15/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Mitochondrial calcium overload plays an important role in the neurological insults in seizure. The Rab7 GTPase-activating protein, Tre-2/Bub2/Cdc16 domain family member 15 (TBC1D15), is involved in the regulation of mitochondrial calcium dynamics by mediating mitochondria-lysosome membrane contact. However, whether TBC1D15-regulated mitochondria-lysosome membrane contact and mitochondrial calcium participate in neuronal injury in seizure is unclear. We aimed to investigate the effect of TBC1D15-regulated mitochondria-lysosome membrane contact on epileptiform discharge-induced neuronal damage and further explore the underlying mechanism. Lentiviral vectors (Lv) infection and stereotaxic adeno-associated virus (AAV) injection were used to regulate TBC1D15 expression before establishing in vitro epileptiform discharge and in vivo status epilepticus (SE) models. TBC1D15's effect on inter-organellar interactions, mitochondrial calcium levels and neuronal injury in seizure was evaluated. The results showed that abnormalities in mitochondria-lysosome membrane contact, mitochondrial calcium overload, mitochondrial dysfunction, increased levels of reactive oxygen species, and prominent neuronal damage were partly relieved by TBC1D15 overexpression, whereas TBC1D15 knockdown markedly deteriorated these phenomena. Further examination revealed that epileptiform discharge-induced mitochondrial calcium overload in primary hippocampal neurons was closely associated with abnormal mitochondria-lysosome membrane contact. This study highlights the crucial role played by TBC1D15-regulated mitochondria-lysosome membrane contact in epileptiform discharge-induced neuronal injury by alleviating mitochondrial calcium overload.
Collapse
Affiliation(s)
- Yinyin Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wanwan Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tingting Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyi Wang
- Institutes of Biological and Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaolei Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiao He
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Cui Wang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Nanchang Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
Alshehri RS, Abuzinadah AR, Alrawaili MS, Alotaibi MK, Alsufyani HA, Alshanketi RM, AlShareef AA. A Review of Biomarkers of Amyotrophic Lateral Sclerosis: A Pathophysiologic Approach. Int J Mol Sci 2024; 25:10900. [PMID: 39456682 PMCID: PMC11507293 DOI: 10.3390/ijms252010900] [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: 09/10/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive degeneration of upper and lower motor neurons. The heterogeneous nature of ALS at the clinical, genetic, and pathological levels makes it challenging to develop diagnostic and prognostic tools that fit all disease phenotypes. Limitations associated with the functional scales and the qualitative nature of mainstay electrophysiological testing prompt the investigation of more objective quantitative assessment. Biofluid biomarkers have the potential to fill that gap by providing evidence of a disease process potentially early in the disease, its progression, and its response to therapy. In contrast to other neurodegenerative diseases, no biomarker has yet been validated in clinical use for ALS. Several fluid biomarkers have been investigated in clinical studies in ALS. Biofluid biomarkers reflect the different pathophysiological processes, from protein aggregation to muscle denervation. This review takes a pathophysiologic approach to summarizing the findings of clinical studies utilizing quantitative biofluid biomarkers in ALS, discusses the utility and shortcomings of each biomarker, and highlights the superiority of neurofilaments as biomarkers of neurodegeneration over other candidate biomarkers.
Collapse
Affiliation(s)
- Rawiah S. Alshehri
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Ahmad R. Abuzinadah
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Moafaq S. Alrawaili
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Muteb K. Alotaibi
- Neurology Department, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia;
| | - Hadeel A. Alsufyani
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Rajaa M. Alshanketi
- Internal Medicine Department, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia;
| | - Aysha A. AlShareef
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| |
Collapse
|
7
|
Zhang L, Wu C, Liu T, Tian Y, Wang D, Wang B, Yin Y. Propofol Protects the Blood-Brain Barrier After Traumatic Brain Injury by Stabilizing the Extracellular Matrix via Prrx1: From Neuroglioma to Neurotrauma. Neurochem Res 2024; 49:2743-2762. [PMID: 38951281 DOI: 10.1007/s11064-024-04202-z] [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: 03/30/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
The purpose of this study is to explore the shared molecular pathogenesis of traumatic brain injury (TBI) and high-grade glioma and investigate the mechanism of propofol (PF) as a potential protective agent. By analyzing the Chinese glioma genome atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases, we compared the transcriptomic data of high-grade glioma and TBI patients to identify common pathological mechanisms. Through bioinformatics analysis, in vitro experiments and in vivo TBI model, we investigated the regulatory effect of PF on extracellular matrix (ECM)-related genes through Prrx1 under oxidative stress. The impact of PF on BBB integrity under oxidative stress was investigated using a dual-layer BBB model, and we explored the protective effect of PF on tight junction proteins and ECM-related genes in mice after TBI. The study found that high-grade glioma and TBI share ECM instability as an important molecular pathological mechanism. PF stabilizes the ECM and protects the BBB by directly binding to Prrx1 or indirectly regulating Prrx1 through miRNAs. In addition, PF reduces intracellular calcium ions and ROS levels under oxidative stress, thereby preserving BBB integrity. In a TBI mouse model, PF protected BBB integrity through up-regulated tight junction proteins and stabilized the expression of ECM-related genes. Our study reveals the shared molecular pathogenesis between TBI and glioblastoma and demonstrate the potential of PF as a protective agent of BBB. This provides new targets and approaches for the development of novel neurotrauma therapeutic drugs.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Chenrui Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Tian
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo Wang
- Department of Neurosurgery, Tianjin University Huanhu Hospital, Tianjin, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China.
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
8
|
Bhat AA, Moglad E, Afzal M, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Ali H, Pant K, Singh TG, Dureja H, Singh SK, Dua K, Gupta G, Subramaniyan V. Therapeutic approaches targeting aging and cellular senescence in Huntington's disease. CNS Neurosci Ther 2024; 30:e70053. [PMID: 39428700 PMCID: PMC11491556 DOI: 10.1111/cns.70053] [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: 05/29/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disease that is manifested by a gradual loss of physical, cognitive, and mental abilities. As the disease advances, age has a major impact on the pathogenic signature of mutant huntingtin (mHTT) protein aggregation. This review aims to explore the intricate relationship between aging, mHTT toxicity, and cellular senescence in HD. Scientific data on the interplay between aging, mHTT, and cellular senescence in HD were collected from several academic databases, including PubMed, Google Scholar, Google, and ScienceDirect. The search terms employed were "AGING," "HUNTINGTON'S DISEASE," "MUTANT HUNTINGTIN," and "CELLULAR SENESCENCE." Additionally, to gather information on the molecular mechanisms and potential therapeutic targets, the search was extended to include relevant terms such as "DNA DAMAGE," "OXIDATIVE STRESS," and "AUTOPHAGY." According to research, aging leads to worsening HD pathophysiology through some processes. As a result of the mHTT accumulation, cellular senescence is promoted, which causes DNA damage, oxidative stress, decreased autophagy, and increased inflammatory responses. Pro-inflammatory cytokines and other substances are released by senescent cells, which may worsen the neuronal damage and the course of the disease. It has been shown that treatments directed at these pathways reduce some of the HD symptoms and enhance longevity in experimental animals, pointing to a new possibility of treating the condition. Through their amplification of the harmful effects of mHTT, aging and cellular senescence play crucial roles in the development of HD. Comprehending these interplays creates novel opportunities for therapeutic measures targeted at alleviating cellular aging and enhancing HD patients' quality of life.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical SciencesUttaranchal UniversityDehradunIndia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of PharmacyPrince Sattam Bin Abdulaziz UniversityAl KharjSaudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy ProgramBatterjee Medical CollegeJeddahSaudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical SciencesUttaranchal UniversityDehradunIndia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of PharmacyUmm Al‐Qura UniversityMakkahSaudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of PharmacyJouf UniversitySakakaAl‐JoufSaudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Department of PharmacologyKyrgyz State Medical CollegeBishkekKyrgyzstan
| | - Kumud Pant
- Graphic Era (Deemed to be University), Dehradun, India
| | | | - Harish Dureja
- Department of Pharmaceutical SciencesMaharshi Dayanand UniversityRohtakIndia
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraPunjabIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of PharmacyChitkara UniversityRajpuraPunjabIndia
- Centre of Medical and Bio‐Allied Health Sciences ResearchAjman UniversityAjmanUnited Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health SciencesMonash UniversityBandar SunwaySelangor Darul EhsanMalaysia
- Department of Medical SciencesSchool of Medical and Life Sciences Sunway UniversityBandar SunwaySelangor Darul EhsanMalaysia
| |
Collapse
|
9
|
de la Harpe A, Beukes N, Frost C. Mitochondrial calcium overload contributes to cannabinoid-induced paraptosis in hormone-responsive breast cancer cells. Cell Prolif 2024; 57:e13650. [PMID: 38721827 PMCID: PMC11471428 DOI: 10.1111/cpr.13650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 10/15/2024] Open
Abstract
Studies have shown that natural products can induce paraptosis in tumour cell lines. Paraptosis is characterized by cytoplasmic vacuolation arising from the endoplasmic reticulum (ER) and mitochondria. The mechanism of paraptosis is unclear; however, dysregulation of Ca2+ homeostasis is believed to affect paraptosis induction. This study investigated the mechanism of cell death induced by a phytocannabinoid ratio in the MCF7 breast cancer cell line. The crystal violet assay was used to detect changes in viability and morphology changes were investigated using light and transmission electron microscopy. Various inhibitors, fluorescent staining with high-content screening, and Western blot analysis were used to investigate different cell death mechanisms. The phytocannabinoid ratio induced significant cell death and cytoplasmic vacuolation in MCF7 cells; however, no apoptosis, necrosis, autophagy, or ferroptosis was detected. Vacuolation induced by phytocannabinoid treatment was inhibited by cycloheximide, suggesting paraptosis induction. The mechanism of paraptosis induction was investigated, and it was found that treatment (1) induced ER dilation and mitochondrial swelling, (2) induced significant ER stress and mitochondrial Ca2+ overload and dysfunction, which appeared to be mediated by the voltage-dependent anion channel, and (3) significantly impaired all mitochondrial metabolic pathways. The data demonstrated that paraptosis induced by the cannabinoid ratio was mediated by Ca2+ flux from the ER to the mitochondria. These findings highlight a novel mechanism of cannabinoid-induced cell death and emphasize the anti-cancer potential of cannabinoid ratios, which exhibited enhanced effects compared to individual cannabinoids.
Collapse
Affiliation(s)
- A. de la Harpe
- Department of Biochemistry and MicrobiologyNelson Mandela UniversityPort ElizabethSouth Africa
| | - N. Beukes
- Department of Biochemistry and MicrobiologyNelson Mandela UniversityPort ElizabethSouth Africa
| | - C. Frost
- Department of Biochemistry and MicrobiologyNelson Mandela UniversityPort ElizabethSouth Africa
| |
Collapse
|
10
|
Xie W, Koppula S, Kale MB, Ali LS, Wankhede NL, Umare MD, Upaganlawar AB, Abdeen A, Ebrahim EE, El-Sherbiny M, Behl T, Shen B, Singla RK. Unraveling the nexus of age, epilepsy, and mitochondria: exploring the dynamics of cellular energy and excitability. Front Pharmacol 2024; 15:1469053. [PMID: 39309002 PMCID: PMC11413492 DOI: 10.3389/fphar.2024.1469053] [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: 07/23/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Epilepsy, a complex neurological condition marked by recurring seizures, is increasingly recognized for its intricate relationship with mitochondria, the cellular powerhouses responsible for energy production and calcium regulation. This review offers an in-depth examination of the interplay between epilepsy, mitochondrial function, and aging. Many factors might account for the correlation between epilepsy and aging. Mitochondria, integral to cellular energy dynamics and neuronal excitability, perform a critical role in the pathophysiology of epilepsy. The mechanisms linking epilepsy and mitochondria are multifaceted, involving mitochondrial dysfunction, reactive oxygen species (ROS), and mitochondrial dynamics. Mitochondrial dysfunction can trigger seizures by compromising ATP production, increasing glutamate release, and altering ion channel function. ROS, natural byproducts of mitochondrial respiration, contribute to oxidative stress and neuroinflammation, critical factors in epileptogenesis. Mitochondrial dynamics govern fusion and fission processes, influence seizure threshold and calcium buffering, and impact seizure propagation. Energy demands during seizures highlight the critical role of mitochondrial ATP generation in maintaining neuronal membrane potential. Mitochondrial calcium handling dynamically modulates neuronal excitability, affecting synaptic transmission and action potential generation. Dysregulated mitochondrial calcium handling is a hallmark of epilepsy, contributing to excitotoxicity. Epigenetic modifications in epilepsy influence mitochondrial function through histone modifications, DNA methylation, and non-coding RNA expression. Potential therapeutic avenues targeting mitochondria in epilepsy include mitochondria-targeted antioxidants, ketogenic diets, and metabolic therapies. The review concludes by outlining future directions in epilepsy research, emphasizing integrative approaches, advancements in mitochondrial research, and ethical considerations. Mitochondria emerge as central players in the complex narrative of epilepsy, offering profound insights and therapeutic potential for this challenging neurological disorder.
Collapse
Affiliation(s)
- Wen Xie
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Republic of Korea
| | - Mayur B. Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, India
| | - Lashin S. Ali
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
| | | | - Mohit D. Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, India
| | | | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Elturabi E. Ebrahim
- Medical-Surgical Nursing Department, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, India
| | - Bairong Shen
- Institutes for Systems Genetics, West China Tianfu Hospital, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K. Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| |
Collapse
|
11
|
Herath HMUL, Piao MJ, Kang KA, Fernando PDSM, Kang HK, Koh YS, Hyun JW. The inhibitory effect of chlorogenic acid on oxidative stress and apoptosis induced by PM 2.5 in HaCaT keratinocytes. J Biochem Mol Toxicol 2024; 38:e23806. [PMID: 39148258 DOI: 10.1002/jbt.23806] [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: 12/10/2023] [Revised: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 08/17/2024]
Abstract
Exposure to fine particulate matter with an aerodynamic diameter of less than 2.5 μm (PM2.5) can cause oxidative damage and apoptosis in the human skin. Chlorogenic acid (CGA) is a bioactive polyphenolic compound with antioxidant, antifungal, and antiviral properties. The objective of this study was to identify the ameliorating impact of CGA that might protect human HaCaT cells against PM2.5. CGA significantly scavenged the reactive oxygen species (ROS) generated by PM2.5, attenuated oxidative cellular/organelle damage, mitochondrial membrane depolarization, and suppressed cytochrome c release into the cytosol. The application of CGA led to a reduction in the expression levels of Bcl-2-associated X protein, caspase-9, and caspase-3, while simultaneously increasing the expression of B-cell lymphoma 2. In addition, CGA was able to reverse the decrease in cell viability caused by PM2.5 via the inhibition of extracellular signal-regulated kinase (ERK). This effect was further confirmed by the use of the mitogen-activated protein kinase kinase inhibitor, which acted upstream of ERK. In conclusion, CGA protected keratinocytes from mitochondrial damage and apoptosis via ameliorating PM2.5-induced oxidative stress and ERK activation.
Collapse
Affiliation(s)
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Pincha Devage Sameera Madushan Fernando
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Hee Kyoung Kang
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Young Sang Koh
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
12
|
Piao MJ, Kang KA, Fernando PDSM, Herath HMUL, Hyun JW. Silver nanoparticle-induced cell damage via impaired mtROS-JNK/MnSOD signaling pathway. Toxicol Mech Methods 2024; 34:803-812. [PMID: 38736318 DOI: 10.1080/15376516.2024.2350595] [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: 02/22/2024] [Accepted: 04/28/2024] [Indexed: 05/14/2024]
Abstract
This study investigated the mechanism of silver nanoparticle (AgNP) cytotoxicity from a mitochondrial perspective. The effect of AgNP on manganese superoxide dismutase (MnSOD), a mitochondrial antioxidant enzyme, against oxidative stress has not been studied in detail. We demonstrated that AgNP decreased MnSOD mRNA level, protein expression, and activity in human Chang liver cells in a time-dependent manner. AgNP induced the production of mitochondrial reactive oxygen species (mtROS), particularly superoxide anion. AgNP was found to increase mitochondrial calcium level and disrupt mitochondrial function, leading to reduced ATP level, succinate dehydrogenase activity, and mitochondrial permeability. AgNP induced cytochrome c release from the mitochondria into the cytoplasm, attenuated the expression of the anti-apoptotic proteins phospho Bcl-2 and Mcl-1, and induced the expression of the pro-apoptotic proteins Bim and Bax. In addition, c-Jun N-terminal kinase (JNK) phosphorylation was significantly increased by AgNP. Treatment with elamipretide (a mitochondria-targeted antioxidant) and SP600125 (a JNK inhibitor) showed the involvement of MnSOD and JNK in these processes. These results indicated that AgNP damaged human Chang liver cells by destroying mitochondrial function through the accumulation of mtROS.
Collapse
Affiliation(s)
- Mei Jing Piao
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | | | | | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
13
|
Danylovych HV, Danylovych YV, Pavliuk MR, Kosterin SO. Products of oxidative and non-oxidative metabolism of L-arginine as potential regulators of Ca 2+ transport in mitochondria of uterine smooth muscle. Biochim Biophys Acta Gen Subj 2024; 1868:130652. [PMID: 38857773 DOI: 10.1016/j.bbagen.2024.130652] [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: 03/14/2024] [Revised: 05/15/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Mitochondria play a crucial role in maintaining Ca2+ homeostasis in cells. Due to the critical regulatory role of the products of oxidative and non-oxidative metabolism of L-arginine, it is essential to clarify their effect on Ca2+ transport in smooth muscle mitochondria. Experiments were performed on the uterine myocytes of rats and isolated mitochondria. The possibility of NO synthesis by mitochondria was demonstrated by confocal microscopy and spectrofluorimetry methods using the NO-sensitive fluorescent probe DAF-FM and Mitotracker Orange CM-H2TMRos. It was shown that 50 μM L-arginine stimulates the energy-dependent accumulation of Ca2+ in mitochondria using the fluorescent probe Fluo-4 AM. A similar effect occurred when using nitric oxide donors 100 μM SNP, SNAP, and sodium nitrite (SN) directly. The stimulating effect was eliminated in the presence of the NO scavenger C-PTIO. Nitric oxide reduces the electrical potential in mitochondria without causing them to swell. The stimulatory effect of spermine on the accumulation of Ca2+ by mitochondria is attributed to the enhancement of NO synthesis, which was demonstrated with the use of C-PTIO, NO-synthase inhibitors (100 μM NA and L-NAME), as well as by direct monitoring of NO synthesis fluorescent probe DAF-FM. A conclusion was drawn about the potential regulatory effect of the product of the oxidative metabolism of L-arginine - NO on the transport of Ca2+ in the mitochondria of the myometrium, as well as the corresponding effect of the product of non-oxidative metabolism -spermine by increasing the synthesis of NO in these subcellular structures.
Collapse
Affiliation(s)
- Hanna V Danylovych
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine.
| | - Yuriy V Danylovych
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Maksym R Pavliuk
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Sergiy O Kosterin
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
14
|
Mohamed Yusoff AA, Mohd Khair SZN. Unraveling mitochondrial dysfunction: comprehensive perspectives on its impact on neurodegenerative diseases. Rev Neurosci 2024:revneuro-2024-0080. [PMID: 39174305 DOI: 10.1515/revneuro-2024-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024]
Abstract
Neurodegenerative diseases represent a significant challenge to modern medicine, with their complex etiology and progressive nature posing hurdles to effective treatment strategies. Among the various contributing factors, mitochondrial dysfunction has emerged as a pivotal player in the pathogenesis of several neurodegenerative disorders. This review paper provides a comprehensive overview of how mitochondrial impairment contributes to the development of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, driven by bioenergetic defects, biogenesis impairment, alterations in mitochondrial dynamics (such as fusion or fission), disruptions in calcium buffering, lipid metabolism dysregulation and mitophagy dysfunction. It also covers current therapeutic interventions targeting mitochondrial dysfunction in these diseases.
Collapse
Affiliation(s)
- Abdul Aziz Mohamed Yusoff
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Siti Zulaikha Nashwa Mohd Khair
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
15
|
Petrikonis K, Bernatoniene J, Kopustinskiene DM, Casale R, Davinelli S, Saso L. The Antinociceptive Role of Nrf2 in Neuropathic Pain: From Mechanisms to Clinical Perspectives. Pharmaceutics 2024; 16:1068. [PMID: 39204413 PMCID: PMC11358986 DOI: 10.3390/pharmaceutics16081068] [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: 06/29/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Neuropathic pain, a chronic condition resulting from nerve injury or dysfunction, presents significant therapeutic challenges and is closely associated with oxidative stress and inflammation, both of which can lead to mitochondrial dysfunction. The nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, a critical cellular defense mechanism against oxidative stress, has emerged as a promising target for neuropathic pain management. Nrf2 modulators enhance the expression of antioxidant and cytoprotective genes, thereby reducing oxidative damage, inflammation, and mitochondrial impairment. This review explores the antinociceptive effects of Nrf2, highlighting how pharmacological agents and natural compounds may be used as potential therapeutic strategies against neuropathic pain. Although preclinical studies demonstrate significant pain reduction and improved nerve function through Nrf2 activation, several clinical challenges need to be addressed. However, emerging clinical evidence suggests potential benefits of Nrf2 modulators in several conditions, such as diabetic neuropathy and multiple sclerosis. Future research should focus on further elucidating the molecular role of Nrf2 in neuropathic pain to optimize its modulation efficacy and maximize clinical utility.
Collapse
Affiliation(s)
- Kestutis Petrikonis
- Department of Neurology, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50009 Kaunas, Lithuania;
| | - Jurga Bernatoniene
- Department of Drug Technology and Social Pharmacy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania;
| | - Dalia M. Kopustinskiene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania;
| | - Roberto Casale
- Opusmedica Persons, Care & Research-NPO, 29121 Piacenza, Italy;
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
16
|
Khan S, Bano N, Ahamad S, John U, Dar NJ, Bhat SA. Excitotoxicity, Oxytosis/Ferroptosis, and Neurodegeneration: Emerging Insights into Mitochondrial Mechanisms. Aging Dis 2024:AD.2024.0125-1. [PMID: 39122453 DOI: 10.14336/ad.2024.0125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Mitochondrial dysfunction plays a pivotal role in the development of age-related diseases, particularly neurodegenerative disorders. The etiology of mitochondrial dysfunction involves a multitude of factors that remain elusive. This review centers on elucidating the role(s) of excitotoxicity, oxytosis/ferroptosis and neurodegeneration within the context of mitochondrial bioenergetics, biogenesis, mitophagy and oxidative stress and explores their intricate interplay in the pathogenesis of neurodegenerative diseases. The effective coordination of mitochondrial turnover processes, notably mitophagy and biogenesis, is assumed to be critically important for cellular resilience and longevity. However, the age-associated decrease in mitophagy impedes the elimination of dysfunctional mitochondria, consequently impairing mitochondrial biogenesis. This deleterious cascade results in the accumulation of damaged mitochondria and deterioration of cellular functions. Both excitotoxicity and oxytosis/ferroptosis have been demonstrated to contribute significantly to the pathophysiology of neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's Disease (HD), Amyotrophic Lateral Sclerosis (ALS) and Multiple Sclerosis (MS). Excitotoxicity, characterized by excessive glutamate signaling, initiates a cascade of events involving calcium dysregulation, energy depletion, and oxidative stress and is intricately linked to mitochondrial dysfunction. Furthermore, emerging concepts surrounding oxytosis/ferroptosis underscore the importance of iron-dependent lipid peroxidation and mitochondrial engagement in the pathogenesis of neurodegeneration. This review not only discusses the individual contributions of excitotoxicity and ferroptosis but also emphasizes their convergence with mitochondrial dysfunction, a key driver of neurodegenerative diseases. Understanding the intricate crosstalk between excitotoxicity, oxytosis/ferroptosis, and mitochondrial dysfunction holds potential to pave the way for mitochondrion-targeted therapeutic strategies. Such strategies, with a focus on bioenergetics, biogenesis, mitophagy, and oxidative stress, emerge as promising avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh-202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh-202002, India
| | - Urmilla John
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India; School of Studies in Zoology, Jiwaji University, Gwalior, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | | |
Collapse
|
17
|
Pradeepkiran JA, Baig J, Seman A, Reddy PH. Mitochondria in Aging and Alzheimer's Disease: Focus on Mitophagy. Neuroscientist 2024; 30:440-457. [PMID: 36597577 DOI: 10.1177/10738584221139761] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid β and phosphorylated τ protein aggregates in the brain, which leads to the loss of neurons. Under the microscope, the function of mitochondria is uniquely primed to play a pivotal role in neuronal cell survival, energy metabolism, and cell death. Research studies indicate that mitochondrial dysfunction, excessive oxidative damage, and defective mitophagy in neurons are early indicators of AD. This review article summarizes the latest development of mitochondria in AD: 1) disease mechanism pathways, 2) the importance of mitochondria in neuronal functions, 3) metabolic pathways and functions, 4) the link between mitochondrial dysfunction and mitophagy mechanisms in AD, and 5) the development of potential mitochondrial-targeted therapeutics and interventions to treat patients with AD.
Collapse
Affiliation(s)
| | - Javaria Baig
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ashley Seman
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
18
|
Yoo H, Park H. Altered mitochondrial Ca 2+ uptake in presynaptic terminals of cultured striatal and cortical neurons from the zQ175 knock-in mouse model of Huntington's disease. Biochem Biophys Res Commun 2024; 716:150010. [PMID: 38704892 DOI: 10.1016/j.bbrc.2024.150010] [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: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
Calcium (Ca2+) in mitochondria plays crucial roles in neurons including modulating metabolic processes. Moreover, excessive Ca2+ in mitochondria can lead to cell death. Thus, altered mitochondrial Ca2+ regulation has been implicated in several neurodegenerative diseases including Huntington's disease (HD). HD is a progressive hereditary neurodegenerative disorder that results from abnormally expanded cytosine-adenine-guanine trinucleotide repeats in the huntingtin gene. One neuropathological hallmark of HD is neuronal loss in the striatum and cortex. However, mechanisms underlying selective loss of striatal and cortical neurons in HD remain elusive. Here, we measured the basal Ca2+ levels and Ca2+ uptake in single presynaptic mitochondria during 100 external electrical stimuli using highly sensitive mitochondria-targeted Ca2+ indicators in cultured cortical and striatal neurons of a knock-in mouse model of HD (zQ175 mice). We observed elevated presynaptic mitochondrial Ca2+ uptake during 100 electrical stimuli in HD cortical neurons compared with wild-type (WT) cortical neurons. We also found the highly elevated presynaptic mitochondrial basal Ca2+ level and Ca2+ uptake during 100 stimuli in HD striatal neurons. The elevated presynaptic mitochondrial basal Ca2+ level in HD striatal neurons and Ca2+ uptake during stimulation in HD striatal and cortical neurons can disrupt neurotransmission and induce mitochondrial Ca2+ overload, eventually leading to neuronal death in the striatum and cortex of HD.
Collapse
Affiliation(s)
- Hanna Yoo
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Hyokeun Park
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
| |
Collapse
|
19
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
20
|
Baron KR, Oviedo S, Krasny S, Zaman M, Aldakhlallah R, Mathur P, Pfeffer G, Bollong MJ, Shutt T, Grotjahn DA, Wiseman RL. Pharmacologic Activation of Integrated Stress Response Kinases Inhibits Pathologic Mitochondrial Fragmentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598126. [PMID: 38915623 PMCID: PMC11195119 DOI: 10.1101/2024.06.10.598126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Excessive mitochondrial fragmentation is associated with the pathologic mitochondrial dysfunction implicated in the pathogenesis of etiologically-diverse diseases, including many neurodegenerative disorders. The integrated stress response (ISR) - comprising the four eIF2α kinases PERK, GCN2, PKR, and HRI - is a prominent stress-responsive signaling pathway that regulates mitochondrial morphology and function in response to diverse types of pathologic insult. This suggests that pharmacologic, stress-independent activation of the ISR represents a potential strategy to mitigate pathologic mitochondrial fragmentation associated with human disease. Here, we show that pharmacologic, stress-independent activation of the ISR kinases HRI or GCN2 promotes adaptive mitochondrial elongation and prevents mitochondrial fragmentation induced by the calcium ionophore ionomycin. Further, we show that stress-independent activation of these ISR kinases reduces mitochondrial fragmentation and restores basal mitochondrial morphology in patient fibroblasts expressing the pathogenic D414V variant of the pro-fusion mitochondrial GTPase MFN2 associated with neurological dysfunctions including ataxia, optic atrophy, and sensorineural hearing loss. These results identify pharmacologic, stress-independent activation of ISR kinases as a potential strategy to prevent pathologic mitochondrial fragmentation induced by disease-relevant chemical and genetic insults, further motivating the pursuit of highly selective ISR kinase-activating compounds as a therapeutic strategy to mitigate mitochondrial dysfunction implicated in diverse human diseases.
Collapse
Affiliation(s)
- Kelsey R. Baron
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
- These authors contributed equally
| | - Samantha Oviedo
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
- Department of Integrative Structural and Computation Biology, The Scripps Research Institute, La Jolla, CA 92037
- These authors contributed equally
| | - Sophia Krasny
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Mashiat Zaman
- Department of Biochemistry and Molecular Biology, Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rama Aldakhlallah
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Prakhyat Mathur
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary; Alberta Child Health Research Institute, Department of Medical Genetics, Cumming School of Medicine, University of Calgary
| | - Michael J. Bollong
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Timothy Shutt
- Departments of Medical Genetics and Biochemistry & Molecular Biology, Cumming School of Medicine, Hotchkiss Brain Institute, Snyder Institute for Chronic Diseases, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Danielle A. Grotjahn
- Department of Integrative Structural and Computation Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - R. Luke Wiseman
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
21
|
Chung E, Wen X, Jia X, Ciallella HL, Aleksunes LM, Zhu H. Hybrid non-animal modeling: A mechanistic approach to predict chemical hepatotoxicity. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134297. [PMID: 38677119 PMCID: PMC11519847 DOI: 10.1016/j.jhazmat.2024.134297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
Developing mechanistic non-animal testing methods based on the adverse outcome pathway (AOP) framework must incorporate molecular and cellular key events associated with target toxicity. Using data from an in vitro assay and chemical structures, we aimed to create a hybrid model to predict hepatotoxicants. We first curated a reference dataset of 869 compounds for hepatotoxicity modeling. Then, we profiled them against PubChem for existing in vitro toxicity data. Of the 2560 resulting assays, we selected the mitochondrial membrane potential (MMP) assay, a high-throughput screening (HTS) tool that can test chemical disruptors for mitochondrial function. Machine learning was applied to develop quantitative structure-activity relationship (QSAR) models with 2536 compounds tested in the MMP assay for screening new compounds. The MMP assay results, including QSAR model outputs, yielded hepatotoxicity predictions for reference set compounds with a Correct Classification Ratio (CCR) of 0.59. The predictivity improved by including 37 structural alerts (CCR = 0.8). We validated our model by testing 37 reference set compounds in human HepG2 hepatoma cells, and reliably predicting them for hepatotoxicity (CCR = 0.79). This study introduces a novel AOP modeling strategy that combines public HTS data, computational modeling, and experimental testing to predict chemical hepatotoxicity.
Collapse
Affiliation(s)
- Elena Chung
- Department of Chemistry and Biochemistry, Rowan University, NJ, USA; Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA, USA
| | - Xia Wen
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Xuelian Jia
- Department of Chemistry and Biochemistry, Rowan University, NJ, USA; Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA, USA
| | - Heather L Ciallella
- Department of Toxicology, Cuyahoga County Medical Examiner's Office, Cleveland, OH, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Hao Zhu
- Department of Chemistry and Biochemistry, Rowan University, NJ, USA; Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
22
|
Kim YW, Yang SG, Seo BB, Koo DB, Park HJ. Deoxynivalenol leads to endoplasmic reticulum stress-mediated apoptosis via the IRE1/JNK/CHOP pathways in porcine embryos. Food Chem Toxicol 2024; 188:114633. [PMID: 38608924 DOI: 10.1016/j.fct.2024.114633] [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: 02/19/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024]
Abstract
The cytotoxic mycotoxin deoxynivalenol (DON) reportedly has adverse effects on oocyte maturation and embryonic development in pigs. Recently, the interplay between cell apoptosis and endoplasmic reticulum (ER) stress has garnered increasing attention in embryogenesis. However, the involvement of the inositol-requiring enzyme 1 (IRE1)/c-jun N-terminal kinase (JNK)/C/EBP homologous protein (CHOP) pathways of unfolded protein response (UPR) signaling in DON-induced apoptosis in porcine embryos remains unknown. In this study, we revealed that exposure to DON (0.25 μM) substantially decreased cell viability until the blastocyst stage in porcine embryos, concomitant with initiation of cell apoptosis through the IRE1/JNK/CHOP pathways in response to ER stress. Quantitative PCR confirmed that UPR signaling-related transcription factors were upregulated in DON-treated porcine blastocysts. Western blot analysis showed that IRE1/JNK/CHOP signaling was activated in DON-exposed porcine embryos, indicating that ER stress-associated apoptosis was instigated. The ER stress inhibitor tauroursodeoxycholic acid protected against DON-induced ER stress in porcine embryos, indicating that the toxic effects of DON on early developmental competence of porcine embryos can be prevented. In conclusion, DON exposure impairs the developmental ability of porcine embryos by inducing ER stress-mediated apoptosis via IRE1/JNK/CHOP signaling.
Collapse
Affiliation(s)
- Ye-Won Kim
- Department of Biotechnology, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, Gyeongbuk, 38453, Republic of Korea; DU Center for Infertility, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Seul-Gi Yang
- DU Center for Infertility, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, Gyeongbuk, 38453, Republic of Korea; Department of Companion Animal Industry, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, 38453, Republic of Korea
| | - Byoung-Boo Seo
- Department of Companion Animal Industry, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, 38453, Republic of Korea
| | - Deog-Bon Koo
- Department of Biotechnology, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, Gyeongbuk, 38453, Republic of Korea; DU Center for Infertility, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, Gyeongbuk, 38453, Republic of Korea; Department of Companion Animal Industry, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, 38453, Republic of Korea.
| | - Hyo-Jin Park
- Department of Biotechnology, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, Gyeongbuk, 38453, Republic of Korea; DU Center for Infertility, Daegu University, 201 Daegudae-ro, Jillyang, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
23
|
Ezhilarasan D. Deciphering the molecular pathways of saroglitazar: A dual PPAR α/γ agonist for managing metabolic NAFLD. Metabolism 2024; 155:155912. [PMID: 38609038 DOI: 10.1016/j.metabol.2024.155912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Saroglitazar (SARO), a dual peroxisome proliferator activated receptor (PPAR)-α/γ agonist, has been used to treat metabolic diseases such as insulin resistance and diabetic dyslipidemia in patients with non-alcoholic fatty liver disease (NAFLD). SARO, administered at a dose of 4 mg/day, has been consistently studied in clinical trials with different time points ranging from 4 to 24 weeks with NAFLD patients. Due to its PPAR-γ agonistic action, SARO prevents adipose tissue-mediated fatty acid delivery to the liver by increasing insulin sensitivity and regulating adiponectin and leptin levels in adipose tissue. In hepatocytes, SARO induces fatty acid β-oxidation in mitochondria and transcriptionally activates lipid metabolizing genes in peroxisomes. SARO inhibits insulin resistance, thereby preventing the activation of sterol regulatory element-binding proteins -1c and carbohydrate response element binding protein in hepatocytes through its PPAR-α agonistic action. SARO treatment reduces lipotoxicity-mediated oxidative stress by activating the nuclear factor erythroid 2-related factor 2 and transcriptionally expressing the antioxidants from the antioxidant response element in the nucleus through its PPAR-γ agonistic action. SARO provides a PPAR-α/γ-mediated anti-inflammatory effect by preventing the phosphorylation of mitogen-activated protein kinases (JNK and ERK) and nuclear factor kappa B in hepatocytes. Additionally, SARO interferes with transforming growth factor-β/Smad downstream signaling, thereby reducing liver fibrosis progression through its PPAR-α/γ agonistic actions. Thus, SARO improves insulin resistance and dyslipidemia in NAFLD, reduces lipid accumulation in the liver, and thereby prevents mitochondrial toxicity, oxidative stress, inflammation, and fibrosis progression. This review summarizes the possible molecular mechanism of SARO in the NAFLD.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Hepatology and Molecular Medicine Lab, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600 077, India.
| |
Collapse
|
24
|
Yadav A, Dabur R. Skeletal muscle atrophy after sciatic nerve damage: Mechanistic insights. Eur J Pharmacol 2024; 970:176506. [PMID: 38492879 DOI: 10.1016/j.ejphar.2024.176506] [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: 01/08/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
Sciatic nerve injury leads to molecular events that cause muscular dysfunction advancement in atrophic conditions. Nerve damage renders muscles permanently relaxed which elevates intracellular resting Ca2+ levels. Increased Ca2+ levels are associated with several cellular signaling pathways including AMPK, cGMP, PLC-β, CERB, and calcineurin. Also, multiple enzymes involved in the tricarboxylic acid cycle and oxidative phosphorylation are activated by Ca2+ influx into mitochondria during muscle contraction, to meet increased ATP demand. Nerve damage induces mitophagy and skeletal muscle atrophy through increased sensitivity to Ca2+-induced opening of the permeability transition pore (PTP) in mitochondria attributed to Ca2+, ROS, and AMPK overload in muscle. Activated AMPK interacts negatively with Akt/mTOR is a highly prevalent and well-described central pathway for anabolic processes. Over the decade several reports indicate abnormal behavior of signaling machinery involved in denervation-induced muscle loss but end up with some controversial outcomes. Therefore, understanding how the synthesis and inhibitory stimuli interact with cellular signaling to control muscle mass and morphology may lead to new pharmacological insights toward understanding the underlying mechanism of muscle loss after sciatic nerve damage. Hence, the present review summarizes the existing literature on denervation-induced muscle atrophy to evaluate the regulation and expression of differential regulators during sciatic damage.
Collapse
Affiliation(s)
- Aarti Yadav
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Rajesh Dabur
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| |
Collapse
|
25
|
Kong P, Ahmad RE, Zulkifli A, Krishnan S, Nam HY, Kamarul T. The role of autophagy in mitigating osteoarthritis progression via regulation of chondrocyte apoptosis: A review. Joint Bone Spine 2024; 91:105642. [PMID: 37739213 DOI: 10.1016/j.jbspin.2023.105642] [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: 03/28/2023] [Revised: 07/22/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease with an immense socioeconomic burden; however, no treatment has achieved complete success in effectively halting or reversing cartilage degradation, which is the central pathophysiological feature of OA. Chondrocytes loss or dysfunction is a significant contributing factor to the progressive cartilage deterioration as these sole resident cells have a crucial role to produce extracellular matrix proteins, thus maintaining cartilage structure and homeostasis. It has been previously suggested that death of chondrocytes occurring through apoptosis substantially contributes to cartilage degeneration. Although the occurrence of apoptosis in osteoarthritic cartilage and its correlation with cartilage degradation is evident, the causes of chondrocyte apoptosis leading to matrix loss are still not well-understood. Autophagy, an intracellular degradative mechanism that eliminates dysfunctional cytoplasmic components to aid cell survival in unfavourable conditions, is a potential therapeutic target to inhibit chondrocyte apoptosis and reduce OA severity. Despite accumulating evidence indicating significant cytoprotective effects of autophagy against chondrocyte apoptosis, the mechanistic link between autophagy and apoptosis in chondrocytes remains to be further explored. In this review, we summarize the relevant mechanistic events that perpetuate chondrocyte apoptosis and highlight the prominent role of autophagy in modulating these events to mitigate OA progression.
Collapse
Affiliation(s)
- Peggy Kong
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Raja Elina Ahmad
- Department of Physiology, Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia.
| | - Amirah Zulkifli
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Shaliny Krishnan
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Hui Yin Nam
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia; Nanotechnology and Catalysis Research Centre (NANOCAT), Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- Department of Orthopaedic Surgery, Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning (NOCERAL), Faculty of Medicine, Universiti Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia; Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas Pulau Pinang, Malaysia
| |
Collapse
|
26
|
Pal I, Bhattacharyya A, V-Ghaffari B, Williams ED, Xiao M, Rutherford MA, Rubio ME. Female GluA3-KO mice show early onset hearing loss and afferent swellings in ambient sound levels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.21.581467. [PMID: 38659964 PMCID: PMC11042237 DOI: 10.1101/2024.02.21.581467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
AMPA-type glutamate receptors (AMPAR) mediate excitatory cochlear transmission. However, the unique roles of AMPAR subunits are unresolved. Lack of subunit GluA3 (Gria3KO) in male mice reduced cochlear output by 8-weeks of age. Since Gria3 is X-linked and considering sex differences in hearing vulnerability, we hypothesized accelerated presbycusis in Gria3KO females. Here, auditory brainstem responses (ABR) were similar in 3-week-old female Gria3WT and Gria3KO mice. However, when raised in ambient sound, ABR thresholds were elevated and wave-1 amplitudes were diminished at 5-weeks and older in Gria3KO. In contrast, these metrics were similar between genotypes when raised in quiet. Paired synapses were similar in number, but lone ribbons and ribbonless synapses were increased in female Gria3KO mice in ambient sound compared to Gria3WT or to either genotype raised in quiet. Synaptic GluA4:GluA2 ratios increased relative to Gria3WT, particularly in ambient sound, suggesting an activity-dependent increase in calcium-permeable AMPARs in Gria3KO. Swollen afferent terminals were observed by 5-weeks only in Gria3KO females reared in ambient sound. We propose that lack of GluA3 induces sex-dependent vulnerability to AMPAR-mediated excitotoxicity.
Collapse
Affiliation(s)
- Indra Pal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Atri Bhattacharyya
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - Babak V-Ghaffari
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - Essence D. Williams
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Maolei Xiao
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - Mark A. Rutherford
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO 63110
| | - María Eulalia Rubio
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
27
|
Mareedu S, Fefelova N, Galindo CL, Prakash G, Mukai R, Sadoshima J, Xie LH, Babu GJ. Improved mitochondrial function in the hearts of sarcolipin-deficient dystrophin and utrophin double-knockout mice. JCI Insight 2024; 9:e170185. [PMID: 38564291 PMCID: PMC11141945 DOI: 10.1172/jci.insight.170185] [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: 03/08/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease associated with cardiomyopathy. DMD cardiomyopathy is characterized by abnormal intracellular Ca2+ homeostasis and mitochondrial dysfunction. We used dystrophin and utrophin double-knockout (mdx:utrn-/-) mice in a sarcolipin (SLN) heterozygous-knockout (sln+/-) background to examine the effect of SLN reduction on mitochondrial function in the dystrophic myocardium. Germline reduction of SLN expression in mdx:utrn-/- mice improved cardiac sarco/endoplasmic reticulum (SR) Ca2+ cycling, reduced cardiac fibrosis, and improved cardiac function. At the cellular level, reducing SLN expression prevented mitochondrial Ca2+ overload, reduced mitochondrial membrane potential loss, and improved mitochondrial function. Transmission electron microscopy of myocardial tissues and proteomic analysis of mitochondria-associated membranes showed that reducing SLN expression improved mitochondrial structure and SR-mitochondria interactions in dystrophic cardiomyocytes. These findings indicate that SLN upregulation plays a substantial role in the pathogenesis of cardiomyopathy and that reducing SLN expression has clinical implications in the treatment of DMD cardiomyopathy.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Calcium/metabolism
- Cardiomyopathies/metabolism
- Cardiomyopathies/genetics
- Cardiomyopathies/pathology
- Disease Models, Animal
- Dystrophin/genetics
- Dystrophin/metabolism
- Mice, Inbred mdx
- Mice, Knockout
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/ultrastructure
- Mitochondria, Heart/genetics
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Proteolipids/metabolism
- Proteolipids/genetics
- Utrophin/genetics
- Utrophin/metabolism
Collapse
Affiliation(s)
- Satvik Mareedu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Cristi L. Galindo
- Vascular Medicine Institute and Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Goutham Prakash
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Gopal J. Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
28
|
Abstract
Eryptosis is a regulated cell death (RCD) of mature erythrocytes initially described as a counterpart of apoptosis for enucleated cells. However, over the recent years, a growing number of studies have emphasized certain differences between both cell death modalities. In this review paper, we underline the hallmarks of eryptosis and apoptosis and highlight resemblances and dissimilarities between both RCDs. We summarize and critically discuss differences in the impact of caspase-3, Ca2+ signaling, ROS signaling pathways, opposing roles of casein kinase 1α, protein kinase C, Janus kinase 3, cyclin-dependent kinase 4, and AMP-activated protein kinase to highlight a certain degree of divergence between apoptosis and eryptosis. This review emphasizes the crucial importance of further studies that focus on deepening our knowledge of cell death machinery and identifying novel differences between cell death of nucleated and enucleated cells. This might provide evidence that erythrocytes can be defined as viable entities capable of programmed cell destruction. Additionally, the revealed cell type-specific patterns in cell death can facilitate the development of cell death-modulating therapeutic agents.
Collapse
Affiliation(s)
- Anton Tkachenko
- 1st Faculty of Medicine, BIOCEV, Charles University, Průmyslová 595, 25250, Vestec, Czech Republic.
| |
Collapse
|
29
|
Danylovych H, Danylovych Y, Chunikhin A, Cherenok S, Kalchenko V, Kosterin S. Use of thiacalix[4]arene C-1193 for a directed influence on the functional activity of mitochondria and simulation of this process using a Petri nets. BIOTECHNOLOGIA 2024; 105:69-81. [PMID: 38633893 PMCID: PMC11020151 DOI: 10.5114/bta.2024.135643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 04/19/2024] Open
Abstract
In molecular biological studies, considerable attention is paid to macrocyclic nanoscale compounds known as calix[4]arenes. An imperative concern in biochemical membranology and molecular biotechnology is the exploration of effectors capable of modifying the intensity of redox reactions within the inner mitochondrial membrane and influencing the activity of its Ca2+ transport systems. The simulation model development is relevant to formalize and generalize the experimental data and assess the conformity of experimental results with theoretical predictions. Experiments were carried out on a suspension of isolated rat myometrial mitochondria. The synthesized thiacalix[4]arene C-1193, containing four sulfur atoms, was employed. Demonstrations of time-dependent and concentration-dependent (0.01-10 μM) inhibition of Ca2+ accumulation and reactive oxygen species (ROS) formation by mitochondria in the presence of C-1193 were observed. While C-1193 inhibited the oxidation of NADH and FADH2, it did not induce mitochondrial swelling. The thiacalix[4]arene also inhibited the synthesis of nitric oxide, with a Ki of 5.5 ± 1.7 nM, positioning it as a high-affinity blocker of endogenous NO generation in mitochondria. These results are the basis for the possible application of the synthesized thiacalix[4]arene as a tool in researching biochemical processes in mitochondria. A simulation model employing functional hybrid Petri nets was developed, reproducing the functional activity of mitochondria, including simultaneous NADH oxidation, ROS formation, NO synthesis, and Ca2+ accumulation. The derived equations formalize and describe the time dependencies of the listed processes in the medium under the influence of thiacalix[4]arene C-1193.
Collapse
Affiliation(s)
- Hanna Danylovych
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Yurii Danylovych
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Alexander Chunikhin
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Sergiy Cherenok
- Institute of Organic Chemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Vitaly Kalchenko
- Institute of Organic Chemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Sergiy Kosterin
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| |
Collapse
|
30
|
Hartsoe P, Holguin F, Chu HW. Mitochondrial Dysfunction and Metabolic Reprogramming in Obesity and Asthma. Int J Mol Sci 2024; 25:2944. [PMID: 38474191 PMCID: PMC10931700 DOI: 10.3390/ijms25052944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Mitochondrial dysfunction and metabolic reprogramming have been extensively studied in many disorders ranging from cardiovascular to neurodegenerative disease. Obesity has previously been associated with mitochondrial fragmentation, dysregulated glycolysis, and oxidative phosphorylation, as well as increased reactive oxygen species production. Current treatments focus on reducing cellular stress to restore homeostasis through the use of antioxidants or alterations of mitochondrial dynamics. This review focuses on the role of mitochondrial dysfunction in obesity particularly for those suffering from asthma and examines mitochondrial transfer from mesenchymal stem cells to restore function as a potential therapy. Mitochondrial targeted therapy to restore healthy metabolism may provide a unique approach to alleviate dysregulation in individuals with this unique endotype.
Collapse
Affiliation(s)
- Paige Hartsoe
- Department of Medicine, National Jewish Health, Denver, CO 80222, USA
| | - Fernando Holguin
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO 80222, USA
| |
Collapse
|
31
|
Mohanad M, Mohamed SK, Aboulhoda BE, Ahmed MAE. Neuroprotective effects of vitamin D in an Alzheimer's disease rat model: Improvement of mitochondrial dysfunction via calcium/calmodulin-dependent protein kinase kinase 2 activation of Sirtuin1 phosphorylation. Biofactors 2024; 50:371-391. [PMID: 37801071 DOI: 10.1002/biof.2013] [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: 05/12/2023] [Accepted: 09/17/2023] [Indexed: 10/07/2023]
Abstract
Mitochondrial dysfunction is an early event in Alzheimer's disease (AD) pathogenesis. To assess the impact of vitamin D3 (Vit.D) on neurogenesis, we investigated its role in mitigating cognitive impairment and mitochondrial dysfunction through calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2)-mediated phosphorylation of Sirtuin1 (SIRT1) in an aluminum-chloride-D-galactose (AlCl3-D-gal)-induced AD rat model. Rats were distributed into four groups: control, AlCl3 + D-gal (10 + 60 mg/kg, ip), Vit.D (500 IU/kg, po), and AlCl3 + D-gal+Vit.D. Novel object recognition (NOR), Morris Water Maze, and passive avoidance (PA) tests were used to measure memory abilities. The hippocampal tissue was used to assess vitamin D3 receptor (VDR) and peroxisome-proliferator-activated-receptor-γ-coactivator-1α (PGC-1α) expression by quantitative real-time polymerase chain reaction (qRT-PCR), CAMKK2, p-SIRT1, phosphorylated-AMP-activated protein kinase (p-AMPK), dynamin-related-protein-1 (Drp1), and mitofusin-1 (Mnf1) proteins by western blot and Ca2+ levels, endothelial nitic oxide synthase (eNOS), superoxide dismutase (SOD), amyloid beta (Aβ), and phospho tau (p-Tau) via enzyme-linked immunosorbent assay(ELISA) in addition to histological and ultrastructural examination of rat's brain tissue. Vit.D-attenuated hippocampal injury reversed the cognitive decline and Aβ aggregation, and elevated p-Tau levels in the AlCl3 + D-gal-induced AD rat model. In AlCl3 + D-gal-exposed rats, Vit.D induced VDR expression, normalized Ca2+ levels, elevated CAMKK2, p-AMPK, p-SIRT1, and PGC-1α expression. Vit.D reduced Drp1, induced Mnf1, increased mitochondrial membrane potential, preserved mitochondrial structure, restored normal mitochondrial function, and retained normal eNOS level and SOD activity in AlCl3 + D-gal rats. In conclusion, our findings proved that Vit.D may ameliorate cognitive deficits in AlCl3 + D-gal-induced AD by restoring normal mitochondrial function and reducing inflammatory and oxidative stress via CAMKK2-AMPK/SIRT1 pathway upregulation.
Collapse
Affiliation(s)
- Marwa Mohanad
- Department of Biochemistry, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| | - Shimaa K Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Basma E Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha A E Ahmed
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), Giza, Egypt
| |
Collapse
|
32
|
Czerwińska K, Januszewska L, Markiewicz-Górka I, Jaremków A, Martynowicz H, Pawlas K, Mazur G, Poręba R, Gać P. Selenoprotein P, peroxiredoxin-5, renalase, and total antioxidant status in patients with suspected obstructive sleep apnea. Sleep Breath 2024; 28:211-219. [PMID: 37495908 PMCID: PMC10954901 DOI: 10.1007/s11325-023-02880-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023]
Abstract
PURPOSE The aim of this study was to investigate the relationship between selenoprotein P, peroxiredoxin-5, renalase, total antioxidant status (TAS), mean blood pressure (mBP), and apnea-hypopnea index (AHI). METHODS The study group consisted of 112 patients hospitalized to verify the diagnosis of obstructive sleep apnea (OSA). The inclusion criteria were consent to participate in the study and age ≥ 18 years. Patients with active proliferative disease, severe systemic diseases, or mental diseases were excluded from the study. Each patient underwent full polysomnography and had blood pressure measured. Blood samples were collected and laboratory test was performed. RESULTS Among 112 patients enrolled, there was a statistically significant negative linear correlation between blood pressure values (sBP, dBP, mBP) and selenoprotein P, renalase, and TAS levels. Similarly, there was a negative linear correlation between AHI and selenoprotein P, renalase, and TAS levels, but none between AHI and peroxiredoxin-5. Based on the obtained regression models, higher selenoprotein P, peroxiredoxin-5, and renalase levels were independently associated with higher TAS. Lower mBP values were independently associated with the use of antihypertensive drugs, higher TAS, and younger age. Male gender, higher BMI, and higher mBP were independently associated with higher AHI. CONCLUSIONS Higher concentrations of selenoprotein P, peroxiredoxin-5, and renalase were associated with higher TAS, which confirms their antioxidant properties. There was an indirect connection between tested antioxidants and blood pressure values.
Collapse
Affiliation(s)
- Karolina Czerwińska
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Lidia Januszewska
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Iwona Markiewicz-Górka
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Aleksandra Jaremków
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Helena Martynowicz
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, PL, Poland
| | - Krystyna Pawlas
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland
| | - Grzegorz Mazur
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, PL, Poland
| | - Rafał Poręba
- Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556, Wroclaw, PL, Poland
| | - Paweł Gać
- Division of Environmental Health and Occupational Medicine, Department of Population Health, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368, Wroclaw, PL, Poland.
| |
Collapse
|
33
|
Twyning MJ, Tufi R, Gleeson TP, Kolodziej KM, Campesan S, Terriente-Felix A, Collins L, De Lazzari F, Giorgini F, Whitworth AJ. Partial loss of MCU mitigates pathology in vivo across a diverse range of neurodegenerative disease models. Cell Rep 2024; 43:113681. [PMID: 38236772 DOI: 10.1016/j.celrep.2024.113681] [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: 03/31/2023] [Revised: 11/01/2023] [Accepted: 01/02/2024] [Indexed: 03/02/2024] Open
Abstract
Mitochondrial calcium (Ca2+) uptake augments metabolic processes and buffers cytosolic Ca2+ levels; however, excessive mitochondrial Ca2+ can cause cell death. Disrupted mitochondrial function and Ca2+ homeostasis are linked to numerous neurodegenerative diseases (NDs), but the impact of mitochondrial Ca2+ disruption is not well understood. Here, we show that Drosophila models of multiple NDs (Parkinson's, Huntington's, Alzheimer's, and frontotemporal dementia) reveal a consistent increase in neuronal mitochondrial Ca2+ levels, as well as reduced mitochondrial Ca2+ buffering capacity, associated with increased mitochondria-endoplasmic reticulum contact sites (MERCs). Importantly, loss of the mitochondrial Ca2+ uptake channel MCU or overexpression of the efflux channel NCLX robustly suppresses key pathological phenotypes across these ND models. Thus, mitochondrial Ca2+ imbalance is a common feature of diverse NDs in vivo and is an important contributor to the disease pathogenesis. The broad beneficial effects from partial loss of MCU across these models presents a common, druggable target for therapeutic intervention.
Collapse
Affiliation(s)
- Madeleine J Twyning
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Roberta Tufi
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Thomas P Gleeson
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Kinga M Kolodziej
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Susanna Campesan
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Ana Terriente-Felix
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Lewis Collins
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Federica De Lazzari
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Alexander J Whitworth
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK.
| |
Collapse
|
34
|
O’Day DH. The Complex Interplay between Toxic Hallmark Proteins, Calmodulin-Binding Proteins, Ion Channels, and Receptors Involved in Calcium Dyshomeostasis in Neurodegeneration. Biomolecules 2024; 14:173. [PMID: 38397410 PMCID: PMC10886625 DOI: 10.3390/biom14020173] [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: 01/01/2024] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Calcium dyshomeostasis is an early critical event in neurodegeneration as exemplified by Alzheimer's (AD), Huntington's (HD) and Parkinson's (PD) diseases. Neuronal calcium homeostasis is maintained by a diversity of ion channels, buffers, calcium-binding protein effectors, and intracellular storage in the endoplasmic reticulum, mitochondria, and lysosomes. The function of these components and compartments is impacted by the toxic hallmark proteins of AD (amyloid beta and Tau), HD (huntingtin) and PD (alpha-synuclein) as well as by interactions with downstream calcium-binding proteins, especially calmodulin. Each of the toxic hallmark proteins (amyloid beta, Tau, huntingtin, and alpha-synuclein) binds to calmodulin. Multiple channels and receptors involved in calcium homeostasis and dysregulation also bind to and are regulated by calmodulin. The primary goal of this review is to show the complexity of these interactions and how they can impact research and the search for therapies. A secondary goal is to suggest that therapeutic targets downstream from calcium dyshomeostasis may offer greater opportunities for success.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
35
|
Kushnireva L, Segal M, Korkotian E. Cultured Rat Hippocampal Neurons Exposed to the Mitochondrial Uncoupler Carbonyl Cyanide Chlorophenylhydrazone Undergo a Rapid, Presenilin-Dependent Change in Neuronal Properties. Int J Mol Sci 2024; 25:578. [PMID: 38203751 PMCID: PMC10779238 DOI: 10.3390/ijms25010578] [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: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Presenilin 1 (PS1) is a transmembrane proteolytic subunit of γ-secretase that cleaves amyloid precursor proteins. Mutations in PS1 (mPS1) are associated with early-onset familial Alzheimer's disease (AD). The link between mutated PS1, mitochondrial calcium regulation, and AD has been studied extensively in different test systems. Despite the wide-ranging role of mPS1 in AD, there is a paucity of information on the link between PS1 and neuronal cell death, a hallmark of AD. In the present study, we employed the selective mitochondrial uncoupler carbonyl cyanide chlorophenylhydrazone (CCCP) and compared the reactivity of mPS1-transfected cultured rat hippocampal neurons with PS1 and control neurons in a situation of impaired mitochondrial functions. CCCP causes a slow rise in cytosolic and mitochondrial calcium in all three groups of neurons, with the mPS1 neurons demonstrating a faster rise. Consequently, mPS1 neurons were depolarized by CCCP and measured with TMRM, a mitochondrial voltage indicator, more than the other two groups. Morphologically, CCCP produced more filopodia in mPS1 neurons than in the other two groups, which were similarly affected by the drug. Finally, mPS1 transfected neurons tended to die from prolonged exposure to CCCP sooner than the other groups, indicating an increase in vulnerability associated with a lower ability to regulate excess cytosolic calcium.
Collapse
Affiliation(s)
- Liliia Kushnireva
- Faculty of Biology, Perm State University, 614068 Perm, Russia;
- Department of Immunology and Regenerative Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Menahem Segal
- Department of Brain Sciences, The Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Eduard Korkotian
- Department of Brain Sciences, The Weizmann Institute of Science, Rehovot 7610001, Israel;
| |
Collapse
|
36
|
Sun XN, An YA, Paschoal VA, de Souza CO, Wang MY, Vishvanath L, Bueno LM, Cobb AS, Nieto Carrion JA, Ibe ME, Li C, Kidd HA, Chen S, Li W, Gupta RK, Oh DY. GPR84-mediated signal transduction affects metabolic function by promoting brown adipocyte activity. J Clin Invest 2023; 133:e168992. [PMID: 37856216 PMCID: PMC10721148 DOI: 10.1172/jci168992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023] Open
Abstract
The G protein-coupled receptor 84 (GPR84), a medium-chain fatty acid receptor, has garnered attention because of its potential involvement in a range of metabolic conditions. However, the precise mechanisms underlying this effect remain elusive. Our study has shed light on the pivotal role of GPR84, revealing its robust expression and functional significance within brown adipose tissue (BAT). Mice lacking GPR84 exhibited increased lipid accumulation in BAT, rendering them more susceptible to cold exposure and displaying reduced BAT activity compared with their WT counterparts. Our in vitro experiments with primary brown adipocytes from GPR84-KO mice revealed diminished expression of thermogenic genes and reduced O2 consumption. Furthermore, the application of the GPR84 agonist 6-n-octylaminouracil (6-OAU) counteracted these effects, effectively reinstating the brown adipocyte activity. These compelling in vivo and in vitro findings converge to highlight mitochondrial dysfunction as the primary cause of BAT anomalies in GPR84-KO mice. The activation of GPR84 induced an increase in intracellular Ca2+ levels, which intricately influenced mitochondrial respiration. By modulating mitochondrial Ca2+ levels and respiration, GPR84 acts as a potent molecule involved in BAT activity. These findings suggest that GPR84 is a potential therapeutic target for invigorating BAT and ameliorating metabolic disorders.
Collapse
Affiliation(s)
- Xue-Nan Sun
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yu A. An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| | - Vivian A. Paschoal
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Camila O. de Souza
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - May-yun Wang
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lavanya Vishvanath
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Lorena M.A. Bueno
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ayanna S. Cobb
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph A. Nieto Carrion
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Madison E. Ibe
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chao Li
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Harrison A. Kidd
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Wenhong Li
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rana K. Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Division of Endocrinology, Department of Medicine, Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Da Young Oh
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
37
|
Lee HJ, Chae CW, Han HJ. Enhancing the therapeutic efficacy of mesenchymal stem cell transplantation in diabetes: Amelioration of mitochondrial dysfunction-induced senescence. Biomed Pharmacother 2023; 168:115759. [PMID: 37865993 DOI: 10.1016/j.biopha.2023.115759] [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: 08/25/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
Mesenchymal stem cell (MSC) transplantation offers significant potential for the treatment of diabetes mellitus (DM) and its complications. However, hyperglycemic conditions can induce senescence and dysfunction in both transplanted and resident MSCs, thereby limiting their therapeutic potential. Mitochondrial dysfunction and oxidative stress are key contributors to this process in MSCs exposed to hyperglycemia. As such, strategies aimed at mitigating mitochondrial dysfunction could enhance the therapeutic efficacy of MSC transplantation in DM. In this review, we provide an updated overview of how mitochondrial dysfunction mediates MSC senescence. We present experimental evidence for the molecular mechanisms behind high glucose-induced mitochondrial dysfunction in MSCs, which include impairment of mitochondrial biogenesis, mitochondrial calcium regulation, the mitochondrial antioxidant system, mitochondrial fusion-fission dynamics, mitophagy, and intercellular mitochondrial transfer. Furthermore, we propose potential pharmacological candidates that could improve the efficacy of MSC transplantation by enhancing mitochondrial function in patients with DM and related complications.
Collapse
Affiliation(s)
- Hyun Jik Lee
- Laboratory of Veterinary Physiology, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Republic of South Korea; Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju 28644, Republic of South Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
38
|
Ghosh S, Dahiya M, Kumar A, Bheri M, Pandey GK. Calcium imaging: a technique to monitor calcium dynamics in biological systems. PHYSIOLOGY AND MOLECULAR BIOLOGY OF PLANTS : AN INTERNATIONAL JOURNAL OF FUNCTIONAL PLANT BIOLOGY 2023; 29:1777-1811. [PMID: 38222278 PMCID: PMC10784449 DOI: 10.1007/s12298-023-01405-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/16/2024]
Abstract
Calcium ion (Ca2+) is a multifaceted signaling molecule that acts as an important second messenger. During the course of evolution, plants and animals have developed Ca2+ signaling in order to respond against diverse stimuli, to regulate a large number of physiological and developmental pathways. Our understanding of Ca2+ signaling and its components in physiological phenomena ranging from lower to higher organisms, and from single cell to multiple tissues has grown exponentially. The generation of Ca2+ transients or signatures for various stress factor is a well-known mechanism adopted in plant and animal systems. However, the decoding of such remarkable signatures is an uphill task and is always an interesting goal for the scientific community. In the past few decades, studies on the concentration and dynamics of intracellular Ca2+ are significantly increasing and have become a trend in modern biology. The advancement in approaches from Ca2+ binding dyes to in vivo Ca2+ imaging through the use of Ca2+ biosensors to achieve spatio-temporal resolution in micro and milliseconds range, provide us phenomenal opportunities to study live cell Ca2+ imaging or dynamics. Here, we describe the usage, improvement and advancement of Ca2+ based dyes, genetically encoded probes and sensors to achieve extraordinary Ca2+ imaging in plants and animals. Graphical abstract
Collapse
Affiliation(s)
- Soma Ghosh
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Monika Dahiya
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Amit Kumar
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Malathi Bheri
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| | - Girdhar K. Pandey
- Department of Plant Molecular Biology, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, 110021 India
| |
Collapse
|
39
|
Sakas R, Dan K, Edelman D, Abu-Ata S, Ben-Menashe A, Awad-Igbaria Y, Francois-Soustiel J, Palzur E. Hyperbaric Oxygen Therapy Alleviates Memory and Motor Impairments Following Traumatic Brain Injury via the Modulation of Mitochondrial-Dysfunction-Induced Neuronal Apoptosis in Rats. Antioxidants (Basel) 2023; 12:2034. [PMID: 38136154 PMCID: PMC10740762 DOI: 10.3390/antiox12122034] [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: 10/14/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality in young adults, characterized by primary and secondary injury. Primary injury is the immediate mechanical damage, while secondary injury results from delayed neuronal death, often linked to mitochondrial damage accumulation. Hyperbaric oxygen therapy (HBOT) has been proposed as a potential treatment for modulating secondary post-traumatic neuronal death. However, the specific molecular mechanism by which HBOT modulates secondary brain damage through mitochondrial protection remains unclear. Spatial learning, reference memory, and motor performance were measured in rats before and after Controlled Cortical Impact (CCI) injury. The HBOT (2.5 ATA) was performed 4 h following the CCI and twice daily (12 h intervals) for four consecutive days. Mitochondrial functions were assessed via high-resolution respirometry on day 5 following CCI. Moreover, IHC was performed at the end of the experiment to evaluate cortical apoptosis, neuronal survival, and glial activation. The current result indicates that HBOT exhibits a multi-level neuroprotective effect. Thus, we found that HBOT prevents cortical neuronal loss, reduces the apoptosis marker (cleaved-Caspase3), and modulates glial cell proliferation. Furthermore, HBO treatment prevents the reduction in mitochondrial respiration, including non-phosphorylation state, oxidative phosphorylation, and electron transfer capacity. Additionally, a superior motor and spatial learning performance level was observed in the CCI group treated with HBO compared to the CCI group. In conclusion, our findings demonstrate that HBOT during the critical period following the TBI improves cognitive and motor damage via regulating glial proliferation apoptosis and protecting mitochondrial function, consequently preventing cortex neuronal loss.
Collapse
Affiliation(s)
- Reem Sakas
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; (R.S.); (K.D.); (S.A.-A.); (A.B.-M.); (J.F.-S.)
- Research Institute of Galilee Medical Center, Nahariya 221001, Israel
| | - Katya Dan
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; (R.S.); (K.D.); (S.A.-A.); (A.B.-M.); (J.F.-S.)
- Research Institute of Galilee Medical Center, Nahariya 221001, Israel
| | - Doron Edelman
- Neurosurgery Department, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel;
| | - Saher Abu-Ata
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; (R.S.); (K.D.); (S.A.-A.); (A.B.-M.); (J.F.-S.)
- Research Institute of Galilee Medical Center, Nahariya 221001, Israel
| | - Aviv Ben-Menashe
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; (R.S.); (K.D.); (S.A.-A.); (A.B.-M.); (J.F.-S.)
- Research Institute of Galilee Medical Center, Nahariya 221001, Israel
| | - Yaseen Awad-Igbaria
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; (R.S.); (K.D.); (S.A.-A.); (A.B.-M.); (J.F.-S.)
- Research Institute of Galilee Medical Center, Nahariya 221001, Israel
| | - Jean Francois-Soustiel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; (R.S.); (K.D.); (S.A.-A.); (A.B.-M.); (J.F.-S.)
- Neurosurgery Department, Galilee Medical Center, Nahariya 221001, Israel
| | - Eilam Palzur
- Research Institute of Galilee Medical Center, Nahariya 221001, Israel
| |
Collapse
|
40
|
Camacho-Pereira J, Lai de Souza LO, Chichierchio MS, Rodrigues-Chaves C, Lomba LDS, Fonseca-Oliveira M, Carvalho-Mendonça D, Silva-Rodrigues T, Galina A. The NADase CD38 may not dictate NAD levels in brain mitochondria of aged mice but regulates hydrogen peroxide generation. Free Radic Biol Med 2023; 209:29-39. [PMID: 37774804 DOI: 10.1016/j.freeradbiomed.2023.09.035] [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: 07/31/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023]
Abstract
Aging is a time-related functional decline that affects many species. One of the hallmarks of aging is mitochondrial dysfunction, which leads to metabolic decline. The NAD decline during aging, in several tissues, correlates with increase in NADase activity of CD38. Knock out or pharmacological inhibition of CD38 activity can rescue mitochondrial function in several tissues, however, the role of CD38 in controlling NAD levels and metabolic function in the aging brain is unknown. In this work, we investigated CD38 NADase activity controlling NAD levels and mitochondrial function in mice brain with aging. We demonstrate that NADase activity of CD38 does not dictate NAD total levels in brain of aging mice and does not control mitochondrial oxygen consumption nor other oxygen parameters markers of mitochondrial dysfunction. However, for the first time we show that CD38 regulates hydrogen peroxide (H2O2) generation, one of the reactive oxygen species (ROS) in aging brain, through regulation of pyruvate dehydrogenase and alfa-ketoglutarate dehydrogenase, as mitochondria H2O2 leakage sites. The effect may be related to mitochondrial calcium handling differences in CD38 absence. Our study highlights a novel role of CD38 in brain energy metabolism and aging.
Collapse
Affiliation(s)
- Juliana Camacho-Pereira
- Laboratório de Biologia Molecular de Leveduras, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Leonardo Osbourne Lai de Souza
- Laboratório de Biologia Molecular de Leveduras, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Marina Santos Chichierchio
- Laboratório de Biologia Molecular de Leveduras, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Camila Rodrigues-Chaves
- Laboratório de Bioenergética e Fisiologia Mitocondrial, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Luiza de Sousa Lomba
- Laboratório de Bioenergética e Fisiologia Mitocondrial, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Manoel Fonseca-Oliveira
- Laboratório de Bioenergética e Fisiologia Mitocondrial, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Daniel Carvalho-Mendonça
- Laboratório de Biologia Molecular de Leveduras, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Thaia Silva-Rodrigues
- Laboratório de Bioenergética e Fisiologia Mitocondrial, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| | - Antonio Galina
- Laboratório de Bioenergética e Fisiologia Mitocondrial, Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Cidade Universitária, Av. Carlos Chagas Filho 373, Ilha do Fundão, Rio de Janeiro, 21941-590, Brazil.
| |
Collapse
|
41
|
Mironov VF, Dimukhametov MN, Nemtarev AV, Pashirova TN, Tsepaeva OV, Voloshina AD, Vyshtakalyuk AB, Litvinov IA, Lyubina AP, Sapunova AS, Abramova DF, Zobov VV. Novel Mitochondria-Targeted Amphiphilic Aminophosphonium Salts and Lipids Nanoparticles: Synthesis, Antitumor Activity and Toxicity. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2840. [PMID: 37947686 PMCID: PMC10649961 DOI: 10.3390/nano13212840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/17/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023]
Abstract
The creation of mitochondria-targeted vector systems is a new tool for the treatment of socially significant diseases. Phosphonium groups provide targeted delivery of drugs through biological barriers to organelles. For this purpose, a new class of alkyl(diethylAmino)(Phenyl) Phosphonium halides (APPs) containing one, two, or three diethylamino groups was obtained by the reaction of alkyl iodides (bromides) with (diethylamino)(phenyl)phosphines under mild conditions (20 °C) and high yields (93-98%). The structure of APP was established by NMR and XRD. A high in vitro cytotoxicity of APPs against M-HeLa, HuTu 80, PC3, DU-145, PANC-1, and MCF-7 lines was found. The selectivity index is in the range of 0.06-4.0 μM (SI 17-277) for the most active APPs. The effect of APPs on cancer cells is characterized by hyperproduction of ROS and depolarization of the mitochondrial membrane. APPs induce apoptosis, proceeding along the mitochondrial pathway. Incorporation of APPs into lipid systems (liposomes and solid lipid nanoparticles) improves cytotoxicity toward tumor cells and decrease toxicity against normal cell lines. The IC50s of lipid systems are lower than for the reference drug DOX, with a high SI (30-56) toward MCF-7 and DU-145. APPs exhibit high selective activity against Gram-positive bacteria S. aureus 209P and B. segeus 8035, including methicillin-resistant S. aureus (MRSA-1, MRSA-2), comparable to the activity of the fluoroquinolone antibiotic norfloxacin. A moderate in vivo toxicity in CD-1 mice was established for the lead APP.
Collapse
Affiliation(s)
- Vladimir F. Mironov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
- Alexander Butlerov Institute of Chemistry, Kazan (Volga Region) Federal University, 18 Kremlevskaya St., 420008 Kazan, Russia
| | - Mudaris N. Dimukhametov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Andrey V. Nemtarev
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
- Alexander Butlerov Institute of Chemistry, Kazan (Volga Region) Federal University, 18 Kremlevskaya St., 420008 Kazan, Russia
| | - Tatiana N. Pashirova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Olga V. Tsepaeva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Alexandra D. Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Alexandra B. Vyshtakalyuk
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Igor A. Litvinov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Anna P. Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Anastasiia S. Sapunova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Dinara F. Abramova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| | - Vladimir V. Zobov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia; (M.N.D.); (A.V.N.); (T.N.P.); (O.V.T.); (A.D.V.); (A.B.V.); (I.A.L.); (A.P.L.); (A.S.S.); (D.F.A.); (V.V.Z.)
| |
Collapse
|
42
|
Hao S, Huang H, Ma RY, Zeng X, Duan CY. Multifaceted functions of Drp1 in hypoxia/ischemia-induced mitochondrial quality imbalance: from regulatory mechanism to targeted therapeutic strategy. Mil Med Res 2023; 10:46. [PMID: 37833768 PMCID: PMC10571487 DOI: 10.1186/s40779-023-00482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Hypoxic-ischemic injury is a common pathological dysfunction in clinical settings. Mitochondria are sensitive organelles that are readily damaged following ischemia and hypoxia. Dynamin-related protein 1 (Drp1) regulates mitochondrial quality and cellular functions via its oligomeric changes and multiple modifications, which plays a role in mediating the induction of multiple organ damage during hypoxic-ischemic injury. However, there is active controversy and gaps in knowledge regarding the modification, protein interaction, and functions of Drp1, which both hinder and promote development of Drp1 as a novel therapeutic target. Here, we summarize recent findings on the oligomeric changes, modification types, and protein interactions of Drp1 in various hypoxic-ischemic diseases, as well as the Drp1-mediated regulation of mitochondrial quality and cell functions following ischemia and hypoxia. Additionally, potential clinical translation prospects for targeting Drp1 are discussed. This review provides new ideas and targets for proactive interventions on multiple organ damage induced by various hypoxic-ischemic diseases.
Collapse
Affiliation(s)
- Shuai Hao
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002 China
| | - He Huang
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Rui-Yan Ma
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037 China
| | - Xue Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400010 China
| | - Chen-Yang Duan
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| |
Collapse
|
43
|
Shao F, Han J, Tian Z, Wang Z, Liu S, Wu Y. Synergistic ROS generation and directional overloading of endogenous calcium induce mitochondrial dysfunction in living cells. Biomaterials 2023; 301:122284. [PMID: 37619266 DOI: 10.1016/j.biomaterials.2023.122284] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Taking advantage of endogenous Ca2+ to upregulate intramitochondrial Ca2+ level has become a powerful mean for mitochondrial dysfunction-mediated tumor therapy. However, the Ca2+ entered into mitochondria is limited ascribing to the uncontrollability and non-selectivity of endogenous Ca2+ transport. It remains a great challenge to make the maximum use of endogenous Ca2+ to ensure sufficient Ca2+ overloading in mitochondria. Herein, we smartly fabricate an intracellular Ca2+ directional transport channel to selectively transport endogenous Ca2+ from endoplasmic reticulum (ER) to mitochondria based on cascade release nanoplatform ABT-199@liposomes/doxorubicin@FeIII-tannic acid (ABT@Lip/DOX@Fe-TA). In tumor acidic microenvironment, Fe3+ ions are firstly released and reduced by tannic acid (TA) to Fe2+ for ROS generation. Subsequently, under the NIR light irradiation, the released ABT-199 molecules combine with ROS contribute to the formation of IP3R-Grp75-VDAC1 channel between ER and mitochondria, thus Ca2+ ions are directionally delivered and intramitochondrial Ca2+ level is significantly upregulated. The synergetic ROS generation and mitochondrial Ca2+ overloading effectively intensifies mitochondrial dysfunction, thereby achieving efficient tumor inhibition. This work presents a new insight and promising avenue for endogenous Ca2+-involved tumor therapies.
Collapse
Affiliation(s)
- Fengying Shao
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, State Key Laboratory of Digital Medical Engineering, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Jianyu Han
- School of Energy and Environment, Southeast University, Nanjing 211189, China
| | - Zhaoyan Tian
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Zhi Wang
- Wuxi Institute of Inspection, Testing and Certification, Wuxi 214125, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, State Key Laboratory of Digital Medical Engineering, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yafeng Wu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, State Key Laboratory of Digital Medical Engineering, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| |
Collapse
|
44
|
Harrington JS, Ryter SW, Plataki M, Price DR, Choi AMK. Mitochondria in health, disease, and aging. Physiol Rev 2023; 103:2349-2422. [PMID: 37021870 PMCID: PMC10393386 DOI: 10.1152/physrev.00058.2021] [Citation(s) in RCA: 129] [Impact Index Per Article: 129.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are well known as organelles responsible for the maintenance of cellular bioenergetics through the production of ATP. Although oxidative phosphorylation may be their most important function, mitochondria are also integral for the synthesis of metabolic precursors, calcium regulation, the production of reactive oxygen species, immune signaling, and apoptosis. Considering the breadth of their responsibilities, mitochondria are fundamental for cellular metabolism and homeostasis. Appreciating this significance, translational medicine has begun to investigate how mitochondrial dysfunction can represent a harbinger of disease. In this review, we provide a detailed overview of mitochondrial metabolism, cellular bioenergetics, mitochondrial dynamics, autophagy, mitochondrial damage-associated molecular patterns, mitochondria-mediated cell death pathways, and how mitochondrial dysfunction at any of these levels is associated with disease pathogenesis. Mitochondria-dependent pathways may thereby represent an attractive therapeutic target for ameliorating human disease.
Collapse
Affiliation(s)
- John S Harrington
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | | | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - David R Price
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
45
|
Leri M, Vasarri M, Barletta E, Schiavone N, Bergonzi MC, Bucciantini M, Degl’Innocenti D. The Protective Role of Oleuropein Aglycone against Pesticide-Induced Toxicity in a Human Keratinocytes Cell Model. Int J Mol Sci 2023; 24:14553. [PMID: 37834001 PMCID: PMC10572371 DOI: 10.3390/ijms241914553] [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: 08/16/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
The extensive use of agricultural pesticides to improve crop quality and yield significantly increased the risk to the public of exposure to small but repeated doses of pesticides over time through various routes, including skin, by increasing the risk of disease outbreaks. Although much work was conducted to reduce the use of pesticides in agriculture, little attention was paid to prevention, which could reduce the toxicity of pesticide exposure by reducing its impact on human health. Extra virgin olive oil (EVOO), a major component of the Mediterranean diet, exerts numerous health-promoting properties, many of which are attributed to oleuropein aglycone (OleA), the deglycosylated form of oleuropein, which is the main polyphenolic component of EVOO. In this work, three pesticides with different physicochemical and biological properties, namely oxadiazon (OXA), imidacloprid (IMID), and glyphosate (GLYPHO), were compared in terms of metabolic activity, mitochondrial function and epigenetic modulation in an in vitro cellular model of human HaCaT keratinocytes to mimic the pathway of dermal exposure. The potential protective effect of OleA against pesticide-induced cellular toxicity was then evaluated in a cell pre-treatment condition. This study showed that sub-lethal doses of OXA and IMID reduced the metabolic activity and mitochondrial functionality of HaCaT cells by inducing oxidative stress and altering intracellular calcium flux and caused epigenetic modification by reducing histone acetylation H3 and H4. GLYPHO, on the other hand, showed no evidence of cellular toxicity at the doses tested. Pretreatment of cells with OleA was able to protect cells from the damaging effects of the pesticides OXA and IMID by maintaining metabolic activity and mitochondrial function at a controlled level and preventing acetylation reduction, particularly of histone H3. In conclusion, the bioactive properties of OleA reported here could be of great pharmaceutical and health interest, as they could be further studied to design new formulations for the prevention of toxicity from exposure to pesticide use.
Collapse
Affiliation(s)
- Manuela Leri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Marzia Vasarri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
- Department of Chemistry, University of Florence, Via U. Schiff 6, 50519 Sesto Fiorentino, Italy;
| | - Emanuela Barletta
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Nicola Schiavone
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Maria Camilla Bergonzi
- Department of Chemistry, University of Florence, Via U. Schiff 6, 50519 Sesto Fiorentino, Italy;
| | - Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| | - Donatella Degl’Innocenti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.L.); (M.V.); (E.B.); (N.S.); (M.B.)
| |
Collapse
|
46
|
Sharlo KA, Lvova ID, Tyganov SA, Zaripova KA, Belova SP, Kostrominova TY, Shenkman BS, Nemirovskaya TL. The Effect of SERCA Activation on Functional Characteristics and Signaling of Rat Soleus Muscle upon 7 Days of Unloading. Biomolecules 2023; 13:1354. [PMID: 37759754 PMCID: PMC10526198 DOI: 10.3390/biom13091354] [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: 07/11/2023] [Revised: 08/28/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Skeletal muscle abnormalities and atrophy during unloading are accompanied by the accumulation of excess calcium in the sarcoplasm. We hypothesized that calcium accumulation may occur, among other mechanisms, due to the inhibition of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) activity. Consequently, the use of the SERCA activator will reduce the level of calcium in the sarcoplasm and prevent the negative consequences of muscle unloading. Wistar rats were randomly assigned into one of three groups (eight rats per group): control rats with placebo (C), 7 days of unloading/hindlimb suspension with placebo (7HS), and 7 days of unloading treated with SERCA activator CDN1163 (7HSC). After seven days of unloading the soleus muscle, the 7HS group displayed increased fatigue in the ex vivo test, a significant increase in the level of calcium-dependent CaMK II phosphorylation and the level of tropomyosin oxidation, as well as a decrease in the content of mitochondrial DNA and protein, slow-type myosin mRNA, and the percentage of slow-type muscle fibers. All of these changes were prevented in the 7HSC group. Moreover, treatment with CDN1163 blocked a decrease in the phosphorylation of p70S6k, an increase in eEF2 phosphorylation, and an increase in MuRF-1 mRNA expression. Nevertheless, there were no differences in the degree of fast and slow muscle fiber atrophy between the 7HS and 7HSC groups. Conclusion: SERCA activation during 7 days of unloading prevented an increase in soleus fatigue, the decrease of slow-type myosin, mitochondrial markers, and markers of calcium homeostasis but had no effect on muscle atrophy.
Collapse
Affiliation(s)
- Kristina A. Sharlo
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Irina D. Lvova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Sergey A. Tyganov
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Ksenia A. Zaripova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Svetlana P. Belova
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana Y. Kostrominova
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine-Northwest, Gary, IN 46202, USA;
| | - Boris S. Shenkman
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| | - Tatiana L. Nemirovskaya
- Myology Laboratory, Institute of Biomedical Problems, RAS (Russian Academy of Sciences), Moscow 123007, Russia; (K.A.S.); (I.D.L.); (S.A.T.); (K.A.Z.); (S.P.B.); (B.S.S.)
| |
Collapse
|
47
|
Roy R, Mandal PK, Maroon JC. Oxidative Stress Occurs Prior to Amyloid Aβ Plaque Formation and Tau Phosphorylation in Alzheimer's Disease: Role of Glutathione and Metal Ions. ACS Chem Neurosci 2023; 14:2944-2954. [PMID: 37561556 PMCID: PMC10485904 DOI: 10.1021/acschemneuro.3c00486] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Alzheimer's disease (AD) is an insidious and progressive neurodegenerative disorder that affects millions of people worldwide. Although the pathogenesis remains obscure, there are two dominant causal hypotheses. Since last three decades, amyloid beta (Aβ) deposition was the most prominent hypothesis, and the other is the tau hyperphosphorylation hypothesis. The confirmed diagnostic criterion for AD is the presence of neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau and the deposition of toxic oligomeric Aβ in the autopsied brain. Consistent with these hypotheses, oxidative stress (OS) is garnering major attention in AD research. OS results from an imbalance of pro-oxidants and antioxidants. There is a considerable debate in the scientific community on which process occurs first, OS or plaque deposition/tau hyperphosphorylation. Based on recent scientific observations of various laboratories including ours along with critical analysis of those information, we believe that OS is the early event that leads to oligomeric Aβ deposition as well as dimerization of tau protein and its subsequent hyperphosphorylation. This OS hypothesis immediately suggests the consideration of novel therapeutic approaches to include antioxidants involving glutathione enrichment in the brain by supplementation with or without an iron chelator.
Collapse
Affiliation(s)
- Rimil
Guha Roy
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122052, India
| | - Pravat K Mandal
- Neuroimaging
and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122052, India
- Florey
Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Melbourne, 3052 VIC, Australia
| | - Joseph C. Maroon
- Department
of Neurosurgery, University of Pittsburgh
Medical School, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
48
|
Popov LD. Mitochondria as intracellular signalling organelles. An update. Cell Signal 2023:110794. [PMID: 37422005 DOI: 10.1016/j.cellsig.2023.110794] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/23/2023] [Accepted: 07/02/2023] [Indexed: 07/10/2023]
Abstract
Traditionally, mitochondria are known as "the powerhouse of the cell," responsible for energy (ATP) generation (by the electron transport chain, oxidative phosphorylation, the tricarboxylic acid cycle, and fatty acid ß-oxidation), and for the regulation of several metabolic processes, including redox homeostasis, calcium signalling, and cellular apoptosis. The extensive studies conducted in the last decades portray mitochondria as multifaceted signalling organelles that ultimately command cells' survival or death. Based on current knowledge, we'll outline the mitochondrial signalling to other intracellular compartments in homeostasis and pathology-related mitochondrial stress conditions here. The following topics are discussed: (i) oxidative stress and mtROS signalling in mitohormesis, (ii) mitochondrial Ca2+ signalling; (iii) the anterograde (nucleus-to-mitochondria) and retrograde (mitochondria-to-nucleus) signal transduction, (iv) the mtDNA role in immunity and inflammation, (v) the induction of mitophagy- and apoptosis - signalling cascades, (vi) the mitochondrial dysfunctions (mitochondriopathies) in cardiovascular, neurodegenerative, and malignant diseases. The novel insights into molecular mechanisms of mitochondria-mediated signalling can explain mitochondria adaptation to metabolic and environmental stresses to achieve cell survival.
Collapse
Affiliation(s)
- Lucia-Doina Popov
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania.
| |
Collapse
|
49
|
Prasad Panda S, Kesharwani A. Micronutrients/miRs/ATP networking in mitochondria: Clinical intervention with ferroptosis, cuproptosis, and calcium burden. Mitochondrion 2023; 71:1-16. [PMID: 37172668 DOI: 10.1016/j.mito.2023.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/12/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
The mitochondrial electron transport chain (mtETC) requires mainly coenzyme Q10 (CoQ10), copper (Cu2+), calcium (Ca2+), and iron (Fe2+) ions for efficient ATP production. According to cross-sectional research, up to 50% of patients with micronutrient imbalances have been linked to oxidative stress, mitochondrial dysfunction, reduced ATP production, and the prognosis of various diseases. The condition of ferroptosis, which is caused by the downregulation of CoQ10 and the activation of non-coding micro RNAs (miRs), is strongly linked to free radical accumulation, cancer, and neurodegenerative diseases. The entry of micronutrients into the mitochondrial matrix depends upon the higher threshold level of mitochondrial membrane potential (ΔΨm), and high cytosolic micronutrients. The elevated micronutrient in the mitochondrial matrix causes the utilization of all ATP, leading to a drop in ATP levels. Mitochondrial calcium uniporter (MCU) and Na+/Ca2+ exchanger (NCX) play a major role in Ca2+ influx in the mitochondrial matrix. The mitochondrial Ca2+ overload is regulated by specific miRs such as miR1, miR7, miR25, miR145, miR138, and miR214, thereby reducing apoptosis and improving ATP production. Cuproptosis is primarily brought on by increased Cu+ build-up and mitochondrial proteotoxic stress, mediated by ferredoxin-1 (FDX1) and long non-coding RNAs. Cu importers (SLC31A1) and exporters (ATP7B) influence intracellular Cu2+ levels to control cuproptosis. According to literature reviews, very few randomized micronutrient interventions have been carried out, despite the identification of a high prevalence of micronutrient deficiencies. In this review, we concentrated on essential micronutrients and specific miRs associated with ATP production that balance oxidative stress in mitochondria.
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| |
Collapse
|
50
|
Dhureja M, Arthur R, Soni D, Upadhayay S, Temgire P, Kumar P. Calcium channelopathies in neurodegenerative disorder: an untold story of RyR and SERCA. Expert Opin Ther Targets 2023; 27:1159-1172. [PMID: 37971192 DOI: 10.1080/14728222.2023.2277863] [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: 08/26/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Recent neuroscience breakthroughs have shed light on the sophisticated relationship between calcium channelopathies and movement disorders, exposing a previously undiscovered tale focusing on the Ryanodine Receptor (RyR) and the Sarco/Endoplasmic Reticulum Calcium ATPase (SERCA). Calcium signaling mainly orchestrates neural communication, which regulates synaptic transmission and total network activity. It has been determined that RyR play a significant role in managing neuronal functions, most notably in releasing intracellular calcium from the endoplasmic reticulum. AREAS COVERED It highlights the involvement of calcium channels such as RyR and SERCA in physiological and pathophysiological conditions. EXPERT OPINION Links between RyR and SERCA activity dysregulation, aberrant calcium levels, motor and cognitive dysfunction have brought attention to the importance of RyR and SERCA modulation in neurodegenerative disorders. Understanding the obscure function of these proteins will open up new therapeutic possibilities to address the underlying causes of neurodegenerative diseases. The unreported RyR and SERCA narrative broadens the understanding of calcium channelopathies in movement disorders and calls for more research into cutting-edge therapeutic approaches.
Collapse
Affiliation(s)
- Maanvi Dhureja
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Divya Soni
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Pooja Temgire
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|