1
|
Yu H, Ren K, Jin Y, Zhang L, Liu H, Huang Z, Zhang Z, Chen X, Yang Y, Wei Z. Mitochondrial DAMPs: Key mediators in neuroinflammation and neurodegenerative disease pathogenesis. Neuropharmacology 2025; 264:110217. [PMID: 39557152 DOI: 10.1016/j.neuropharm.2024.110217] [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: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are increasingly linked to mitochondrial dysfunction and neuroinflammation. Central to this link are mitochondrial damage-associated molecular patterns (mtDAMPs), including mitochondrial DNA, ATP, and reactive oxygen species, released during mitochondrial stress or damage. These mtDAMPs activate inflammatory pathways, such as the NLRP3 inflammasome and cGAS-STING, contributing to the progression of neurodegenerative diseases. This review delves into the mechanisms by which mtDAMPs drive neuroinflammation and discusses potential therapeutic strategies targeting these pathways to mitigate neurodegeneration. Additionally, it explores the cross-talk between mitochondria and the immune system, highlighting the complex interplay that exacerbates neuronal damage. Understanding the role of mtDAMPs could pave the way for novel treatments aimed at modulating neuroinflammation and slowing disease progression, ultimately improving patient outcome.
Collapse
Affiliation(s)
- Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Ziheng Zhang
- College of Life Sciences, Xinjiang University, Urumqi, Xinjiang, 830046, PR China
| | - Xing Chen
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
2
|
Dubey S, Yu Z, Stephens EM, Lazrak A, Ahmad I, Aggarwal S, Andrabi S, Hossain MI, Jilling T, Fernadez SR, Bartels JL, Lapi SE, Mobley J, Pastukh VM, Gillespie M, Matalon S. Oxidative Injury to Lung Mitochondrial DNA is a Key Contributor for the Development of Chemical Lung Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624949. [PMID: 39651262 PMCID: PMC11623505 DOI: 10.1101/2024.11.22.624949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The mechanisms and extent to which inhalation of oxidant gases damage the mitochondrial genome contributing to the development of acute and chronic lung injury have not been investigated. C57BL/6 mice exposed to chlorine (Cl 2 ) gas and returned to room air, developed progressive loss of lung DNA glycosylase OGG1, significant oxidative injury to mtDNA, decreased intact lung mitochondrial (mt) DNA, generation of inflammatory pathway by DAMPs causing airway and alveolar injury with significant mortality. Global proteomics identified over 1400 lung proteins with alteration of key mitochondrial proteins at 24 h post Cl 2 exposure. Intranasal instillation of a recombinant protein containing mitochondrial targeted OGG1 (mitoOGG1) post exposure, decreased oxidative injury to mtDNA, lung mitochondrial proteome, severity of the acute and chronic lung injury and increased survival. These data show that injury to the mt-genome is a key contributor to the development of acute and chronic lung injury.
Collapse
|
3
|
Pan H, Su Q, Hong P, You Y, Zhou L, Zou J, Sun J, Zhong G, Liao J, Zhang H, Tang Z, Hu L. Arsenic-induced mtDNA release promotes inflammatory responses through cGAS-STING signaling in chicken hepatocytes. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 205:106129. [PMID: 39477583 DOI: 10.1016/j.pestbp.2024.106129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 11/07/2024]
Abstract
Arsenic is a toxic element that can cause severe liver damage in humans and animals. Arsenic-based inorganic pesticides, such as lead arsenate, copper arsenate, and calcium arsenate, are widely used for insect control and can eventually affect human health through accumulation in the food chain. However, the relationship between arsenic trioxide (ATO)-induced hepatotoxicity and the cGAS-STING signaling pathway has not been reported. The aim of this study was to investigate the potential role of inflammatory response in ATO-induced hepatotoxicity in chickens. In this study, we found that ATO exposure resulted in mtDNA leakage into the cytoplasm of chicken hepatocytes, which activated the cGAS-STING pathway and significantly increased the cGAS, STING, TBK1, and IRF7 mRNA and protein expression levels. Moreover, type I interferon response was activated. Concurrently, STING triggered the activation of the traditional NF-κB signaling pathway and promoted the expression of pro-inflammatory cytokine genes, including TNF-α, IL-6, and IL-1β. Subsequently, we found that both mtDNA clearance with EtBr and inhibition of the cGAS-STING pathway with H-151 reversed the ATO-induced innate immune and inflammatory responses. In summary, the above findings indicate that chicken hepatocytes can induce innate immune responses and inflammatory responses via mtDNA-cGAS-STING under ATO-exposure conditions, which is of great significance for further studies on the toxicity mechanism of ATO.
Collapse
Affiliation(s)
- Hang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Qian Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Panjing Hong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Yanli You
- College of Life Science, Yantai University, Yantai, 264005, Shandong Province, China.
| | - Limeng Zhou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Junbo Zou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jingping Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
4
|
Zhou G, Wang X, Guo M, Qu C, Gao L, Yu J, Li Y, Luo S, Shi Q, Guo Y. Mitophagy deficiency activates stimulator of interferon genes activation and aggravates pathogenetic cardiac remodeling. Genes Dis 2024; 11:101074. [PMID: 39281830 PMCID: PMC11399633 DOI: 10.1016/j.gendis.2023.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/11/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2024] Open
Abstract
Stimulator of interferon genes (STING) has recently been found to play a crucial role in cardiac sterile inflammation and dysfunction. The role of stimulator of interferon genes (STING) in cardiac sterile inflammation and dysfunction has been recently discovered. This study aims to examine the involvement of STING in pathological cardiac remodeling and the mechanisms that govern the activation of the STING pathway. To investigate this, transverse aortic constriction (TAC) was performed on STING knockout mice to induce pressure overload-induced cardiac remodeling. Subsequently, cardiac function, remodeling, and inflammation levels were evaluated. The STING pathway was found to be activated in the pressure overload-stressed heart and angiotensin II (Ang II)-stimulated cardiac fibroblasts. Loss of STING expression led to a significant reduction in inflammatory responses, mitochondrial fragmentation, and oxidative stress in the heart, resulting in attenuated cardiac remodeling and dysfunction. Furthermore, the exacerbation of pressure overload-induced STING-mediated inflammation and pathological cardiac remodeling was observed when mitophagy was suppressed through the silencing of Parkin, an E3 ubiquitin ligase. Taken together, these findings indicate that STING represents a newly identified and significant molecule implicated in the process of pathological cardiac remodeling and that mitophagy is an upstream mechanism that regulates STING activation. Targeting STING may therefore provide a novel therapeutic strategy for pathological cardiac remodeling and heart failure.
Collapse
Affiliation(s)
- Guoxiang Zhou
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaowen Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mingyu Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Can Qu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lei Gao
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiang Yu
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuanjing Li
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Suxin Luo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qiong Shi
- The Department of Laboratory Medicine, M.O.E. Key Laboratory of Laboratory Medical Diagnostics, Chongqing Medical University, Chongqing 400016, China
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
5
|
Kim J, Yuan Y, Agaronyan K, Zhao A, D Wang V, Gupta G, Essayas H, Kaminski A, McGovern J, Yu S, Woo S, Lee CJ, Gandhi S, Saber T, Saleh T, Hu B, Sun Y, Ishikawa G, Bain W, Evankovich J, Chen L, Yun H, Herzog EL, Dela Cruz CS, Ryu C, Sharma L. Damage sensing through TLR9 Regulates Inflammatory and Antiviral Responses During Influenza Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583378. [PMID: 38496452 PMCID: PMC10942338 DOI: 10.1101/2024.03.04.583378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Host response aimed at eliminating the infecting pathogen, as well as the pathogen itself, can cause tissue injury. Tissue injury leads to the release of a myriad of cellular components including mitochondrial DNA, which the host senses through pattern recognition receptors. How the sensing of tissue injury by the host shapes the anti-pathogen response remains poorly understood. In this study, we utilized mice that are deficient in toll-like receptor-9 (TLR9), which binds to unmethylated CpG DNA sequences such as those present in bacterial and mitochondrial DNA. To avoid direct pathogen sensing by TLR9, we utilized the influenza virus, which lacks ligands for TLR9, to determine how damage sensing by TLR9 contributes to anti-influenza immunity. Our data show that TLR9-mediated sensing of tissue damage promotes an inflammatory response during early infection, driven by the epithelial and myeloid cells. Along with the diminished inflammatory response, the absence of TLR9 led to impaired viral clearance manifested as a higher and prolonged influenza components in myeloid cells including monocytes and macrophages rendering them highly inflammatory. The persistent inflammation driven by infected myeloid cells led to persistent lung injury and impaired recovery in influenza-infected TLR9-/- mice. Further, we show elevated TLR9 activation in the plasma samples of patients with influenza and its association with the disease severity in hospitalized patients, demonstrating its clinical relevance. Overall, we demonstrate an essential role of damage sensing through TLR9 in promoting anti-influenza immunity and inflammatory response.
Collapse
Affiliation(s)
- Jooyoung Kim
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Yifan Yuan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- University of Maryland, MD
| | - Karen Agaronyan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- Howard Hughes Medical Institute
| | - Amy Zhao
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Victoria D Wang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Gayatri Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Heran Essayas
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Ayelet Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - John McGovern
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Sheeline Yu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Samuel Woo
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Chris J. Lee
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Shifa Gandhi
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Tina Saber
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Tayebeh Saleh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Ying Sun
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- VA Medical Center, Pittsburgh, PA
| | - John Evankovich
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Lujia Chen
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - HongDuck Yun
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Erica L. Herzog
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Charles S. Dela Cruz
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- VA Medical Center, Pittsburgh, PA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| |
Collapse
|
6
|
Islam MA, Sehar U, Sultana OF, Mukherjee U, Brownell M, Kshirsagar S, Reddy PH. SuperAgers and centenarians, dynamics of healthy ageing with cognitive resilience. Mech Ageing Dev 2024; 219:111936. [PMID: 38657874 DOI: 10.1016/j.mad.2024.111936] [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: 03/08/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Graceful healthy ageing and extended longevity is the most desired goal for human race. The process of ageing is inevitable and has a profound impact on the gradual deterioration of our physiology and health since it triggers the onset of many chronic conditions like dementia, osteoporosis, diabetes, arthritis, cancer, and cardiovascular disease. However, some people who lived/live more than 100 years called 'Centenarians" and how do they achieve their extended lifespans are not completely understood. Studying these unknown factors of longevity is important not only to establish a longer human lifespan but also to manage and treat people with shortened lifespans suffering from age-related morbidities. Furthermore, older adults who maintain strong cognitive function are referred to as "SuperAgers" and may be resistant to risk factors linked to cognitive decline. Investigating the mechanisms underlying their cognitive resilience may contribute to the development of therapeutic strategies that support the preservation of cognitive function as people age. The key to a long, physically, and cognitively healthy life has been a mystery to scientists for ages. Developments in the medical sciences helps us to a better understanding of human physiological function and greater access to medical care has led us to an increase in life expectancy. Moreover, inheriting favorable genetic traits and adopting a healthy lifestyle play pivotal roles in promoting longer and healthier lives. Engaging in regular physical activity, maintaining a balanced diet, and avoiding harmful habits such as smoking contribute to overall well-being. The synergy between positive lifestyle choices, access to education, socio-economic factors, environmental determinants and genetic supremacy enhances the potential for a longer and healthier life. Our article aims to examine the factors associated with healthy ageing, particularly focusing on cognitive health in centenarians. We will also be discussing different aspects of ageing including genomic instability, metabolic burden, oxidative stress and inflammation, mitochondrial dysfunction, cellular senescence, immunosenescence, and sarcopenia.
Collapse
Affiliation(s)
- Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Upasana Mukherjee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Malcolm Brownell
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- 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; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX 79409, USA.
| |
Collapse
|
7
|
Atarashi N, Morishita M, Matsuda S. Activation of innate immune receptor TLR9 by mitochondrial DNA plays essential roles in the chemical long-term depression of hippocampal neurons. J Biol Chem 2024; 300:105744. [PMID: 38354781 PMCID: PMC10943477 DOI: 10.1016/j.jbc.2024.105744] [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: 09/18/2023] [Revised: 01/11/2024] [Accepted: 02/04/2024] [Indexed: 02/16/2024] Open
Abstract
Synaptic plasticity is believed to be the cellular basis for experience-dependent learning and memory. Although long-term depression (LTD), a form of synaptic plasticity, is caused by the activity-dependent reduction of cell surface α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)-type glutamate receptors (AMPA receptors) at postsynaptic sites, its regulation by neuronal activity is not completely understood. In this study, we showed that the inhibition of toll-like receptor-9 (TLR9), an innate immune receptor, suppresses N-methyl-d-aspartate (NMDA)-induced reduction of cell surface AMPA receptors in cultured hippocampal neurons. We found that inhibition of TLR9 also blocked NMDA-induced activation of caspase-3, which plays an essential role in the induction of LTD. siRNA-based knockdown of TLR9 also suppressed the NMDA-induced reduction of cell surface AMPA receptors, although the scrambled RNA had no effect on the NMDA-induced trafficking of AMPA receptors. Overexpression of the siRNA-resistant form of TLR9 rescued the AMPA receptor trafficking abolished by siRNA. Furthermore, NMDA stimulation induced rapid mitochondrial morphological changes, mitophagy, and the binding of mitochondrial DNA (mtDNA) to TLR9. Treatment with dideoxycytidine and mitochondrial division inhibitor-1, which block mtDNA replication and mitophagy, respectively, inhibited NMDA-dependent AMPA receptor internalization. These results suggest that mitophagy induced by NMDA receptor activation releases mtDNA and activates TLR9, which plays an essential role in the trafficking of AMPA receptors during the induction of LTD.
Collapse
Affiliation(s)
- Naoya Atarashi
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - Misaki Morishita
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan
| | - Shinji Matsuda
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan; Center for Neuroscience and Biomedical Engineering (CNBE), The University of Electro-Communications, Tokyo, Japan.
| |
Collapse
|
8
|
Wang Y, Wang J, Tao SY, Liang Z, Xie R, Liu NN, Deng R, Zhang Y, Deng D, Jiang G. Mitochondrial damage-associated molecular patterns: A new insight into metabolic inflammation in type 2 diabetes mellitus. Diabetes Metab Res Rev 2024; 40:e3733. [PMID: 37823338 DOI: 10.1002/dmrr.3733] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/18/2023] [Accepted: 09/08/2023] [Indexed: 10/13/2023]
Abstract
The pathogenesis of diabetes is accompanied by increased levels of inflammatory factors, also known as "metabolic inflammation", which runs through the whole process of the occurrence and development of the disease. Mitochondria, as the key site of glucose and lipid metabolism, is often accompanied by mitochondrial function damage in type 2 diabetes mellitus (T2DM). Damaged mitochondria release pro-inflammatory factors through damage-related molecular patterns that activate inflammation pathways and reactions to oxidative stress, further aggravate metabolic disorders, and form a vicious circle. Currently, the pathogenesis of diabetes is still unclear, and clinical treatment focuses primarily on symptomatic intervention of the internal environment of disorders of glucose and lipid metabolism with limited clinical efficacy. The proinflammatory effect of mitochondrial damage-associated molecular pattern (mtDAMP) in T2DM provides a new research direction for exploring the pathogenesis and intervention targets of T2DM. Therefore, this review covers the most recent findings on the molecular mechanism and related signalling cascades of inflammation caused by mtDAMP in T2DM and discusses its pathogenic role of it in the pathological process of T2DM to search potential intervention targets.
Collapse
Affiliation(s)
- Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jingwu Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Si-Yu Tao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | | | - Rong Xie
- Xinjiang Medical University, Urumqi, China
| | - Nan-Nan Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ruxue Deng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuelin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Deqiang Deng
- Department of Endocrinology, Urumqi Hospital of Traditional Chinese Medicine, Urumqi, China
| | - Guangjian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Raghul Kannan S, Tamizhselvi R. N-acetyltransferase and inflammation: Bridging an unexplored niche. Gene 2023; 887:147730. [PMID: 37625560 DOI: 10.1016/j.gene.2023.147730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Protein N-terminal (Nt) acetylation is an essential post-translational process catalysed by N-acetyltransferases or N-terminal acetyltransferases (NATs). Over the past several decades, several types of NATs (NatA- NatH) have been identified along with their substrates, explaining their significance in eukaryotes. It affects protein stability, protein degradation, protein translocation, and protein-protein interaction. NATs have recently drawn attention as they are associated with the pathogenesis of human diseases. In particular, NAT-induced epigenetic modifications play an important role in the control of mitochondrial function, which may lead to inflammatory diseases. NatC knockdown causes a marked reduction in mitochondrial membrane proteins, impairing their functions, and NatA affects mitophagy via reduced phosphorylation and transcription of the autophagy receptor. However, the NAT-mediated mitochondrial epigenetic mechanisms involved in the inflammatory process remain unexplored. The current review will impart an overview of the biological functions and aberrations of various NAT, which may provide a novel therapeutic strategy for inflammatory disorders.
Collapse
Affiliation(s)
- Sampath Raghul Kannan
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Ramasamy Tamizhselvi
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
10
|
Yoon J, Kim S, Lee M, Kim Y. Mitochondrial nucleic acids in innate immunity and beyond. Exp Mol Med 2023; 55:2508-2518. [PMID: 38036728 PMCID: PMC10766607 DOI: 10.1038/s12276-023-01121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/12/2023] [Accepted: 08/23/2023] [Indexed: 12/02/2023] Open
Abstract
Mitochondria participate in a wide range of cellular processes. One essential function of mitochondria is to be a platform for antiviral signaling proteins during the innate immune response to viral infection. Recently, studies have revealed that mitochondrion-derived DNAs and RNAs are recognized as non-self molecules and act as immunogenic ligands. More importantly, the cytosolic release of these mitochondrial nucleic acids (mt-NAs) is closely associated with the pathogenesis of human diseases accompanying aberrant immune activation. The release of mitochondrial DNAs (mtDNAs) via BAX/BAK activation and/or VDAC1 oligomerization activates the innate immune response and inflammasome assembly. In addition, mitochondrial double-stranded RNAs (mt-dsRNAs) are sensed by pattern recognition receptors in the cytosol to induce type I interferon expression and initiate apoptotic programs. Notably, these cytosolic mt-NAs also mediate adipocyte differentiation and contribute to mitogenesis and mitochondrial thermogenesis. In this review, we summarize recent studies of innate immune signaling pathways regulated by mt-NAs, human diseases associated with mt-NAs, and the emerging physiological roles of mt-NAs.
Collapse
Affiliation(s)
- Jimin Yoon
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sujin Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Mihye Lee
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| | - Yoosik Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
- Graduate School of Engineering Biology, KAIST, Daejeon, 34141, Republic of Korea.
- KAIST Institute for BioCentury (KIB), KAIST, Daejeon, 34141, Republic of Korea.
- KAIST Institute for Health Science and Technology (KIHST), KAIST, Daejeon, 34141, Republic of Korea.
- BioProcess Engineering Research Center and BioInformatics Research Center, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
11
|
Galizzi G, Di Carlo M. Mitochondrial DNA and Inflammation in Alzheimer's Disease. Curr Issues Mol Biol 2023; 45:8586-8606. [PMID: 37998717 PMCID: PMC10670154 DOI: 10.3390/cimb45110540] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/15/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023] Open
Abstract
Mitochondrial dysfunction and neuroinflammation are implicated in the pathogenesis of most neurodegenerative diseases, such as Alzheimer's disease (AD). In fact, although a growing number of studies show crosstalk between these two processes, there remain numerous gaps in our knowledge of the mechanisms involved, which requires further clarification. On the one hand, mitochondrial dysfunction may lead to the release of mitochondrial damage-associated molecular patterns (mtDAMPs) which are recognized by microglial immune receptors and contribute to neuroinflammation progression. On the other hand, inflammatory molecules released by glial cells can influence and regulate mitochondrial function. A deeper understanding of these mechanisms may help identify biomarkers and molecular targets useful for the treatment of neurodegenerative diseases. This review of works published in recent years is focused on the description of the mitochondrial contribution to neuroinflammation and neurodegeneration, with particular attention to mitochondrial DNA (mtDNA) and AD.
Collapse
Affiliation(s)
- Giacoma Galizzi
- Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Via Ugo La Malfa, 153-90146 Palermo, Italy;
| | | |
Collapse
|
12
|
Lyu Y, Wang T, Huang S, Zhang Z. Mitochondrial Damage-Associated Molecular Patterns and Metabolism in the Regulation of Innate Immunity. J Innate Immun 2023; 15:665-679. [PMID: 37666239 PMCID: PMC10601681 DOI: 10.1159/000533602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/10/2023] [Indexed: 09/06/2023] Open
Abstract
The innate immune system, as the host's first line of defense against intruders, plays a critical role in recognizing, identifying, and reacting to a wide range of microbial intruders. There is increasing evidence that mitochondrial stress is a major initiator of innate immune responses. When mitochondria's integrity is disrupted or dysfunction occurs, the mitochondria's contents are released into the cytosol. These contents, like reactive oxygen species, mitochondrial DNA, and double-stranded RNA, among others, act as damage-related molecular patterns (DAMPs) that can bind to multiple innate immune sensors, particularly pattern recognition receptors, thereby leading to inflammation. To avoid the production of DAMPs, in addition to safeguarding organelles integrity and functionality, mitochondria may activate mitophagy or apoptosis. Moreover, mitochondrial components and specific metabolic regulations modify properties of innate immune cells. These include macrophages, dendritic cells, innate lymphoid cells, and so on, in steady state or in stimulation that are involved in processes ranging from the tricarboxylic acid cycle to oxidative phosphorylation and fatty acid metabolism. Here we provide a brief summary of mitochondrial DAMPs' initiated and potentiated inflammatory response in the innate immune system. We also provide insights into how the state of activation, differentiation, and functional polarization of innate immune cells can be influenced by alteration to the metabolic pathways in mitochondria.
Collapse
Affiliation(s)
- Yanmin Lyu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Tianyu Wang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuhong Huang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhaoqiang Zhang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
13
|
Huang W, Wen L, Tian H, Jiang J, Liu M, Ye Y, Gao J, Zhang R, Wang F, Li H, Shen L, Peng F, Tu Y. Self-Propelled Proteomotors with Active Cell-Free mtDNA Clearance for Enhanced Therapy of Sepsis-Associated Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301635. [PMID: 37518854 PMCID: PMC10520684 DOI: 10.1002/advs.202301635] [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: 03/13/2023] [Revised: 07/10/2023] [Indexed: 08/01/2023]
Abstract
Acute lung injury (ALI) is a frequent and serious complication of sepsis with limited therapeutic options. Gaining insights into the inflammatory dysregulation that causes sepsis-associated ALI can help develop new therapeutic strategies. Herein, the crucial role of cell-free mitochondrial DNA (cf-mtDNA) in the regulation of alveolar macrophage activation during sepsis-associated ALI is identified. Most importantly, a biocompatible hybrid protein nanomotor (NM) composed of recombinant deoxyribonuclease I (DNase-I) and human serum albumin (HSA) via glutaraldehyde-mediated crosslinking is prepared to obtain an inhalable nanotherapeutic platform targeting pulmonary cf-mtDNA clearance. The synthesized DNase-I/HSA NMs are endowed with self-propulsive capability and demonstrate superior performances in stability, DNA hydrolysis, and biosafety. Pulmonary delivery of DNase-I/HSA NMs effectively eliminates cf-mtDNAs in the lungs, and also improves sepsis survival by attenuating pulmonary inflammation and lung injury. Therefore, pulmonary cf-mtDNA clearance strategy using DNase-I/HSA NMs is considered to be an attractive approach for sepsis-associated ALI.
Collapse
Affiliation(s)
- Weichang Huang
- Department of Critical Care MedicineDongguan Institute of Respiratory and Critical Care MedicineAffiliated Dongguan HospitalSouthern Medical UniversityDongguan523059China
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Lihong Wen
- Department of Plastic SurgerySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhou510120China
| | - Hao Tian
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jiamiao Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Meihuan Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Yicheng Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Junbin Gao
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Ruotian Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Fei Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Huaan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Lihan Shen
- Department of Critical Care MedicineDongguan Institute of Respiratory and Critical Care MedicineAffiliated Dongguan HospitalSouthern Medical UniversityDongguan523059China
| | - Fei Peng
- School of Materials Science and EngineeringSun Yat‐Sen UniversityGuangzhou510275China
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
14
|
Main EN, Cruz TM, Bowlin GL. Mitochondria as a therapeutic: a potential new frontier in driving the shift from tissue repair to regeneration. Regen Biomater 2023; 10:rbad070. [PMID: 37663015 PMCID: PMC10468651 DOI: 10.1093/rb/rbad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/12/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
Fibrosis, or scar tissue development, is associated with numerous pathologies and is often considered a worst-case scenario in terms of wound healing or the implantation of a biomaterial. All that remains is a disorganized, densely packed and poorly vascularized bundle of connective tissue, which was once functional tissue. This creates a significant obstacle to the restoration of tissue function or integration with any biomaterial. Therefore, it is of paramount importance in tissue engineering and regenerative medicine to emphasize regeneration, the successful recovery of native tissue function, as opposed to repair, the replacement of the native tissue (often with scar tissue). A technique dubbed 'mitochondrial transplantation' is a burgeoning field of research that shows promise in in vitro, in vivo and various clinical applications in preventing cell death, reducing inflammation, restoring cell metabolism and proper oxidative balance, among other reported benefits. However, there is currently a lack of research regarding the potential for mitochondrial therapies within tissue engineering and regenerative biomaterials. Thus, this review explores these promising findings and outlines the potential for mitochondrial transplantation-based therapies as a new frontier of scientific research with respect to driving regeneration in wound healing and host-biomaterial interactions, the current successes of mitochondrial transplantation that warrant this potential and the critical questions and remaining obstacles that remain in the field.
Collapse
Affiliation(s)
- Evan N Main
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| | - Thaiz M Cruz
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| | - Gary L Bowlin
- Department of Biomedical Engineering, University of Memphis, 330 Engineering Technology Building, Memphis, TN 38152, USA
| |
Collapse
|
15
|
Ma W, Zhu J, Bai L, Zhao P, Li F, Zhang S. The role of neutrophil extracellular traps and proinflammatory damage-associated molecular patterns in idiopathic inflammatory myopathies. Clin Exp Immunol 2023; 213:202-208. [PMID: 37289984 PMCID: PMC10361739 DOI: 10.1093/cei/uxad059] [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: 02/11/2023] [Revised: 04/13/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of systemic autoimmune diseases characterized by immune-mediated muscle injury. Abnormal neutrophil extracellular traps (NETs) can be used as a biomarker of IIM disease activity, but the mechanism of NET involvement in IIMs needs to be elucidated. Important components of NETs, including high-mobility group box 1, DNA, histones, extracellular matrix, serum amyloid A, and S100A8/A9, act as damage-associated molecular patterns (DAMPs) to promote inflammation in IIMs. NETs can act on different cells to release large amounts of cytokines and activate the inflammasome, which can subsequently aggravate the inflammatory response. Based on the idea that NETs may be proinflammatory DAMPs of IIMs, we describe the role of NETs, DAMPs, and their interaction in the pathogenesis of IIMs and discuss the possible targeted treatment strategies in IIMs.
Collapse
Affiliation(s)
- Wenlan Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiarui Zhu
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Bai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Peipei Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Feifei Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
16
|
Al Amir Dache Z, Thierry AR. Mitochondria-derived cell-to-cell communication. Cell Rep 2023; 42:112728. [PMID: 37440408 DOI: 10.1016/j.celrep.2023.112728] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 02/21/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
In addition to their intracellular mobility, mitochondria and their components can exist outside the cells from which they originate. As a result, they are capable of acting on non-parental distant cells and mediate intercellular communication in physiological conditions and in a variety of pathologies. It has recently been demonstrated that this horizontal transfer governs a wide range of biological processes, such as tissue homeostasis, the rescue of injured recipient cells, and tumorigenesis. In addition, due to mitochondria's bacterial ancestry, they and their components can be recognized as damage-associated molecular patterns (DAMPs) by the immune cells, leading to inflammation. Here, we provide an overview of the most current and significant findings concerning the different structures of extracellular mitochondria and their by-products and their functions in the physiological and pathological context. This account illustrates the ongoing expansion of our understanding of mitochondria's biological role and functions in mammalian organisms.
Collapse
Affiliation(s)
- Zahra Al Amir Dache
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France; INSERM U1316, CNRS UMR7057, Université Paris Cité, Paris, France
| | - Alain R Thierry
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France; ICM, Institut Régional du Cancer de Montpellier, 34298 Montpellier, France.
| |
Collapse
|
17
|
Ju WK, Perkins GA, Kim KY, Bastola T, Choi WY, Choi SH. Glaucomatous optic neuropathy: Mitochondrial dynamics, dysfunction and protection in retinal ganglion cells. Prog Retin Eye Res 2023; 95:101136. [PMID: 36400670 DOI: 10.1016/j.preteyeres.2022.101136] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/04/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by a slow, progressive, and multifactorial degeneration of retinal ganglion cells (RGCs) and their axons, resulting in vision loss. Despite its high prevalence in individuals 60 years of age and older, the causing factors contributing to glaucoma progression are currently not well characterized. Intraocular pressure (IOP) is the only proven treatable risk factor. However, lowering IOP is insufficient for preventing disease progression. One of the significant interests in glaucoma pathogenesis is understanding the structural and functional impairment of mitochondria in RGCs and their axons and synapses. Glaucomatous risk factors such as IOP elevation, aging, genetic variation, neuroinflammation, neurotrophic factor deprivation, and vascular dysregulation, are potential inducers for mitochondrial dysfunction in glaucoma. Because oxidative phosphorylation stress-mediated mitochondrial dysfunction is associated with structural and functional impairment of mitochondria in glaucomatous RGCs, understanding the underlying mechanisms and relationship between structural and functional alterations in mitochondria would be beneficial to developing mitochondria-related neuroprotection in RGCs and their axons and synapses against glaucomatous neurodegeneration. Here, we review the current studies focusing on mitochondrial dynamics-based structural and functional alterations in the mitochondria of glaucomatous RGCs and therapeutic strategies to protect RGCs against glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Won-Kyu Ju
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tonking Bastola
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA
| | - Woo-Young Choi
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA; Department of Plastic Surgery, College of Medicine, Chosun University, Gwang-ju, South Korea
| | - Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
18
|
Huang H, Oo TT, Apaijai N, Chattipakorn N, Chattipakorn SC. An Updated Review of Mitochondrial Transplantation as a Potential Therapeutic Strategy Against Cerebral Ischemia and Cerebral Ischemia/Reperfusion Injury. Mol Neurobiol 2023; 60:1865-1883. [PMID: 36595193 DOI: 10.1007/s12035-022-03200-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023]
Abstract
Regardless of the progress made in the pathogenesis of ischemic stroke, it remains a leading cause of adult disability and death. To date, the most effective treatment for ischemic stroke is the timely recanalization of the occluded artery. However, the short time window and reperfusion injury have greatly limited its application and efficacy. Mitochondrial dysfunction and ATP depletion have become regarded as being hallmarks of neuropathophysiology following ischemic stroke. Mitochondrial transplantation is a novel potential therapeutic intervention for ischemic stroke that has sparked widespread concern during the past few years. This review summarizes and discusses the effects of mitochondrial transplantation in in vitro and in vivo ischemic stroke models. In addition, pharmacological interventions promoting mitochondrial transplantation are reviewed and discussed. We also discuss the potential challenges to the clinical application of mitochondrial transplantation in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Huatuo Huang
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Thura Tun Oo
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, 50200, Chiang Mai, Thailand. .,Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai, 50200, Thailand. .,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
19
|
Kalani K, Chaturvedi P, Chaturvedi P, Kumar Verma V, Lal N, Awasthi SK, Kalani A. Mitochondrial mechanisms in Alzheimer's disease: Quest for therapeutics. Drug Discov Today 2023; 28:103547. [PMID: 36871845 DOI: 10.1016/j.drudis.2023.103547] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/05/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Mitochondrial function is essential for maintaining neuronal integrity, because neurons have a high energy demand. Neurodegenerative diseases, such as Alzheimer's disease (AD), are exacerbated by mitochondrial dysfunction. Mitochondrial autophagy (mitophagy) attenuates neurodegenerative diseases by eradicating dysfunctional mitochondria. In neurodegenerative disorders, there is disruption of the mitophagy process. High levels of iron also interfere with the mitophagy process and the mtDNA released after mitophagy is proinflammatory and triggers the cGAS-STING pathway that aids AD pathology. In this review, we critically discuss the factors that affect mitochondrial impairment and different mitophagy processes in AD. Furthermore, we discuss the molecules used in mouse studies as well as clinical trials that could result in potential therapeutics in the future.
Collapse
Affiliation(s)
- Komal Kalani
- Department of Chemistry, The University of Texas at San Antonio, San Antonio 78249, TX, USA; Regulatory Scientist, Vestaron Cooperation, Durham 27703, NC, USA
| | - Poonam Chaturvedi
- Department of Physiotherapy, Lovely Professional University, Phagwara 144402, Punjab, India
| | - Pankaj Chaturvedi
- Department of Physiology, University of Louisville, Louisville 40202, KY, USA
| | - Vinod Kumar Verma
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur 208024, Uttar Pradesh, India
| | - Nand Lal
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur 208024, Uttar Pradesh, India
| | - Sudhir K Awasthi
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur 208024, Uttar Pradesh, India
| | - Anuradha Kalani
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur 208024, Uttar Pradesh, India.
| |
Collapse
|
20
|
Fischer I, Chaudhry SR, Hänggi D, Muhammad S. Clustering of serum biomarkers involved in post-aneurysmal subarachnoid hemorrhage (aSAH) complications. Neurosurg Rev 2023; 46:63. [PMID: 36864213 PMCID: PMC9981718 DOI: 10.1007/s10143-023-01967-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/30/2023] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
Post-aSAH sequela is characterized by the onset of life-threatening complications along with the upregulated underlying inflammation. Cerebral vasospasm (CVS) is one of the most frequent complication after aSAH, which contributes majorly to delayed cerebral ischemia and poor clinical outcome. The objective of this study was to identify the clusters of serum biomarkers that are associated with cerebral vasospasm (CVS) after suffering from aneurysmal subarachnoid hemorrhage (aSAH). In this single-center study, serum concentrations of 10 potential biomarkers, together with clinical and demographic parameters, for 66 aSAH patients were recorded within 24 h after aSAH. The dataset was split into a training set (43 patients) and a validation set. Correlation heatmaps for both datasets were computed. Variables with inconsistent correlations on the two subsets were excluded. Clusters of relevant biomarkers were identified on the complete set, separately for patients who developed post-aSAH CVS and those who did not. Two clusters were found to be specific for patients who suffered from CVS: mitochondrial gene fragments (cytochrome B (Cyt-B), cytochrome C oxidase subunit-1 (Cox-1), displacement loop (D-loop), and IL-23, and the other one, containing IL-6, IL-10, age, and Hunt and Hess score. Clusters of serum biomarkers, analyzed within 24 h of the onset of aSAH, days before the CVS development, are expressed differently in patients suffering from post-aSAH CVS, compared to patients without CVS. This suggests that these biomarkers may be involved in the pathophysiological processes leading to CVS and may be used as its early predictors. These interesting findings are potentially highly relevant for the management of CVS and call for validation on a larger sample of patients.
Collapse
Affiliation(s)
- Igor Fischer
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Shafqat Rasul Chaudhry
- Department of Pharmacy, Obaid Noor Institute of Medical Sciences (ONIMS), Mianwali, 42200, Punjab, Pakistan
| | - Daniel Hänggi
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
21
|
Karmanova E, Chernikov A, Usacheva A, Ivanov V, Bruskov V. Metformin counters oxidative stress and mitigates adverse effects of radiation exposure: An overview. Fundam Clin Pharmacol 2023. [PMID: 36852652 DOI: 10.1111/fcp.12884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/19/2023] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
Metformin (1,1-dimethylbiguanidine hydrochloride) (MF) is a drug that has long been in use for the treatment of type 2 diabetes mellitus and recently is coming into use in the radiation therapy of cancer and other conditions. Exposure to ionizing radiation disturbs the redox homeostasis of cells and causes damage to proteins, membranes, and mitochondria, destroying a number of biological processes. After irradiation, MF activates cellular antioxidant and repair systems by signaling to eliminate the harmful consequences of disruption of redox homeostasis. The use of MF in the treatment of the negative effects of irradiation has great potential in medical patients after radiotherapy and in victims of nuclear accidents or radiologic terrorism.
Collapse
Affiliation(s)
- Ekaterina Karmanova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.,Institute of Cell Biophysics, Pushchino Scientific Center for Biological Research, Federal Research Center of the Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Anatoly Chernikov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Anna Usacheva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vladimir Ivanov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vadim Bruskov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
22
|
Liu Z, Shan S, Yuan Z, Wu F, Zheng M, Wang Y, Gui J, Xu W, Wang C, Ren T, Wen Z. Mitophagy bridges DNA sensing with metabolic adaption to expand lung cancer stem-like cells. EMBO Rep 2023; 24:e54006. [PMID: 36416244 PMCID: PMC9900345 DOI: 10.15252/embr.202154006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/01/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
While previous studies have identified cancer stem-like cells (CSCs) as a crucial driver for chemoresistance and tumor recurrence, the underlying mechanisms for populating the CSC pool remain unclear. Here, we identify hypermitophagy as a feature of human lung CSCs, promoting metabolic adaption via the Notch1-AMPK axis to drive CSC expansion. Specifically, mitophagy is highly active in CSCs, resulting in increased mitochondrial DNA (mtDNA) content in the lysosome. Lysosomal mtDNA acts as an endogenous ligand for Toll-like receptor 9 (TLR9) that promotes Notch1 activity. Notch1 interacts with AMPK to drive lysosomal AMPK activation by inducing metabolic stress and LKB1 phosphorylation. This TLR9-Notch1-AMPK axis supports mitochondrial metabolism to fuel CSC expansion. In patient-derived xenograft chimeras, targeting mitophagy and TLR9-dependent Notch1-AMPK pathway restricts tumor growth and CSC expansion. Taken together, mitochondrial hemostasis is interlinked with innate immune sensing and Notch1-AMPK activity to increase the CSC pool of human lung cancer.
Collapse
Affiliation(s)
- Zhen Liu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Shan Shan
- Department of Respiratory MedicineShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zixin Yuan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Ming Zheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Ying Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Jun Gui
- State Key Laboratory of Oncogenes and Related Genes; Renji‐Med X Clinical Stem Cell Research Center, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Xu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Chunhong Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology of Jiangsu Province, State Key Laboratory of Radiation Medicine and ProtectionSoochow UniversitySuzhouChina
| | - Tao Ren
- Department of Respiratory MedicineShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Kay Laboratory of Sleep Disordered BreathingShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| |
Collapse
|
23
|
Li Y, Yang S, Jin X, Li D, Lu J, Wang X, Wu M. Mitochondria as novel mediators linking gut microbiota to atherosclerosis that is ameliorated by herbal medicine: A review. Front Pharmacol 2023; 14:1082817. [PMID: 36733506 PMCID: PMC9886688 DOI: 10.3389/fphar.2023.1082817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Atherosclerosis (AS) is the main cause of cardiovascular disease (CVD) and is characterized by endothelial damage, lipid deposition, and chronic inflammation. Gut microbiota plays an important role in the occurrence and development of AS by regulating host metabolism and immunity. As human mitochondria evolved from primordial bacteria have homologous characteristics, they are attacked by microbial pathogens as target organelles, thus contributing to energy metabolism disorders, oxidative stress, and apoptosis. Therefore, mitochondria may be a key mediator of intestinal microbiota disorders and AS aggravation. Microbial metabolites, such as short-chain fatty acids, trimethylamine, hydrogen sulfide, and bile acids, also affect mitochondrial function, including mtDNA mutation, oxidative stress, and mitophagy, promoting low-grade inflammation. This further damages cellular homeostasis and the balance of innate immunity, aggravating AS. Herbal medicines and their monomers can effectively ameliorate the intestinal flora and their metabolites, improve mitochondrial function, and inhibit atherosclerotic plaques. This review focuses on the interaction between gut microbiota and mitochondria in AS and explores a therapeutic strategy for restoring mitochondrial function and intestinal microbiota disorders using herbal medicines, aiming to provide new insights for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yujuan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Jin
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Beijing University of Chinese Medicine, Beijing, China
| | - Xinyue Wang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Min Wu,
| |
Collapse
|
24
|
Mitrofanova A, Fontanella AM, Burke GW, Merscher S, Fornoni A. Mitochondrial Contribution to Inflammation in Diabetic Kidney Disease. Cells 2022; 11:3635. [PMID: 36429063 PMCID: PMC9688941 DOI: 10.3390/cells11223635] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes is the leading cause of chronic kidney disease worldwide. Despite the burden, the factors contributing to the development and progression of diabetic kidney disease (DKD) remain to be fully elucidated. In recent years, increasing evidence suggests that mitochondrial dysfunction is a pathological mediator in DKD as the kidney is a highly metabolic organ rich in mitochondria. Furthermore, low grade chronic inflammation also contributes to the progression of DKD, and several inflammatory biomarkers have been reported as prognostic markers to risk-stratify patients for disease progression and all-cause mortality. Interestingly, the term "sterile inflammation" appears to be used in the context of DKD describing the development of intracellular inflammation in the absence of bacterial or viral pathogens. Therefore, a link between mitochondrial dysfunction and inflammation in DKD exists and is a hot topic in both basic research and clinical investigations. This review summarizes how mitochondria contribute to sterile inflammation in renal cells in DKD.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Antonio M. Fontanella
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - George W. Burke
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
25
|
Estrogen receptor β activation inhibits colitis by promoting NLRP6-mediated autophagy. Cell Rep 2022; 41:111454. [DOI: 10.1016/j.celrep.2022.111454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/24/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022] Open
|
26
|
Long G, Gong R, Wang Q, Zhang D, Huang C. Role of released mitochondrial DNA in acute lung injury. Front Immunol 2022; 13:973089. [PMID: 36059472 PMCID: PMC9433898 DOI: 10.3389/fimmu.2022.973089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/01/2022] [Indexed: 12/02/2022] Open
Abstract
Acute lung injury(ALI)/acute respiratory distress syndrome(ARDS) is a form of acute-onset hypoxemic respiratory failure characterised by an acute, diffuse, inflammatory lung injury, and increased alveolar-capillary permeability, which is caused by a variety of pulmonary or nonpulmonary insults. Recently, aberrant mitochondria and mitochondrial DNA(mtDNA) level are associated with the development of ALI/ARDS, and plasma mtDNA level shows the potential to be a promising biomarker for clinical diagnosis and evaluation of lung injury severity. In mechanism, the mtDNA and its oxidised form, which are released from impaired mitochondria, play a crucial role in the inflammatory response and histopathological changes in the lung. In this review, we discuss mitochondrial outer membrane permeabilisation (MOMP), mitochondrial permeability transition pore(mPTP), extracellular vesicles (EVs), extracellular traps (ETs), and passive release as the principal mechanisms for the release of mitochondrial DNA into the cytoplasm and extracellular compartments respectively. Further, we explain how the released mtDNA and its oxidised form can induce inflammatory cytokine production and aggravate lung injury through the Toll-like receptor 9(TLR9) signalling, cytosolic cGAS-stimulator of interferon genes (STING) signalling (cGAS-STING) pathway, and inflammasomes activation. Additionally, we propose targeting mtDNA-mediated inflammatory pathways as a novel therapeutic approach for treating ALI/ARDS.
Collapse
Affiliation(s)
- Gangyu Long
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Rui Gong
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qian Wang
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Dingyu Zhang
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, China
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Dingyu Zhang, ; Chaolin Huang,
| | - Chaolin Huang
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, China
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Dingyu Zhang, ; Chaolin Huang,
| |
Collapse
|
27
|
Nofi CP, Wang P, Aziz M. Chromatin-Associated Molecular Patterns (CAMPs) in sepsis. Cell Death Dis 2022; 13:700. [PMID: 35961978 PMCID: PMC9372964 DOI: 10.1038/s41419-022-05155-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 01/21/2023]
Abstract
Several molecular patterns have been identified that recognize pattern recognition receptors. Pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) are commonly used terminologies to classify molecules originating from pathogen and endogenous molecules, respectively, to heighten the immune response in sepsis. Herein, we focus on a subgroup of endogenous molecules that may be detected as foreign and similarly trigger immune signaling pathways. These chromatin-associated molecules, i.e., chromatin containing nuclear DNA and histones, extracellular RNA, mitochondrial DNA, telomeric repeat-containing RNA, DNA- or RNA-binding proteins, and extracellular traps, may be newly classified as chromatin-associated molecular patterns (CAMPs). Herein, we review the release of CAMPs from cells, their mechanism of action and downstream immune signaling pathways, and targeted therapeutic approaches to mitigate inflammation and tissue injury in inflammation and sepsis.
Collapse
Affiliation(s)
- Colleen P. Nofi
- grid.250903.d0000 0000 9566 0634Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY USA ,Elmezi Graduate School of Molecular Medicine, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA
| | - Ping Wang
- grid.250903.d0000 0000 9566 0634Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY USA ,Elmezi Graduate School of Molecular Medicine, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA
| | - Monowar Aziz
- grid.250903.d0000 0000 9566 0634Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY USA ,Elmezi Graduate School of Molecular Medicine, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA
| |
Collapse
|
28
|
Fathima N, Manorenj S, Vishwakarma SK, Khan AA. Cell-free mitochondrial DNA quantification in ischemic stroke patients for non-invasive and real-time monitoring of disease status. World J Transl Med 2022; 10:14-28. [DOI: 10.5528/wjtm.v10.i2.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/14/2022] [Accepted: 07/17/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) is one of the major causes of the continuous increasing rate of global mortality due to the lack of timely diagnosis, prognosis, and management. This study provides a primitive platform for non-invasive and cost-effective diagnosis and prognosis of patients with AIS using circulating cell-free mitochondrial DNA (cf-mtDNA) quantification and validation.
AIM To evaluate the role of cf-mtDNA as s non-invasive, and affordable tool for real-time monitoring and prognosticating AIS patients at disease onset and during treatment.
METHODS This study enrolled 88 participants including 44 patients with AIS and 44 healthy controls with almost similar mean age group at stroke onset, and at 24 h and 72 h of treatment. Peripheral blood samples were collected from each study participant and plasma was separated using centrifugation. The cf-mtDNA concentration was quantified using nanodrop reading and validated through real-time quantitative polymerase chain reaction (RT-qPCR) of NADH-ubiquinone oxidoreductase chain 1 (ND1) relative transcript expression levels.
RESULTS Comparative analysis of cf-mtDNA concentration in patients at disease onset showed significantly increased levels compared to control individuals for both nanodrop reading, as well as ND1 relative expression levels (P < 0.0001). Intergroup analysis of cf-mtDNA concentration using nanodrop showed significantly reduced levels in patients at 72 h of treatment compared to onset (P < 0.01). However, RT-qPCR analysis showed a significant reduction at 24 h and 72 h of treatment compared to the disease onset (P < 0.001). The sensitivity and specificity were relatively higher for RT-qPCR than nanodrop-based cf-mtDNA quantification. Correlation analysis of both cf-mtDNA concentration as well as ND1 relative expression with National Institute of Health Stroke Scale score at baseline showed a positive trend.
CONCLUSION In summary, quantitative estimation of highly pure cf-mtDNA provides a simple, highly sensitive and specific, non-invasive, and affordable approach for real-time monitoring and prognosticating AIS patients at onset and during treatment.
Collapse
Affiliation(s)
- Nusrath Fathima
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Sandhya Manorenj
- Department of Neurology, Princess Esra Hospital, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Sandeep Kumar Vishwakarma
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| | - Aleem Ahmed Khan
- Central Laboratory for Stem Cell Research and Translational Medicine, Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Hyderabad 500058, Telangana, India
| |
Collapse
|
29
|
Liu C, Yang M, Li L, Luo S, Yang J, Li C, Liu H, Sun L. A Glimpse of Inflammation and Anti-Inflammation Therapy in Diabetic Kidney Disease. Front Physiol 2022; 13:909569. [PMID: 35874522 PMCID: PMC9298824 DOI: 10.3389/fphys.2022.909569] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/18/2022] [Indexed: 02/06/2023] Open
Abstract
Diabetic kidney disease (DKD) is a common complication of diabetes mellitus and a major cause of end-stage kidney disease (ESKD). The pathogenesis of DKD is very complex and not completely understood. Recently, accumulated evidence from in vitro and in vivo studies has demonstrated that inflammation plays an important role in the pathogenesis and the development of DKD. It has been well known that a variety of pro-inflammatory cytokines and related signaling pathways are involved in the procession of DKD. Additionally, some anti-hyperglycemic agents and mineralocorticoid receptor antagonists (MRAs) that are effective in alleviating the progression of DKD have anti-inflammatory properties, which might have beneficial effects on delaying the progression of DKD. However, there is currently a lack of systematic overviews. In this review, we focus on the novel pro-inflammatory signaling pathways in the development of DKD, including the nuclear factor kappa B (NF-κB) signaling pathway, toll-like receptors (TLRs) and myeloid differentiation primary response 88 (TLRs/MyD88) signaling pathway, adenosine 5′-monophosphate-activated protein kinase (AMPK) signaling pathways, inflammasome activation, mitochondrial DNA (mtDNA) release as well as hypoxia-inducible factor-1(HIF-1) signaling pathway. We also discuss the related anti-inflammation mechanisms of metformin, finerenone, sodium-dependent glucose transporters 2 (SGLT2) inhibitors, Dipeptidyl peptidase-4 (DPP-4) inhibitors, Glucagon-like peptide-1 (GLP-1) receptor agonist and traditional Chinese medicines (TCM).
Collapse
Affiliation(s)
- Chongbin Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South Unibersity, Changsha, China.,Hunan Key Laboratory of kidney Disease and Blood Purification, Changsha, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South Unibersity, Changsha, China.,Hunan Key Laboratory of kidney Disease and Blood Purification, Changsha, China
| | - Li Li
- Department of Nephrology, The Second Xiangya Hospital, Central South Unibersity, Changsha, China.,Hunan Key Laboratory of kidney Disease and Blood Purification, Changsha, China
| | - Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital, Central South Unibersity, Changsha, China
| | - Jinfei Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South Unibersity, Changsha, China
| | - Chenrui Li
- Department of Nephrology, The Second Xiangya Hospital, Central South Unibersity, Changsha, China
| | - Huafeng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases & Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South Unibersity, Changsha, China.,Hunan Key Laboratory of kidney Disease and Blood Purification, Changsha, China
| |
Collapse
|
30
|
Farooq M, Khan AW, Ahmad B, Kim MS, Choi S. Therapeutic Targeting of Innate Immune Receptors Against SARS-CoV-2 Infection. Front Pharmacol 2022; 13:915565. [PMID: 35847031 PMCID: PMC9280161 DOI: 10.3389/fphar.2022.915565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
The innate immune system is the first line of host's defense against invading pathogens. Multiple cellular sensors that detect viral components can induce innate antiviral immune responses. As a result, interferons and pro-inflammatory cytokines are produced which help in the elimination of invading viruses. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) belongs to Coronaviridae family, and has a single-stranded, positive-sense RNA genome. It can infect multiple hosts; in humans, it is responsible for the novel coronavirus disease 2019 (COVID-19). Successful, timely, and appropriate detection of SARS-CoV-2 can be very important for the early generation of the immune response. Several drugs that target the innate immune receptors as well as other signaling molecules generated during the innate immune response are currently being investigated in clinical trials. In this review, we summarized the current knowledge of the mechanisms underlying host sensing and innate immune responses against SARS-CoV-2 infection, as well as the role of innate immune receptors in terms of their therapeutic potential against SARS-CoV-2. Moreover, we discussed the drugs undergoing clinical trials and the FDA approved drugs against SARS-CoV-2. This review will help in understanding the interactions between SARS-CoV-2 and innate immune receptors and thus will point towards new dimensions for the development of new therapeutics, which can be beneficial in the current pandemic.
Collapse
Affiliation(s)
- Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- S&K Therapeutics, Ajou University, Suwon, South Korea
| | - Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Bilal Ahmad
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- S&K Therapeutics, Ajou University, Suwon, South Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- S&K Therapeutics, Ajou University, Suwon, South Korea
| |
Collapse
|
31
|
Guo Y, Gan D, Hu F, Cheng Y, Yu J, Lei B, Shu Q, Gu R, Xu G. Intravitreal injection of mitochondrial DNA induces cell damage and retinal dysfunction in rats. Biol Res 2022; 55:22. [PMID: 35659309 PMCID: PMC9164539 DOI: 10.1186/s40659-022-00390-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/03/2022] [Indexed: 11/28/2022] Open
Abstract
Background Retinal neurodegeneration is induced by a variety of environmental insults and stresses, but the exact mechanisms are unclear. In the present study, we explored the involvement of cytosolic mitochondrial DNA (mtDNA), resulting in the cGAS-STING dependent inflammatory response and apoptosis in retinal damage in vivo. Methods Retinal injury was induced with white light or intravitreal injection of lipopolysaccharide (LPS). After light- or LPS-induced injury, the amount of cytosolic mtDNA in the retina was detected by PCR. The mtDNA was isolated and used to transfect retinas in vivo. WB and real-time PCR were used to evaluate the activation of cGAS-STING pathway and the levels of apoptosis-associated protein at different times after mtDNA injection. Retinal cell apoptosis rate was detected by TUNEL staining. Full-field electroretinography (ERG) was used to assess the retinal function. Results Light injury and the intravitreal injection of LPS both caused the leakage of mtDNA into the cytoplasm in retinal tissue. After the transfection of mtDNA in vivo, the levels of cGAS, STING, and IFN-β mRNAs and the protein levels of STING, phosph-TBK1, phospho-IRF3, and IFN-β were upregulated. mtDNA injection also induced the activation of caspase 3 and caspase 9. BAX and BAK were increased at both the mRNA and protein levels. The release of cytochrome c from the mitochondria to the cytosol was increased after mtDNA injection. The wave amplitudes on ERG decreased and retinal cell apoptosis was detected after mtDNA injection. Conclusions Cytosolic mtDNA triggers an inflammatory response. It also promotes apoptosis and the dysfunction of the retina. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-022-00390-6.
Collapse
|
32
|
Wu W, Zhou S, Liu T, Liang D. Mitochondrial transcription factor B2 overexpression increases M2 macrophage infiltration via cytosolic mitochondrial DNA-stimulated Interleukin-6 secretion in ovarian cancer. Bioengineered 2022; 13:12211-12223. [PMID: 35577351 PMCID: PMC9275939 DOI: 10.1080/21655979.2022.2074615] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mitochondrial transcription factor B2 (TFB2M) is a protein modulating both mitochondrial DNA (mtDNA) transcription and compacting. In this study, we explored the expression profile of TFB2M in ovarian cancer, its association with infiltration of tumor-associated macrophages (TAMs), and its influence on macrophage polarization. Serial sections of ovarian cancer tissue arrays were stained to detect TFB2M and CD163 expression. Epithelial ovarian cancer cell line OVISE and CAOV4 were used to assess the influence of TFB2M on IL-6 expression. THP-1 cells were utilized as an in vitro model for macrophage migration and polarization. Results showed that higher TFB2M expression is associated with poor survival in ovarian cancer patients. IHC staining confirmed a moderately positive correlation between TFB2M expression and the infiltration of CD163-positive cells in 68 primary ovarian cancer cases. TFB2M overexpression was associated with increased mtDNA outside the mitochondria and elevated IL-6 expression in ovarian cancer cells. When cytosolic mtDNA was selectively inhibited by DNase I, TFB2M-induced IL-6 upregulation was canceled. TFB2M overexpression could activate the nuclear factor kappa-B (NF-κB) signaling pathway via promoting nucleus entry of p65 and p-p65, which was abrogated by inhibiting cytosolic mtDNA, TLR9, or NF-κB signaling pathway. Conditioned medium from OIVSE cells with TFB2M overexpression could induce macrophage migration and M2 polarization. However, these inducing effects were abrogated by DNase I, TLR9 inhibitor, and anti-IL-6 R pretreatment. In conclusion, this study showed a novel role of TFB2M in the immunosuppressive tumor microenvironment. It promotes M2 macrophage infiltration via a cytosolic mtDNA/TLR9/NF-κB/IL-6 pathway in ovarian cancer.
Collapse
Affiliation(s)
- Weilu Wu
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Shijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tianmin Liu
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Dongni Liang
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
33
|
Xie B, Luo A. Nucleic Acid Sensing Pathways in DNA Repair Targeted Cancer Therapy. Front Cell Dev Biol 2022; 10:903781. [PMID: 35557952 PMCID: PMC9089908 DOI: 10.3389/fcell.2022.903781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022] Open
Abstract
The repair of DNA damage is a complex process, which helps to maintain genome fidelity, and the ability of cancer cells to repair therapeutically DNA damage induced by clinical treatments will affect the therapeutic efficacy. In the past decade, great success has been achieved by targeting the DNA repair network in tumors. Recent studies suggest that DNA damage impacts cellular innate and adaptive immune responses through nucleic acid-sensing pathways, which play essential roles in the efficacy of DNA repair targeted therapy. In this review, we summarize the current understanding of the molecular mechanism of innate immune response triggered by DNA damage through nucleic acid-sensing pathways, including DNA sensing via the cyclic GMP-AMP synthase (cGAS), Toll-like receptor 9 (TLR9), absent in melanoma 2 (AIM2), DNA-dependent protein kinase (DNA-PK), and Mre11-Rad50-Nbs1 complex (MRN) complex, and RNA sensing via the TLR3/7/8 and retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs). Furthermore, we will focus on the recent developments in the impacts of nucleic acid-sensing pathways on the DNA damage response (DDR). Elucidating the DDR-immune response interplay will be critical to harness immunomodulatory effects to improve the efficacy of antitumor immunity therapeutic strategies and build future therapeutic approaches.
Collapse
Affiliation(s)
- Bingteng Xie
- School of Life Science, Beijing Institute of Technology, Beijing, China.,Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment, Beijing Institute of Technology, Ministry of Industry and Information Technology, Beijing, China
| | - Aiqin Luo
- School of Life Science, Beijing Institute of Technology, Beijing, China.,Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment, Beijing Institute of Technology, Ministry of Industry and Information Technology, Beijing, China
| |
Collapse
|
34
|
Pastorek M, Dúbrava M, Celec P. On the Origin of Neutrophil Extracellular Traps in COVID-19. Front Immunol 2022; 13:821007. [PMID: 35359960 PMCID: PMC8961727 DOI: 10.3389/fimmu.2022.821007] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Despite ongoing vaccination COVID-19 is a global healthcare problem because of the lack of an effective targeted therapy. In severe COVID-19 manifesting as acute respiratory distress syndrome, uncontrolled innate immune system activation results in cytokine deregulation, damage-associated molecular patterns release upon tissue damage and high occurrence of thrombotic events. These pathomechanisms are linked to neutrophil function and dysfunction, particularly increased formation of neutrophil extracellular traps (NETs). While the association of NETs and severity of COVID-19 has been shown and proved, the causes of NETs formation are unclear. The aim of this review is to summarize potential inducers of NETs formation in severe COVID-19 and to discuss potential treatment options targeting NETs formation of removal.
Collapse
Affiliation(s)
- Michal Pastorek
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Martin Dúbrava
- Department of Geriatric Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| |
Collapse
|
35
|
Guo Y, Tsai HI, Zhang L, Zhu H. Mitochondrial DNA on Tumor-Associated Macrophages Polarization and Immunity. Cancers (Basel) 2022; 14:1452. [PMID: 35326602 PMCID: PMC8946090 DOI: 10.3390/cancers14061452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/26/2022] [Accepted: 03/09/2022] [Indexed: 12/04/2022] Open
Abstract
As the richest immune cells in most tumor microenvironments (TMEs), tumor-associated macrophages (TAMs) play an important role in tumor development and treatment sensitivity. The phenotypes and functions of TAMs vary according to their sources and tumor progression. Different TAM phenotypes display distinct behaviors in terms of tumor immunity and are regulated by intracellular and exogenous molecules. Additionally, dysfunctional and oxidatively stressed mitochondrial-derived mitochondrial DNA (mtDNA) plays an important role in remodeling the phenotypes and functions of TAMs. This article reviews the interactions between mtDNA and TAMs in the TME and further discusses the influence of their performance on tumor genesis and development.
Collapse
Affiliation(s)
- Yaxin Guo
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Hsiang-i Tsai
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Lirong Zhang
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Haitao Zhu
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
- Laboratory of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| |
Collapse
|
36
|
Lyamzaev KG, Zinovkin RA, Chernyak BV. Extrusion of mitochondria: Garbage clearance or cell–cell communication signals? J Cell Physiol 2022; 237:2345-2356. [DOI: 10.1002/jcp.30711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Konstantin G. Lyamzaev
- Belozersky Institute of Physico‐Chemical Biology Lomonosov Moscow State University Moscow Russia
- The “Russian Clinical Research Center for Gerontology” of the Ministry of Healthcare of the Russian Federation Pirogov Russian National Research Medical University Moscow Russia
| | - Roman A. Zinovkin
- Belozersky Institute of Physico‐Chemical Biology Lomonosov Moscow State University Moscow Russia
| | - Boris V. Chernyak
- Belozersky Institute of Physico‐Chemical Biology Lomonosov Moscow State University Moscow Russia
| |
Collapse
|
37
|
Kong C, Song W, Fu T. Systemic inflammatory response syndrome is triggered by mitochondrial damage (Review). Mol Med Rep 2022; 25:147. [PMID: 35234261 PMCID: PMC8915392 DOI: 10.3892/mmr.2022.12663] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/11/2022] [Indexed: 11/30/2022] Open
Abstract
Mitochondria are key organelles of cellular energy metabolism; both mitochondrial function and metabolism determine the physiological function of cells and serve an essential role in immune responses. Key damage-associated molecular patterns (DAMPs), such as mitochondrial DNA and N-formyl peptides, released following severe trauma-induced mitochondrial damage may affect the respiratory chain, enhance oxidative stress and activate systemic inflammatory responses via a variety of inflammation-associated signaling pathways. Severe trauma can lead to sepsis, multiple organ dysfunction syndrome and death. The present review aimed to summarize the pathophysiological mechanisms underlying the effects of human mitochondrial injury-released DAMPs on triggering systemic inflammatory responses and to determine their potential future clinical applications in preventing and treating sepsis.
Collapse
Affiliation(s)
- Can Kong
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Song
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tao Fu
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
38
|
Chowdhury A, Witte S, Aich A. Role of Mitochondrial Nucleic Acid Sensing Pathways in Health and Patho-Physiology. Front Cell Dev Biol 2022; 10:796066. [PMID: 35223833 PMCID: PMC8873532 DOI: 10.3389/fcell.2022.796066] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/14/2022] [Indexed: 12/23/2022] Open
Abstract
Mitochondria, in symbiosis with the host cell, carry out a wide variety of functions from generating energy, regulating the metabolic processes, cell death to inflammation. The most prominent function of mitochondria relies on the oxidative phosphorylation (OXPHOS) system. OXPHOS heavily influences the mitochondrial-nuclear communication through a plethora of interconnected signaling pathways. Additionally, owing to the bacterial ancestry, mitochondria also harbor a large number of Damage Associated Molecular Patterns (DAMPs). These molecules relay the information about the state of the mitochondrial health and dysfunction to the innate immune system. Consequently, depending on the intracellular or extracellular nature of detection, different inflammatory pathways are elicited. One group of DAMPs, the mitochondrial nucleic acids, hijack the antiviral DNA or RNA sensing mechanisms such as the cGAS/STING and RIG-1/MAVS pathways. A pro-inflammatory response is invoked by these signals predominantly through type I interferon (T1-IFN) cytokines. This affects a wide range of organ systems which exhibit clinical presentations of auto-immune disorders. Interestingly, tumor cells too, have devised ingenious ways to use the mitochondrial DNA mediated cGAS-STING-IRF3 response to promote neoplastic transformations and develop tumor micro-environments. Thus, mitochondrial nucleic acid-sensing pathways are fundamental in understanding the source and nature of disease initiation and development. Apart from the pathological interest, recent studies also attempt to delineate the structural considerations for the release of nucleic acids across the mitochondrial membranes. Hence, this review presents a comprehensive overview of the different aspects of mitochondrial nucleic acid-sensing. It attempts to summarize the nature of the molecular patterns involved, their release and recognition in the cytoplasm and signaling. Finally, a major emphasis is given to elaborate the resulting patho-physiologies.
Collapse
Affiliation(s)
- Arpita Chowdhury
- Department of Cellular Biochemistry, University Medical Center, Göttingen, Germany
| | - Steffen Witte
- Department of Cellular Biochemistry, University Medical Center, Göttingen, Germany
| | - Abhishek Aich
- Department of Cellular Biochemistry, University Medical Center, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging, from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
39
|
Austad SN, Ballinger S, Buford TW, Carter CS, Smith DL, Darley-Usmar V, Zhang J. Targeting whole body metabolism and mitochondrial bioenergetics in the drug development for Alzheimer's disease. Acta Pharm Sin B 2022; 12:511-531. [PMID: 35256932 PMCID: PMC8897048 DOI: 10.1016/j.apsb.2021.06.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/26/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Aging is by far the most prominent risk factor for Alzheimer's disease (AD), and both aging and AD are associated with apparent metabolic alterations. As developing effective therapeutic interventions to treat AD is clearly in urgent need, the impact of modulating whole-body and intracellular metabolism in preclinical models and in human patients, on disease pathogenesis, have been explored. There is also an increasing awareness of differential risk and potential targeting strategies related to biological sex, microbiome, and circadian regulation. As a major part of intracellular metabolism, mitochondrial bioenergetics, mitochondrial quality-control mechanisms, and mitochondria-linked inflammatory responses have been considered for AD therapeutic interventions. This review summarizes and highlights these efforts.
Collapse
Key Words
- ACE2, angiotensin I converting enzyme (peptidyl-dipeptidase A) 2
- AD, Alzheimer's disease
- ADP, adenosine diphosphate
- ADRD, AD-related dementias
- Aβ, amyloid β
- CSF, cerebrospinal fluid
- Circadian regulation
- DAMPs
- DAMPs, damage-associated molecular patterns
- Diabetes
- ER, estrogen receptor
- ETC, electron transport chain
- FCCP, trifluoromethoxy carbonylcyanide phenylhydrazone
- FPR-1, formyl peptide receptor 1
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide-1
- HBP, hexoamine biosynthesis pathway
- HTRA, high temperature requirement A
- Hexokinase biosynthesis pathway
- I3A, indole-3-carboxaldehyde
- IRF-3, interferon regulatory factor 3
- LC3, microtubule associated protein light chain 3
- LPS, lipopolysaccharide
- LRR, leucine-rich repeat
- MAVS, mitochondrial anti-viral signaling
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- MRS, magnetic resonance spectroscopy
- Mdivi-1, mitochondrial division inhibitor 1
- Microbiome
- Mitochondrial DNA
- Mitochondrial electron transport chain
- Mitochondrial quality control
- NLRP3, leucine-rich repeat (LRR)-containing protein (NLR)-like receptor family pyrin domain containing 3
- NOD, nucleotide-binding oligomerization domain
- NeuN, neuronal nuclear protein
- PET, fluorodeoxyglucose (FDG)-positron emission tomography
- PKA, protein kinase A
- POLβ, the base-excision repair enzyme DNA polymerase β
- ROS, reactive oxygen species
- Reactive species
- SAMP8, senescence-accelerated mice
- SCFAs, short-chain fatty acids
- SIRT3, NAD-dependent deacetylase sirtuin-3
- STING, stimulator of interferon genes
- STZ, streptozotocin
- SkQ1, plastoquinonyldecyltriphenylphosphonium
- T2D, type 2 diabetes
- TCA, Tricarboxylic acid
- TLR9, toll-like receptor 9
- TMAO, trimethylamine N-oxide
- TP, tricyclic pyrone
- TRF, time-restricted feeding
- cAMP, cyclic adenosine monophosphate
- cGAS, cyclic GMP/AMP synthase
- hAPP, human amyloid precursor protein
- hPREP, human presequence protease
- i.p., intraperitoneal
- mTOR, mechanistic target of rapamycin
- mtDNA, mitochondrial DNA
- αkG, alpha-ketoglutarate
Collapse
Affiliation(s)
- Steven N. Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Thomas W. Buford
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christy S. Carter
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
40
|
Wade H, Duan Q, Su Q. Interaction between Sars-CoV-2 structural proteins and host cellular receptors: From basic mechanisms to clinical perspectives. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:243-277. [PMID: 36088078 PMCID: PMC9182089 DOI: 10.1016/bs.apcsb.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (Sars-CoV-2) has caused a global pandemic that has affected the lives of billions of individuals. Sars-CoV-2 primarily infects human cells by binding of the viral spike protein to angiotensin-converting enzyme 2 (ACE2). In addition, novel means of viral entry are currently being investigated, including Neuropillin 1, toll-like receptors (TLRs), cluster of differentiation 147 (CD147), and integrin α5β1. Enriched expression of these proteins across metabolic regulatory organs/tissues, including the circulatory system, liver, pancreas, and intestine contributes to major clinical complications among COVID-19 patients, particularly the development of hypertension, myocardial injury, arrhythmia, acute coronary syndrome and increased coagulation in the circulatory system during and post-infection. Pre-existing metabolic disease, such as cardiovascular disease, obesity, diabetes, and non-alcoholic fatty liver disease, is associated with increased risk of hospitalization, persistent post-infection complications and worse outcomes in patients with COVID-19. This review overviews the biological features of Sars-CoV-2, highlights recent findings that delineate the pathological mechanisms of COVID-19 and the consequent clinical diseases.
Collapse
|
41
|
Role of Damage-Associated Molecular Pattern/Cell Death Pathways in Vaccine-Induced Immunity. Viruses 2021; 13:v13122340. [PMID: 34960608 PMCID: PMC8708515 DOI: 10.3390/v13122340] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
Immune responses induced by natural infection and vaccination are known to be initiated by the recognition of microbial patterns by cognate receptors, since microbes and most vaccine components contain pathogen-associated molecular patterns. Recent discoveries on the roles of damage-associated molecular patterns (DAMPs) and cell death in immunogenicity have improved our understanding of the mechanism underlying vaccine-induced immunity. DAMPs are usually immunologically inert, but can transform into alarming signals to activate the resting immune system in response to pathogenic infection, cellular stress and death, or tissue damage. The activation of DAMPs and cell death pathways can trigger local inflammation, occasionally mediating adaptive immunity, including antibody- and cell-mediated immune responses. Emerging evidence indicates that the components of vaccines and adjuvants induce immunogenicity via the stimulation of DAMP/cell death pathways. Furthermore, strategies for targeting this pathway to enhance immunogenicity are being investigated actively. In this review, we describe various DAMPs and focus on the roles of DAMP/cell death pathways in the context of vaccines for infectious diseases and cancer.
Collapse
|
42
|
Itagaki K, Riça I, Konecna B, Kim HI, Park J, Kaczmarek E, Hauser CJ. Role of Mitochondria-Derived Danger Signals Released After Injury in Systemic Inflammation and Sepsis. Antioxid Redox Signal 2021; 35:1273-1290. [PMID: 33847158 PMCID: PMC8905257 DOI: 10.1089/ars.2021.0052] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Sepsis is a major public health concern, with high mortality and morbidity, especially among patients undergoing trauma. It is characterized by a systemic inflammatory response syndrome (SIRS) occurring in response to infection. Although classically associated with pathogens, many patients with SIRS do not have infection. The variability of the disease course cannot be fully explained by our current understanding of its pathogenesis. Thus, other factors are likely to play key roles in the development and progression of SIRS/sepsis. Recent Advances: Circulating levels of damage-associated molecular patterns (DAMPs) seem to correlate with SIRS/sepsis morbidity and mortality. Of the known DAMPs, those of mitochondrial (mt) origin have been of particular interest, since their DNA (mtDNA) and formyl peptides (mtFPs) resemble bacterial DNA and peptides, and hence, when released, may be recognized as "danger signals." Critical Issues: mtDAMPs released after tissue injury trigger immune responses similar to those induced by pathogens. Thus, they can result in systemic inflammation and organ damage, similar to that observed in SIRS/sepsis. We will discuss recent findings on the roles of mtDAMPs, particularly regarding the less recognized mtFPs, in the activation of inflammatory responses and development of SIRS/sepsis. Future Directions: There are no established methods to predict the course of SIRS/sepsis, but clinical studies reveal that plasma levels of mtDAMPs may correlate with the outcome of the disease. We propose that non-pathogen-initiated, mtDAMPs-induced SIRS/sepsis events need further studies aimed at early clinical recognition and better treatment of this disease.
Collapse
Affiliation(s)
- Kiyoshi Itagaki
- Department of Surgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Ingred Riça
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Barbora Konecna
- Institute of Molecular Biomedicine, Comenius University, Bratislava, Slovakia
| | - Hyo In Kim
- Department of Surgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Jinbong Park
- Department of Surgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Elzbieta Kaczmarek
- Department of Surgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA.,Center for Vascular Biology Research, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Carl J Hauser
- Department of Surgery, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Lam LKM, Murphy S, Kokkinaki D, Venosa A, Sherrill-Mix S, Casu C, Rivella S, Weiner A, Park J, Shin S, Vaughan AE, Hahn BH, Odom John AR, Meyer NJ, Hunter CA, Worthen GS, Mangalmurti NS. DNA binding to TLR9 expressed by red blood cells promotes innate immune activation and anemia. Sci Transl Med 2021; 13:eabj1008. [PMID: 34669439 DOI: 10.1126/scitranslmed.abj1008] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- L K Metthew Lam
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sophia Murphy
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dimitra Kokkinaki
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Scott Sherrill-Mix
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carla Casu
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stefano Rivella
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Aaron Weiner
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Jeongho Park
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Beatrice H Hahn
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Audrey R Odom John
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nuala J Meyer
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA.,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - G Scott Worthen
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Division of Neonatalogy, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nilam S Mangalmurti
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
44
|
Sato H, Hoshi M, Ikeda F, Fujiyuki T, Yoneda M, Kai C. Downregulation of mitochondrial biogenesis by virus infection triggers antiviral responses by cyclic GMP-AMP synthase. PLoS Pathog 2021; 17:e1009841. [PMID: 34648591 PMCID: PMC8516216 DOI: 10.1371/journal.ppat.1009841] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/27/2021] [Indexed: 01/23/2023] Open
Abstract
In general, in mammalian cells, cytosolic DNA viruses are sensed by cyclic GMP-AMP synthase (cGAS), and RNA viruses are recognized by retinoic acid-inducible gene I (RIG-I)-like receptors, triggering a series of downstream innate antiviral signaling steps in the host. We previously reported that measles virus (MeV), which possesses an RNA genome, induces rapid antiviral responses, followed by comprehensive downregulation of host gene expression in epithelial cells. Interestingly, gene ontology analysis indicated that genes encoding mitochondrial proteins are enriched among the list of downregulated genes. To evaluate mitochondrial stress after MeV infection, we first observed the mitochondrial morphology of infected cells and found that significantly elongated mitochondrial networks with a hyperfused phenotype were formed. In addition, an increased amount of mitochondrial DNA (mtDNA) in the cytosol was detected during progression of infection. Based on these results, we show that cytosolic mtDNA released from hyperfused mitochondria during MeV infection is captured by cGAS and causes consequent priming of the DNA sensing pathway in addition to canonical RNA sensing. We also ascertained the contribution of cGAS to the in vivo pathogenicity of MeV. In addition, we found that other viruses that induce downregulation of mitochondrial biogenesis as seen for MeV cause similar mitochondrial hyperfusion and cytosolic mtDNA-priming antiviral responses. These findings indicate that the mtDNA-activated cGAS pathway is critical for full innate control of certain viruses, including RNA viruses that cause mitochondrial stress. Viruses exert their pathogenicity by targeting various cellular components in infected cells. In response, host cells have evolved strategies to sense intracellular pathogen-associated molecules, such as nucleic acids derived from infected virus, and trigger subsequent antiviral responses to counteract infection. Measles virus (MeV), the causative agent of human measles, is the most highly contagious virus, killing 300 children per day worldwide; thus MeV has been targeted for eradication by the World Health Organization. In the present study, we found that MeV causes downregulation of mitochondrial biogenesis accompanied with aberrant hyperfusion of mitochondria in the infected cells. Furthermore, we show that cytoplasmic release of mitochondrial DNA activates DNA sensor molecule, cGAS, in addition to the innate immune response induced by the viral component. Importantly, this phenomenon was also observed for viruses, both RNA and DNA, which target mitochondrial biogenesis. Our study provides new insights into the mitochondrial stress by virus infection and an important host defense system to suppress viral propagation.
Collapse
Affiliation(s)
- Hiroki Sato
- Infectious Disease Control Science, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- Molecular Virology, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Miho Hoshi
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Fusako Ikeda
- Division of Virological Medicine, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Tomoko Fujiyuki
- Infectious Disease Control Science, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- Virus Engineering, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Misako Yoneda
- Division of Virological Medicine, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
| | - Chieko Kai
- Infectious Disease Control Science, Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
45
|
Khin PP, Hong Y, Yeon M, Lee DH, Lee JH, Jun HS. Dulaglutide improves muscle function by attenuating inflammation through OPA-1-TLR-9 signaling in aged mice. Aging (Albany NY) 2021; 13:21962-21974. [PMID: 34537761 PMCID: PMC8507261 DOI: 10.18632/aging.203546] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/07/2021] [Indexed: 12/13/2022]
Abstract
Dulaglutide, a glucagon-like peptide-1 receptor (GLP-1R) agonist, is widely used to treat diabetes. However, its effects on muscle wasting due to aging are poorly understood. In the current study, we investigated the therapeutic potential and underlying mechanism of dulaglutide in muscle wasting in aged mice. Dulaglutide improved muscle mass and strength in aged mice. Histological analysis revealed that the cross-sectional area of the tibialis anterior (TA) in the dulaglutide-treated group was thicker than that in the vehicle group. Moreover, dulaglutide increased the shift toward middle and large-sized fibers in both young and aged mice compared to the vehicle. Dulaglutide increased myofiber type I and type IIa in young (18.5% and 8.2%) and aged (1.8% and 19.7%) mice, respectively, compared to the vehicle group. Peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis, decreased but increased by dulaglutide in aged mice. The expression of atrophic factors such as myostatin, atrogin-1, and muscle RING-finger protein-1 was decreased in aged mice, whereas that of the myogenic factor, MyoD, was increased in both young and aged mice following dulaglutide treatment. In aged mice, optic atrophy-1 (OPA-1) protein was decreased, whereas Toll-like receptor-9 (TLR-9) and its targeting inflammatory cytokines (interleukin-6 [IL-6] and tumor necrosis factor-α [TNF-α]) were elevated in the TA and quadriceps (QD) muscles. In contrast, dulaglutide administration reversed this expression pattern, thereby significantly attenuating the expression of inflammatory cytokines in aged mice. These data suggest that dulaglutide may exert beneficial effects in the treatment of muscle wasting due to aging.
Collapse
Affiliation(s)
- Phyu Phyu Khin
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Yeonhee Hong
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
| | - MyeongHoon Yeon
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea.,Gachon Medical and Convergence Institute, Gil Medical Center, Incheon, Korea
| | - Jong Han Lee
- Department of Marine Bio and Medical Science, Hanseo University, Seosan, Korea
| | - Hee-Sook Jun
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| |
Collapse
|
46
|
Grochowska J, Czerwinska J, Borowski LS, Szczesny RJ. Mitochondrial RNA, a new trigger of the innate immune system. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 13:e1690. [PMID: 34498404 DOI: 10.1002/wrna.1690] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria play a pivotal role in numerous cellular processes. One of them is regulation of the innate immune pathway. In this instance, mitochondria function in two different aspects of regulatory mechanisms. First, mitochondria are part of the antiviral signaling cascade that is triggered in the cytoplasm and transmitted to effector proteins through mitochondria-localized proteins. Second, mitochondria can become an endogenous source of innate immune stimuli. Under some pathophysiological conditions, mitochondria release to the cytoplasm immunogenic factors, such as mitochondrial nucleic acids. Here, we focus on immunogenic mitochondrial double-stranded RNA (mt-dsRNA) and its origin and metabolism. We discuss factors that are responsible for regulating mt-dsRNA and its escape from mitochondria, emphasizing the contribution of polynucleotide phosphorylase (PNPase, PNPT1). Finally, we review current knowledge of the role of PNPase in human health and disease. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Joanna Grochowska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Jolanta Czerwinska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Lukasz S Borowski
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Warsaw, Poland
| | - Roman J Szczesny
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
47
|
Targhetta VP, Amaral MA, Camara NOS. Through DNA sensors and hidden mitochondrial effects of SARS-CoV-2. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200183. [PMID: 34471404 PMCID: PMC8383803 DOI: 10.1590/1678-9199-jvatitd-2020-0183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/08/2021] [Indexed: 12/23/2022] Open
Abstract
The COVID-19 pandemic brought attention to studies about viral infections and their impact on the cell machinery. SARS-CoV-2, for example, invades the host cells by ACE2 interaction and possibly hijacks the mitochondria. To better understand the disease and to propose novel treatments, crucial aspects of SARS-CoV-2 enrolment with host mitochondria must be studied. The replicative process of the virus leads to consequences in mitochondrial function, and cell metabolism. The hijacking of mitochondria, on the other hand, can drive the extrusion of mitochondrial DNA (mtDNA) to the cytosol. Extracellular mtDNA evoke robust proinflammatory responses once detected, that may act in different pathways, eliciting important immune responses. However, few receptors are validated and are able to detect and respond to mtDNA. In this review, we propose that the mtDNA and its detection might be important in the immune process generated by SARS-CoV-2 and that this mechanism might be important in the lung pathogenesis seen in clinical symptoms. Therefore, investigating the mtDNA receptors and their signaling pathways might provide important clues for therapeutic interventions.
Collapse
Affiliation(s)
- Vitor Pedro Targhetta
- Department of Immunology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, SP, Brazil
| | - Mariana Abrantes Amaral
- Department of Nephrology, Paulista School of Medicine (EPM), Federal University of São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Niels Olsen Saraiva Camara
- Department of Immunology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, SP, Brazil
| |
Collapse
|
48
|
Schuliga M, Read J, Knight DA. Ageing mechanisms that contribute to tissue remodeling in lung disease. Ageing Res Rev 2021; 70:101405. [PMID: 34242806 DOI: 10.1016/j.arr.2021.101405] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Age is a major risk factor for chronic respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and certain phenotypes of asthma. The recent COVID-19 pandemic also highlights the increased susceptibility of the elderly to acute respiratory distress syndrome (ARDS), a diffuse inflammatory lung injury with often long-term effects (ie parenchymal fibrosis). Collectively, these lung conditions are characterized by a pathogenic reparative process that, rather than restoring organ function, contributes to structural and functional tissue decline. In the ageing lung, the homeostatic control of wound healing following challenge or injury has an increased likelihood of being perturbed, increasing susceptibility to disease. This loss of fidelity is a consequence of a diverse range of underlying ageing mechanisms including senescence, mitochondrial dysfunction, proteostatic stress and diminished autophagy that occur within the lung, as well as in other tissues, organs and systems of the body. These ageing pathways are highly interconnected, involving localized and systemic increases in inflammatory mediators and damage associated molecular patterns (DAMPs); along with corresponding changes in immune cell function, metabolism and composition of the pulmonary and gut microbiomes. Here we comprehensively review the roles of ageing mechanisms in the tissue remodeling of lung disease.
Collapse
Affiliation(s)
- Michael Schuliga
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| | - Jane Read
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Providence Health Care Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
49
|
Mitochondria-Induced Immune Response as a Trigger for Neurodegeneration: A Pathogen from Within. Int J Mol Sci 2021; 22:ijms22168523. [PMID: 34445229 PMCID: PMC8395232 DOI: 10.3390/ijms22168523] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 01/14/2023] Open
Abstract
Symbiosis between the mitochondrion and the ancestor of the eukaryotic cell allowed cellular complexity and supported life. Mitochondria have specialized in many key functions ensuring cell homeostasis and survival. Thus, proper communication between mitochondria and cell nucleus is paramount for cellular health. However, due to their archaebacterial origin, mitochondria possess a high immunogenic potential. Indeed, mitochondria have been identified as an intracellular source of molecules that can elicit cellular responses to pathogens. Compromised mitochondrial integrity leads to release of mitochondrial content into the cytosol, which triggers an unwanted cellular immune response. Mitochondrial nucleic acids (mtDNA and mtRNA) can interact with the same cytoplasmic sensors that are specialized in recognizing genetic material from pathogens. High-energy demanding cells, such as neurons, are highly affected by deficits in mitochondrial function. Notably, mitochondrial dysfunction, neurodegeneration, and chronic inflammation are concurrent events in many severe debilitating disorders. Interestingly in this context of pathology, increasing number of studies have detected immune-activating mtDNA and mtRNA that induce an aberrant production of pro-inflammatory cytokines and interferon effectors. Thus, this review provides new insights on mitochondria-driven inflammation as a potential therapeutic target for neurodegenerative and primary mitochondrial diseases.
Collapse
|
50
|
Son JM, Lee C. Aging: All roads lead to mitochondria. Semin Cell Dev Biol 2021; 116:160-168. [PMID: 33741252 PMCID: PMC9774040 DOI: 10.1016/j.semcdb.2021.02.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023]
Abstract
Mitochondria were described as early as 1890 as ubiquitous intracellular structures by Ernster and Schatz (1981) [1]. Since then, the accretion of knowledge in the past century has revealed much of the molecular details of mitochondria, ranging from mitochondrial origin, structure, metabolism, genetics, and signaling, and their implications in health and disease. We now know that mitochondria are remarkably multifunctional and deeply intertwined with many vital cellular processes. They are quasi-self organelles that still possess remnants of its bacterial ancestry, including an independent genome. The mitochondrial free radical theory of aging (MFRTA), which postulated that aging is a product of oxidative damage to mitochondrial DNA, provided a conceptual framework that put mitochondria on the map of aging research. However, several studies have more recently challenged the general validity of the theory, favoring novel ideas based on emerging evidence to understand how mitochondria contribute to aging and age-related diseases. One prominent topic of investigation lies on the fact that mitochondria are not only production sites for bioenergetics and macromolecules, but also regulatory hubs that communicate and coordinate many vital physiological processes at the cellular and organismal level. The bi-directional communication and coordination between the co-evolved mitochondrial and nuclear genomes is especially interesting in terms of cellular regulation. Mitochondria are dynamic and adaptive, rendering their function sensitive to cellular context. Tissues with high energy demands, such as the brain, seem to be uniquely affected by age-dependent mitochondrial dysfunction, providing a foundation for the development of novel mitochondrial-based therapeutics and diagnostics.
Collapse
Affiliation(s)
- Jyung Mean Son
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Changhan Lee
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA,USC Norris Comprehensive Cancer Center, Los Angeles, CA 90089, USA,Biomedical Sciences, Graduate School, Ajou University, Suwon 16499, South Korea,Corresponding author at: Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|