1
|
Truong TT, Huang CC, Chiu WT. Low-intensity pulsed ultrasound reduces oxidative and endoplasmic reticulum stress in motor neuron cells. ULTRASONICS 2024; 146:107499. [PMID: 39467391 DOI: 10.1016/j.ultras.2024.107499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Endoplasmic reticulum (ER) stress is associated with oxidative stress, which is integral to the development of various pathological conditions, including neurodegenerative disorders. In this study, using NSC-34-a hybrid cell line established by fusing motor neuron-rich embryonic spinal cord cells with mouse neuroblastoma cells-we investigated the effects of low-intensity pulsed ultrasound (LIPUS) stimulation on oxidative (reactive oxygen species)/ER stress-induced neurodegeneration. An ultrasound transducer with a center frequency of 1.15 MHz and a spatial peak temporal average intensity of 357 mW/cm2 was used for delivering ultrasound (for 8 min, via a water-filled tube) to motor neuron cells seeded in a plastic culture dish. LIPUS stimulation significantly increased the level of the antiapoptotic protein B-cell lymphoma 2 (BCL-2) and inhibited the expression of apoptosis-associated proteins such as BCL-2-associated X protein (BAX), CCAAT/enhancer-binding protein-homologous protein (CHOP), and caspase-12, thus extending the survival of motor neurons. LIPUS stimulation also enhanced Ca2+ signaling and activated the Ca2+-dependent transcription factors as nuclear factor of activated T cells (NFAT) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Furthermore, LIPUS stimulation induced the activation of the serine/threonine kinase protein kinase B (AKT). Thus, LIPUS stimulation prevented oxidative/ER stress-mediated mitochondrial dysfunction. In conclusion, as a safe and noninvasive method, LIPUS stimulation can facilitate further development of ultrasound neuromodulation as a tool for neuroscience research.
Collapse
Affiliation(s)
- Thi-Thuyet Truong
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701401, Taiwan
| | - Chih-Chung Huang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701401, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan 701401, Taiwan.
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan 701401, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan 701401, Taiwan.
| |
Collapse
|
2
|
Guo Q, Deng T, Du Y, Yao W, Tian W, Liao H, Wang Y, Li J, Yan W, Li Y. Impact of DEHP on mitochondria-associated endoplasmic reticulum membranes and reproductive toxicity in ovary. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116679. [PMID: 38981393 DOI: 10.1016/j.ecoenv.2024.116679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/12/2024] [Accepted: 06/30/2024] [Indexed: 07/11/2024]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a widely recognized environmental endocrine disruptor that potentially impacts female reproductive function, although the specific mechanisms leading to such impairment remain unclear. A growing body of research has revealed that the endoplasmic reticulum and mitochondrial function significantly influence oocyte quality. The structure of mitochondria-associated endoplasmic reticulum membranes (MAMs) is crucial for facilitating the exchange of Ca2+, lipids, and metabolites. This study aimed to investigate the alterations in the composition and function of MAMs after DEHP exposure and to elucidate the underlying mechanisms of ovarian toxicity. The female mice were exposed to DEHP at doses of 5 and 500 mg/kg/day for one month. The results revealed that DEHP exposure led to reduced serum anti-Müllerian hormone levels and increased atretic follicles in mice. DEHP induced endoplasmic reticulum stress and disrupted calcium homeostasis in oocytes. Furthermore, DEHP impaired the mitochondrial function of oocytes and reduced their membrane potential, and promoting apoptosis. Similar results were observed in human granulosa cells after exposure to mono-(2-ethylhexyl) phthalate (MEHP, metabolites of DEHP) in vitro. Proteomic analysis and transmission electron microscopy revealed modifications in the functional proteins and structure of the MAMs, and the suppression of oxidative phosphorylation pathways. The findings of this investigation provide a new perspective on the mechanism underlying the reproductive toxicity of DEHP in females.
Collapse
Affiliation(s)
- Qingchun Guo
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Taoran Deng
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yaoyao Du
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Wen Yao
- General Hospital of Central Theater Command, Wuhan, Hubei, PR China
| | - Wenqu Tian
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Hongmei Liao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yi Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Juan Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Wei Yan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| | - Yufeng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
3
|
Golmohammadi M, Meibodi SAA, Al-Hawary SIS, Gupta J, Sapaev IB, Najm MAA, Alwave M, Nazifi M, Rahmani M, Zamanian MY, Moriasi G. Neuroprotective effects of resveratrol on retinal ganglion cells in glaucoma in rodents: A narrative review. Animal Model Exp Med 2024; 7:195-207. [PMID: 38808561 PMCID: PMC11228121 DOI: 10.1002/ame2.12438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Glaucoma, an irreversible optic neuropathy, primarily affects retinal ganglion cells (RGC) and causes vision loss and blindness. The damage to RGCs in glaucoma occurs by various mechanisms, including elevated intraocular pressure, oxidative stress, inflammation, and other neurodegenerative processes. As the disease progresses, the loss of RGCs leads to vision loss. Therefore, protecting RGCs from damage and promoting their survival are important goals in managing glaucoma. In this regard, resveratrol (RES), a polyphenolic phytoalexin, exerts antioxidant effects and slows down the evolution and progression of glaucoma. The present review shows that RES plays a protective role in RGCs in cases of ischemic injury and hypoxia as well as in ErbB2 protein expression in the retina. Additionally, RES plays protective roles in RGCs by promoting cell growth, reducing apoptosis, and decreasing oxidative stress in H2O2-exposed RGCs. RES was also found to inhibit oxidative stress damage in RGCs and suppress the activation of mitogen-activated protein kinase signaling pathways. RES could alleviate retinal function impairment by suppressing the hypoxia-inducible factor-1 alpha/vascular endothelial growth factor and p38/p53 axes while stimulating the PI3K/Akt pathway. Therefore, RES might exert potential therapeutic effects for managing glaucoma by protecting RGCs from damage and promoting their survival.
Collapse
Affiliation(s)
- Maryam Golmohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Ibrohim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers, Tashkent, Uzbekistan
- New Uzbekistan University, Tashkent, Uzbekistan
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Marim Alwave
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Mozhgan Nazifi
- Department of Neurology, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammadreza Rahmani
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Gervason Moriasi
- Department of Medical Biochemistry, School of Medicine, College of Health Sciences, Mount Kenya University, Thika, Kenya
| |
Collapse
|
4
|
Zhang SX, Wang JJ, Starr CR, Lee EJ, Park KS, Zhylkibayev A, Medina A, Lin JH, Gorbatyuk M. The endoplasmic reticulum: Homeostasis and crosstalk in retinal health and disease. Prog Retin Eye Res 2024; 98:101231. [PMID: 38092262 PMCID: PMC11056313 DOI: 10.1016/j.preteyeres.2023.101231] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The endoplasmic reticulum (ER) is the largest intracellular organelle carrying out a broad range of important cellular functions including protein biosynthesis, folding, and trafficking, lipid and sterol biosynthesis, carbohydrate metabolism, and calcium storage and gated release. In addition, the ER makes close contact with multiple intracellular organelles such as mitochondria and the plasma membrane to actively regulate the biogenesis, remodeling, and function of these organelles. Therefore, maintaining a homeostatic and functional ER is critical for the survival and function of cells. This vital process is implemented through well-orchestrated signaling pathways of the unfolded protein response (UPR). The UPR is activated when misfolded or unfolded proteins accumulate in the ER, a condition known as ER stress, and functions to restore ER homeostasis thus promoting cell survival. However, prolonged activation or dysregulation of the UPR can lead to cell death and other detrimental events such as inflammation and oxidative stress; these processes are implicated in the pathogenesis of many human diseases including retinal disorders. In this review manuscript, we discuss the unique features of the ER and ER stress signaling in the retina and retinal neurons and describe recent advances in the research to uncover the role of ER stress signaling in neurodegenerative retinal diseases including age-related macular degeneration, inherited retinal degeneration, achromatopsia and cone diseases, and diabetic retinopathy. In some chapters, we highlight the complex interactions between the ER and other intracellular organelles focusing on mitochondria and illustrate how ER stress signaling regulates common cellular stress pathways such as autophagy. We also touch upon the integrated stress response in retinal degeneration and diabetic retinopathy. Finally, we provide an update on the current development of pharmacological agents targeting the UPR response and discuss some unresolved questions and knowledge gaps to be addressed by future research.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States.
| | - Josh J Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Christopher R Starr
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eun-Jin Lee
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Karen Sophia Park
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Assylbek Zhylkibayev
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jonathan H Lin
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
5
|
Tong D, Zhou J, Zhou J, Wang X, Gao B, Rui X, Liu L, Chen Q, Huang C. LAMC2 mitigates ER stress by enhancing ER-mitochondria interaction via binding to MYH9 and MYH10. Cancer Gene Ther 2024; 31:43-57. [PMID: 37891404 PMCID: PMC10794146 DOI: 10.1038/s41417-023-00680-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/01/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023]
Abstract
Highly proliferative and metastatic tumors are constantly exposed to both intrinsic and extrinsic factors that induce adaptation to stressful conditions. Chronic adaptation to endoplasmic reticulum (ER) ER stress is common to many different types of cancers, and poses a major challenge for acquired drug resistance. Here we report that LAMC2, an extracellular matrix protein upregulated in many types of cancers, is localized in the ER of lung, breast, and liver cancer cells. Under tunicamycin-induced ER stress, protein level of LAMC2 is upregulated. Transfection of cancer cells with LAMC2 resulted in the attenuation of ER stress phenotype, accompanied by elevation in mitochondrial membrane potential as well as reduction in reactive oxygen species (ROS) levels and apoptosis. In addition, LAMC2 forms protein complexes with MYH9 and MYH10 to promote mitochondrial aggregation and increased ER-mitochondria interaction at the perinuclear region. Moreover, overexpression of LAMC2 counteracts the effects of ER stress and promotes tumor growth in vivo. Taken together, our results revealed that in complex with MYH9 and MYH10, LAMC2 is essential for promoting ER-mitochondria interaction to alleviate ER stress and allow cancer cells to adapt and proliferate under stressful conditions. This study provides new insights and highlights the promising potential of LAMC2 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Dongdong Tong
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Jun Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Jing Zhou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Xiaofei Wang
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Beibei Gao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Xiaoyi Rui
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Liying Liu
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - QiaoYi Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China.
| | - Chen Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China.
- Biomedical Experimental Center of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, 710061, Xi'an, China.
| |
Collapse
|
6
|
Dong S, Wu Y, Zhang Y, Li S, Zhao Q, Liu S, Guo Y, Li X, Song K, Wu L, Wu L, Shi J, Gong L, Yu J. IP3R-1 aggravates endotoxin-induced acute lung injury in mice by regulating MAM formation and mitochondrial function. Exp Biol Med (Maywood) 2023; 248:2262-2272. [PMID: 38159072 PMCID: PMC10903239 DOI: 10.1177/15353702231220667] [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: 07/25/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024] Open
Abstract
Acute lung injury (ALI) caused by endotoxin represents one of the common clinical emergencies. Mitochondria-associated endoplasmic reticulum membranes (MAM) serve as a critical link between mitochondria and endoplasmic reticulum (ER), which has an essential effect on maintaining intracellular homeostasis. As an important component of MAM, type-1 inositol-1,4,5-trisphosphate receptor (IP3R-1) mediates the ER-to-mitochondrial transport of Ca2+. This study explored the role of IP3R-1 and MAM in ALI. Besides the levels of inflammasome-associated components interleukin (IL)-6, tumor necrosis factor (TNF)-α, and malonyldialdehyde (MDA) were increased in both bronchoalveolar lavage fluid (BALF) and serum, increased cross-sectional area of mitochondria, elevated MAM formation, and decreased respiratory control ratio (RCR) were observed within lung tissues collected in lipopolysaccharide (LPS)-treated mice, accompanied by upregulation of IP3R-1 in total lung lysates and MAM. Ca2+ uptake level in the mitochondria, production of reactive oxygen species (ROS) in the mitochondria, and the formation of MAM were elevated within LPS-treated MLE-12 cells, and all those changes in response to LPS were partly inhibited by knocking down of IP3R-1 expression in MLE-12 cells. Collectively, IP3R-1 has a critical effect on MAM formation and mitochondrial dysfunction, which could be innovative therapeutic targets for ALI caused by endotoxin.
Collapse
Affiliation(s)
| | | | | | - Shaona Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Qin Zhao
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Shasha Liu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Yan Guo
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Xiangyun Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Kai Song
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Lili Wu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Lina Wu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Jia Shi
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Lirong Gong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China
| |
Collapse
|
7
|
Lee EJ, Diaz-Aguilar MS, Min H, Choi J, Valdez Duran DA, Grandjean JM, Wiseman RL, Kroeger H, Lin JH. Mitochondria and Endoplasmic Reticulum Stress in Retinal Organoids from Patients with Vision Loss. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1721-1739. [PMID: 36535406 PMCID: PMC10616714 DOI: 10.1016/j.ajpath.2022.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Activating transcription factor 6 (ATF6), a key regulator of the unfolded protein response, plays a key role in endoplasmic reticulum function and protein homeostasis. Variants of ATF6 that abrogate transcriptional activity cause morphologic and molecular defects in cones, clinically manifesting as the human vision loss disease achromatopsia (ACHM). ATF6 is expressed in all retinal cells. However, the effect of disease-associated ATF6 variants on other retinal cell types remains unclear. Herein, this was investigated by analyzing bulk RNA-sequencing transcriptomes from retinal organoids generated from patients with ACHM, carrying homozygous loss-of-function ATF6 variants. Marked dysregulation in mitochondrial respiratory complex gene expression and disrupted mitochondrial morphology in ACHM retinal organoids were identified. This indicated that loss of ATF6 leads to previously unappreciated mitochondrial defects in the retina. Next, gene expression from control and ACHM retinal organoids were compared with transcriptome profiles of seven major retinal cell types generated from recent single-cell transcriptomic maps of nondiseased human retina. This indicated pronounced down-regulation of cone genes and up-regulation in Müller glia genes, with no significant effects on other retinal cells. Overall, the current analysis of ACHM patient retinal organoids identified new cellular and molecular phenotypes in addition to cone dysfunction: activation of Müller cells, increased endoplasmic reticulum stress, disrupted mitochondrial structure, and elevated respiratory chain activity gene expression.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Ophthalmology, Stanford University, Stanford, California; Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California
| | - Monica S Diaz-Aguilar
- Department of Ophthalmology, Stanford University, Stanford, California; Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California; Department of Medicine, Rush University Medical College, Chicago, Illinois
| | - Hyejung Min
- Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California
| | - Jihee Choi
- Department of Pathology, Stanford University, Stanford, California
| | | | - Julia M Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Heike Kroeger
- Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Jonathan H Lin
- Department of Ophthalmology, Stanford University, Stanford, California; Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California.
| |
Collapse
|
8
|
Li Y, Huang D, Jia L, Shangguan F, Gong S, Lan L, Song Z, Xu J, Yan C, Chen T, Tan Y, Liu Y, Huang X, Suzuki CK, Yang Z, Yang G, Lu B. LonP1 Links Mitochondria-ER Interaction to Regulate Heart Function. RESEARCH (WASHINGTON, D.C.) 2023; 6:0175. [PMID: 37333972 PMCID: PMC10275618 DOI: 10.34133/research.0175] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023]
Abstract
Interorganelle contacts and communications are increasingly recognized to play a vital role in cellular function and homeostasis. In particular, the mitochondria-endoplasmic reticulum (ER) membrane contact site (MAM) is known to regulate ion and lipid transfer, as well as signaling and organelle dynamics. However, the regulatory mechanisms of MAM formation and their function are still elusive. Here, we identify mitochondrial Lon protease (LonP1), a highly conserved mitochondrial matrix protease, as a new MAM tethering protein. The removal of LonP1 substantially reduces MAM formation and causes mitochondrial fragmentation. Furthermore, deletion of LonP1 in the cardiomyocytes of mouse heart impairs MAM integrity and mitochondrial fusion and activates the unfolded protein response within the ER (UPRER). Consequently, cardiac-specific LonP1 deficiency causes aberrant metabolic reprogramming and pathological heart remodeling. These findings demonstrate that LonP1 is a novel MAM-localized protein orchestrating MAM integrity, mitochondrial dynamics, and UPRER, offering exciting new insights into the potential therapeutic strategy for heart failure.
Collapse
Affiliation(s)
- Yujie Li
- The Affiliated Nanhua Hospital and School of Basic Medical Sciences, Hengyang Medical School,
University of South China, Hengyang, Hunan 421001, China
- School of Laboratory Medicine and Life Sciences,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention,
Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
| | - Dawei Huang
- School of Laboratory Medicine and Life Sciences,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lianqun Jia
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Ministry of Education of China,
Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, China
| | - Fugen Shangguan
- School of Laboratory Medicine and Life Sciences,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province,
The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shiwei Gong
- School of Laboratory Medicine and Life Sciences,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Linhua Lan
- School of Laboratory Medicine and Life Sciences,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province,
The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiyin Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences,
Wuhan University, Wuhan, Hubei 430072, China
| | - Juan Xu
- Nanjing Maternity and Child Health Care Hospital,
Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Chaojun Yan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences,
Wuhan University, Wuhan, Hubei 430072, China
| | - Tongke Chen
- Animal Center,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yin Tan
- Department of Cardiology,
The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Yongzhang Liu
- School of Laboratory Medicine and Life Sciences,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xingxu Huang
- School of Life Science and Technology,
Shanghai Tech University, Shanghai 201210, China
| | - Carolyn K. Suzuki
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School-Rutgers,
The State University of New Jersey, Newark, NJ, USA
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center,
Nanjing University, Nanjing 210061, China
| | - Guanlin Yang
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Ministry of Education of China,
Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, China
| | - Bin Lu
- The Affiliated Nanhua Hospital and School of Basic Medical Sciences, Hengyang Medical School,
University of South China, Hengyang, Hunan 421001, China
- School of Laboratory Medicine and Life Sciences,
Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- National Health Commission Key Laboratory of Birth Defect Research and Prevention,
Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
| |
Collapse
|
9
|
Resveratrol Suppresses Bupivacaine-Induced Spinal Neurotoxicity in Rats by Inhibiting Endoplasmic Reticulum Stress via SIRT1 Modulation. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1176232. [PMID: 36865484 PMCID: PMC9974252 DOI: 10.1155/2023/1176232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 02/23/2023]
Abstract
Bupivacaine (BUP) may cause neurotoxic effects after spinal anesthesia. Resveratrol (RSV), a natural agonist of Silent information regulator 1 (SIRT1), protects various tissues and organs from damage by regulating endoplasmic reticulum (ER) stress. The aim of this study is to explore whether RSV could alleviate the neurotoxicity induced by bupivacaine via regulating ER stress. We established a model of bupivacaine-induced spinal neurotoxicity in rats using intrathecal injection of 5% bupivacaine. The protective effect of RSV was evaluated by injecting intrathecally with 30 μg/μL RSV in total of 10 μL per day for 4 consecutive days. On day 3 after bupivacaine administration, tail-flick latency (TFL) tests and the Basso, Beattie, and Bresnahan (BBB) locomotor scores were assessed to neurological function, and the lumbar enlargement of the spinal cord was obtained. H&E and Nissl staining were used to evaluate the histomorphological changes and the number of survival neurons. TUNEL staining was conducted to determine apoptotic cells. The expression of proteins was detected by IHC, immunofluorescence, and western blot. The mRNA level of SIRT1 was determined by RT-PCR. Bupivacaine caused spinal cord neurotoxicity by inducing cell apoptosis and triggering ER stress. RSV treatment promoted the recovery of neurological dysfunction after bupivacaine administration by suppressing neuronal apoptosis and ER stress. Furthermore, RSV upregulated SIRT1 expression and inhibited PERK signaling pathway activation. In summary, resveratrol suppresses bupivacaine-induced spinal neurotoxicity in rats by inhibiting endoplasmic reticulum stress via SIRT1 modulation.
Collapse
|
10
|
Endoplasmic Reticulum Stress Signaling and Neuronal Cell Death. Int J Mol Sci 2022; 23:ijms232315186. [PMID: 36499512 PMCID: PMC9740965 DOI: 10.3390/ijms232315186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
Besides protein processing, the endoplasmic reticulum (ER) has several other functions such as lipid synthesis, the transfer of molecules to other cellular compartments, and the regulation of Ca2+ homeostasis. Before leaving the organelle, proteins must be folded and post-translationally modified. Protein folding and revision require molecular chaperones and a favorable ER environment. When in stressful situations, ER luminal conditions or chaperone capacity are altered, and the cell activates signaling cascades to restore a favorable folding environment triggering the so-called unfolded protein response (UPR) that can lead to autophagy to preserve cell integrity. However, when the UPR is disrupted or insufficient, cell death occurs. This review examines the links between UPR signaling, cell-protective responses, and death following ER stress with a particular focus on those mechanisms that operate in neurons.
Collapse
|
11
|
Endoplasmic Reticulum Stress Underlies Nanosilver-Induced Neurotoxicity in Immature Rat Brain. Int J Mol Sci 2022; 23:ijms232113013. [PMID: 36361797 PMCID: PMC9655133 DOI: 10.3390/ijms232113013] [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: 10/10/2022] [Revised: 10/23/2022] [Accepted: 10/24/2022] [Indexed: 12/03/2022] Open
Abstract
The growing production of silver nanoparticles (AgNPs), and their widespread use in medical and consumer products, poses a potential threat to the environment and raises questions about biosafety. Immature organisms are particularly susceptible to various insults during development. The biological characteristics of immature organisms are different from those of adults, and dictate the consequences of exposure to various toxic substances, including AgNPs. Nanoparticles are highly reactive and can easily cross the blood–brain barrier (BBB) to accumulate in brain tissues. It is therefore important to investigate the molecular mechanisms of AgNP-induced neurotoxicity in the developing brain. Immature 2-week-old rats were exposed to a low dose of AgNPs (0.2 mg/kg b.w.) over a long period. Subsequently, brain tissues of the animals were subjected to ultrastructural and molecular analyses to determine endoplasmic reticulum (ER) stress. Ultrastructural markers of ER stress, such as pathological alterations in the ER and elongated forms of mitochondria accompanied by autophagy structures, were confirmed to be present in AgNP-exposed rat brain. Evidence for induction of ER stress in neurons was also provided by molecular markers. Upregulation of genes related to the ER-stress-induced unfolded protein response (UPR) pathway, such as GRP78, PERK, and CHOP ATF-6, was observed at the transcriptional and translational levels. The results show that prolonged exposure of immature rats to a low dose of AgNPs during the developmental period leads to induction of ER stress in the neurons of the developing brain. Simultaneously, in response to AgNP-induced ER stress, neurons promote protective mechanisms that partially compensate for ER stress by regulating the biodynamic processes of mitochondria and autophagy.
Collapse
|
12
|
Ramaiah P, Patra I, Abbas A, Fadhil AA, Abohassan M, Al-Qaim ZH, Hameed NM, Al-Gazally ME, Kemil Almotlaq SS, Mustafa YF, Shiravand Y. Mitofusin-2 in cancer: Friend or foe? Arch Biochem Biophys 2022; 730:109395. [PMID: 36176224 DOI: 10.1016/j.abb.2022.109395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022]
Abstract
Cancer is a category of disorders characterized by excessive cell proliferation with the ability to infiltrate or disseminate to other organs of the body. Mitochondrial dysfunction, as one of the most prominent hallmarks of cancer cells, has been related to the onset and development of various cancers. Mitofusin 2 (MFN2) is a major mediator of mitochondrial fusion, endoplasmic reticulum (ER)-mitochondria interaction, mitophagy and axonal transport of mitochondria. Available data have shown that MFN2, which its alterations have been associated with mitochondrial dysfunction, could affect cancer initiation and progression. In fact, it showed that MFN2 may have a double-edged sword effect on cancer fate. Precisely, it demonstrated that MFN2, as a tumor suppressor, induces cancer cell apoptosis and inhibits cell proliferation via Ca2+ and Bax-mediated apoptosis and increases P21 and p27 levels, respectively. It also could suppress cell survival via inhibiting PI3K/Akt, Ras-ERK1/2-cyclin D1 and mTORC2/Akt signaling pathways. On the other hand, MFN2, as an oncogene, could increase cancer invasion via snail-mediated epithelial-mesenchymal transition (EMT) and in vivo tumorigenesis. While remarkable progress has been achieved in recent decades, further exploration is required to elucidate whether MFN2 could be a friend or it's an enemy. This study aimed to highlight the different functions of MFN2 in various cancers.
Collapse
Affiliation(s)
| | | | - Anum Abbas
- Basic Health Unit, Foundation University Medical College, Islamabad, Pakistan.
| | - Ali Abdulhussain Fadhil
- College of Medical Technology, Medical Lab Techniques, Al-farahidi University, Baghdad, Iraq
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 9088, Saudi Arabia
| | | | | | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul-41001, Iraq
| | - Yavar Shiravand
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80138, Naples, Italy.
| |
Collapse
|
13
|
Bhardwaj R, Bhardwaj A, Dhawan DK, Tandon C, Kaur T. 4-PBA rescues hyperoxaluria induced nephrolithiasis by modulating urinary glycoproteins: Cross talk between endoplasmic reticulum, calcium homeostasis and mitochondria. Life Sci 2022; 305:120786. [PMID: 35809664 DOI: 10.1016/j.lfs.2022.120786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/15/2022]
Abstract
AIM Urinary glycoproteins such as Tamm Horsfall Protein (THP) and Osteopontin (OPN) are well established key regulators of renal stone formation. Additionally, recent revelations have highlighted the influence of Endoplasmic Reticulum (ER) and mitochondria of crucial importance in nephrolithiasis. However, till date conclusive approach highlighting the influence of ER stress on urinary glycoproteins and chaperone in nephrolithiasis remains elusive. Therefore, the present study was focussed on deciphering the possible effect of 4-PBA mitigating ER stress on urinary glycoproteins and calnexin (chaperone) with emphasis on interlinking calcium homeostasis in hyperoxaluric rats. MATERIAL AND METHODS Post 9 days of treatment, animals were sacrificed, and renal tissues were investigated for urinary glycoproteins, calnexin, calcium homeostasis, ER environment, redox status, and mitochondrial linkage. KEY FINDINGS 4-PBA appreciably reversed the altered levels of THP, OPN, and calnexin observed along with curtailing the disrupted calcium homeostasis when assessed for SERCA activity and intra-cellular calcium levels. Additionally, significant improvement in the perturbed ER environment as verified by escalated ER stress markers, disturbed protein folding-aggregation-degradation (congo red assay) pathway, and redox status was found post 4-PBA intervention. Interestingly, linkage of ER stress and mitochondria was established under hyperoxaluric conditions when assessed for protein levels of VDAC1 and GRP75. SIGNIFICANCE 4-PBA treatment resulted in rectifying the repercussions of ER-mitochondrial caused distress when assessed for protein folding/aggregation/degradation events along with disturbed calcium homeostasis. The present study advocates the necessity to adopt a holistic vision towards hyperoxaluria with emphasis on glycoproteins and ER environment.
Collapse
Affiliation(s)
- Rishi Bhardwaj
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Ankita Bhardwaj
- Department of Biophysics, Panjab University, Chandigarh, India
| | | | | | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, India.
| |
Collapse
|
14
|
Liu R, Liu R, Song G, Li Q, Cui Z, Long Y. Mitochondria Dysfunction and Cell Apoptosis Limit Resistance of Nile Tilapia (Oreochromis niloticus) to Lethal Cold Stress. Animals (Basel) 2022; 12:ani12182382. [PMID: 36139242 PMCID: PMC9495169 DOI: 10.3390/ani12182382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Sensitivity of Nile tilapia (Oreochromis niloticus) to cold stress represents a major concern for both aquaculture and theoretical study; however, the cellular and molecular mechanisms determining cold susceptibility of it remain largely unknown. In this study, we first estimated the median survival time of juvenile Nile tilapia under exposure to lethal cold stress (12 °C). The fish were classified as cold-sensitive or cold-tolerant based on their behavioral manifestation after exposed to 12 °C for 3 days. Subsequently, histological, biochemical and gene expression analyses were performed for the fish with different cold resistance to explore the cellular and molecular events underlying cold susceptibility of Nile tilapia. We found that exposure of Nile tilapia to lethal cold stress caused systemic tissue structure changes, mitochondrial swelling and dysfunction, induction of apoptosis and endoplasmic reticulum (ER) stress-related genes and cell apoptosis. The extent of these adverse cellular and molecular events determines an individual’s ability to survive cold stress. Our data indicate that mitochondria dysfunction and mitochondria-mediated cell apoptosis are the main factors limiting Nile tilapia’s cold resistance. Abstract Inability of Nile tilapia (Oreochromis niloticus) to withstand cold stress represents a major economic concern, which restricts the culture area, limits the growing period and even results in mass mortality in cold seasons. However, the cellular and molecular mechanisms determining cold susceptibility of Nile tilapia remain largely unknown. In this study, we characterized the ability of juvenile Nile tilapia to survive lethal cold stress (12 °C) and the median survival time (LT50) of the experimental fish under exposure to 12 °C cold stress was estimated as 3.14 d. After being exposed to 12 °C for 3 d, the survivors that lost equilibrium (LE) and those that swam normally (NO) were regarded as cold-sensitive and cold-tolerant, respectively. The untreated (Ctrl), NO and LE fish were subjected to histological, biochemical and gene expression analyses to explore the cellular and molecular events underlying cold susceptibility of Nile tilapia. Exposure of Nile tilapia to lethal cold stress caused systemic tissue structure changes, mitochondrial swelling and dysfunction, induction of apoptosis and endoplasmic reticulum (ER) stress-related genes and cell apoptosis. The extent of these adverse cellular and molecular events determines an individual’s ability to survive cold stress. Our data indicate that mitochondria dysfunction and mitochondria-mediated cell apoptosis are the main factors limiting Nile tilapia’s cold resistance.
Collapse
Affiliation(s)
- Ran Liu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Renyan Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guili Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Qing Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zongbin Cui
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yong Long
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Correspondence: ; Tel.: +86-27-68780100
| |
Collapse
|
15
|
Bhardwaj A, Bhardwaj R, Saini A, Dhawan DK, Kaur T. Impact of Calcium Influx on Endoplasmic Reticulum in Excitotoxic Neurons: Role of Chemical Chaperone 4-PBA. Cell Mol Neurobiol 2022; 43:1619-1635. [PMID: 36002608 DOI: 10.1007/s10571-022-01271-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022]
Abstract
Excessive activation of α-amino-3-hydroxy-5-methyl-4-isoxazole propoinic acid (AMPA) receptors instigates excitotoxicity via enhanced calcium influx in the neurons thus inciting deleterious consequences. Additionally, Endoplasmic Reticulum (ER) is pivotal in maintaining the intracellular calcium balance. Considering this, studying the aftermath of enhanced calcium uptake by neurons and its effect on ER environment can assist in delineating the pathophysiological events incurred by excitotoxicty. The current study was premeditated to decipher the role of ER pertaining to calcium homeostasis in AMPA-induced excitotoxicity. The findings showed, increased intracellular calcium levels (measured by flowcytometry and spectroflourimeter using Fura 2AM) in AMPA excitotoxic animals (male Sprague dawely rats) (intra-hippocampal injection of 10 mM AMPA). Further, ER resident proteins like calnexin, PDI and ERp72 were found to be upregulated, which further modulated the functioning of ER membrane calcium channels viz. IP3R, RyR, and SERCA pump. Altered calcium homeostasis further led to ER stress and deranged the protein folding capacity of ER post AMPA toxicity, which was ascertained by unfolded protein response (UPR) pathway markers such as IRE1α, eIF2α, and ATF6α. Chemical chaperone, 4-phenybutric acid (4-PBA), ameliorated the protein folding capacity and subsequent UPR markers. In addition, modulation of calcium channels and calcium regulating machinery of ER post 4-PBA administration restored the calcium homeostasis. Therefore the study reinforces the significance of ER stress, a debilitating outcome of impaired calcium homeostasis, under AMPA-induced excitotoxicity. Also, employing chaperone-based therapeutic approach to curb ER stress can restore the calcium imbalance in the neuropathological diseases.
Collapse
Affiliation(s)
- Ankita Bhardwaj
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Rishi Bhardwaj
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | | | - Tanzeer Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
16
|
Proulx J, Stacy S, Park IW, Borgmann K. A Non-Canonical Role for IRE1α Links ER and Mitochondria as Key Regulators of Astrocyte Dysfunction: Implications in Methamphetamine use and HIV-Associated Neurocognitive Disorders. Front Neurosci 2022; 16:906651. [PMID: 35784841 PMCID: PMC9247407 DOI: 10.3389/fnins.2022.906651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Astrocytes are one of the most numerous glial cells in the central nervous system (CNS) and provide essential support to neurons to ensure CNS health and function. During a neuropathological challenge, such as during human immunodeficiency virus (HIV)-1 infection or (METH)amphetamine exposure, astrocytes shift their neuroprotective functions and can become neurotoxic. Identifying cellular and molecular mechanisms underlying astrocyte dysfunction are of heightened importance to optimize the coupling between astrocytes and neurons and ensure neuronal fitness against CNS pathology, including HIV-1-associated neurocognitive disorders (HAND) and METH use disorder. Mitochondria are essential organelles for regulating metabolic, antioxidant, and inflammatory profiles. Moreover, endoplasmic reticulum (ER)-associated signaling pathways, such as calcium and the unfolded protein response (UPR), are important messengers for cellular fate and function, including inflammation and mitochondrial homeostasis. Increasing evidence supports that the three arms of the UPR are involved in the direct contact and communication between ER and mitochondria through mitochondria-associated ER membranes (MAMs). The current study investigated the effects of HIV-1 infection and chronic METH exposure on astrocyte ER and mitochondrial homeostasis and then examined the three UPR messengers as potential regulators of astrocyte mitochondrial dysfunction. Using primary human astrocytes infected with pseudotyped HIV-1 or exposed to low doses of METH for 7 days, astrocytes had increased mitochondrial oxygen consumption rate (OCR), cytosolic calcium flux and protein expression of UPR mediators. Notably, inositol-requiring protein 1α (IRE1α) was most prominently upregulated following both HIV-1 infection and chronic METH exposure. Moreover, pharmacological inhibition of the three UPR arms highlighted IRE1α as a key regulator of astrocyte metabolic function. To further explore the regulatory role of astrocyte IRE1α, astrocytes were transfected with an IRE1α overexpression vector followed by activation with the proinflammatory cytokine interleukin 1β. Overall, our findings confirm IRE1α modulates astrocyte mitochondrial respiration, glycolytic function, morphological activation, inflammation, and glutamate uptake, highlighting a novel potential target for regulating astrocyte dysfunction. Finally, these findings suggest both canonical and non-canonical UPR mechanisms of astrocyte IRE1α. Thus, additional studies are needed to determine how to best balance astrocyte IRE1α functions to both promote astrocyte neuroprotective properties while preventing neurotoxic properties during CNS pathologies.
Collapse
|
17
|
Kuznetcova I, Bacher F, Alfadul SM, Tham MJR, Ang WH, Babak MV, Rapta P, Arion VB. Elucidation of Structure-Activity Relationships in Indolobenzazepine-Derived Ligands and Their Copper(II) Complexes: the Role of Key Structural Components and Insight into the Mechanism of Action. Inorg Chem 2022; 61:10167-10181. [PMID: 35713376 PMCID: PMC9490829 DOI: 10.1021/acs.inorgchem.2c01375] [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] [Indexed: 11/28/2022]
Abstract
Indolo[3,2-d][1]benzazepines (paullones), indolo[3,2-d][2]benzazepines, and indolo[2,3-d][2]benzazepines (latonduines) are isomeric scaffolds of current medicinal interest. Herein, we prepared a small library of novel indolo[3,2-d][2]benzazepine-derived ligands HL1-HL4 and copper(II) complexes 1-4. All compounds were characterized by spectroscopic methods (1H and 13C NMR, UV-vis, IR) and electrospray ionization (ESI) mass spectrometry, while complexes 2 and 3, in addition, by X-ray crystallography. Their purity was confirmed by HPLC coupled with high-resolution ESI mass spectrometry and/or elemental analysis. The stability of compounds in aqueous solutions in the presence of DMSO was confirmed by 1H NMR and UV-vis spectroscopy measurements. The compounds revealed high antiproliferative activity in vitro in the breast cancer cell line MDA-MB-231 and hepatocellular carcinoma cell line LM3 in the low micromolar to nanomolar concentration range. Important structure-activity relationships were deduced from the comparison of anticancer activities of HL1-HL4 and 1-4 with those of structurally similar paullone-derived (HL5-HL7 and 5-7) and latonduine-derived scaffolds (HL8-HL11 and 8-11). The high anticancer activity of the lead drug candidate 4 was linked to reactive oxygen species and endoplasmic reticulum stress induction, which were confirmed by fluorescent microscopy and Western blot analysis.
Collapse
Affiliation(s)
- Irina Kuznetcova
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Felix Bacher
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| | - Samah Mutasim Alfadul
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Max Jing Rui Tham
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117544, Singapore
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117544, Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong SAR 999077, China
| | - Peter Rapta
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, SK-81237 Bratislava, Slovak Republic
| | - Vladimir B Arion
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse 42, A-1090 Vienna, Austria
| |
Collapse
|
18
|
Li C, Ma D, Chen Y, Liu W, Jin F, Bo L. Selective inhibition of JNK located on mitochondria protects against mitochondrial dysfunction and cell death caused by endoplasmic reticulum stress in mice with LPS‑induced ALI/ARDS. Int J Mol Med 2022; 49:85. [PMID: 35514298 PMCID: PMC9106374 DOI: 10.3892/ijmm.2022.5141] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Few pharmacological interventions are able to improve the mortality rate of acute lung injury and acute respiratory distress syndrome (ALI/ARDS). The aim of this research was to elucidate whether endoplasmic reticulum (ER) stress and c-Jun-N-terminal kinase (JNK)-mitochondria pathways serve important roles in ALI/ARDS and to determine whether the key component Sab is a potential treatment target. The current study investigated the activation of ER stress and the JNK pathway, the content of JNK located on the mitochondria during ER stress and lipopolysaccharide (LPS)-induced ALI/ARDS by western blot analysis. The treatment effects of Tat-SabKIM1, a selective inhibitor of JNK located on mitochondria were explored by multiple methods including histopathological evaluation, lung cell apoptosis tested by TUNEL assay, mitochondrial membrane permeability and survival analysis. The results verified that ER stress was enhanced during LPS-induced ALI/ARDS and could induce activation of the JNK pathway and JNK-mitochondrial localization as well as mitochondrial dysfunction and cell death. Tat-SabKIM1 alleviated LPS injection-induced lung injury and improved mouse survival rates by specifically inhibiting JNK localization to mitochondria and mito-JNK signal activation without affecting cytosolic/nuclear JNK activation. The protective effect of Tat-SabKIM1 against ALI/ARDS was partly caused by inhibition of the excessive activation of mitochondria-mediated apoptosis and autophagy. These results showed the important role of Sab as a treatment target of ALI/ARDS and the potential treatment effect of Tat-SabKIM1. In conclusion, abnormal activation of the JNK-mitochondrial pathway could significantly disrupt the normal physiological function of lung cells, resulting in the occurrence of ALI/ARDS and selective inhibit of JNK located on mitochondria by Tat-SabKIM1 had a protective effect against the mitochondrial dysfunction and cell death caused by endoplasmic reticulum stress in mice with LPS-induced ALI/ARDS.
Collapse
Affiliation(s)
- Congcong Li
- Department of Respiratory and Critical Care Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Debin Ma
- Department of Respiratory and Critical Care Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Yan Chen
- Department of Respiratory and Critical Care Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| | - Wei Liu
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Faguang Jin
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Liyan Bo
- Department of Respiratory and Critical Care Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
19
|
Pereira AC, De Pascale J, Resende R, Cardoso S, Ferreira I, Neves BM, Carrascal MA, Zuzarte M, Madeira N, Morais S, Macedo A, do Carmo A, Moreira PI, Cruz MT, Pereira CF. ER-mitochondria communication is involved in NLRP3 inflammasome activation under stress conditions in the innate immune system. Cell Mol Life Sci 2022; 79:213. [PMID: 35344105 PMCID: PMC11072401 DOI: 10.1007/s00018-022-04211-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022]
Abstract
Endoplasmic reticulum (ER) stress and mitochondrial dysfunction, which are key events in the initiation and/or progression of several diseases, are correlated with alterations at ER-mitochondria contact sites, the so-called "Mitochondria-Associated Membranes" (MAMs). These intracellular structures are also implicated in NLRP3 inflammasome activation which is an important driver of sterile inflammation, however, the underlying molecular basis remains unclear. This work aimed to investigate the role of ER-mitochondria communication during ER stress-induced NLRP3 inflammasome activation in both peripheral and central innate immune systems, by using THP-1 human monocytes and BV2 microglia cells, respectively, as in vitro models. Markers of ER stress, mitochondrial dynamics and mass, as well as NLRP3 inflammasome activation were evaluated by Western Blot, IL-1β secretion was measured by ELISA, and ER-mitochondria contacts were quantified by transmission electron microscopy. Mitochondrial Ca2+ uptake and polarization were analyzed with fluorescent probes, and measurement of aconitase and SOD2 activities monitored mitochondrial ROS accumulation. ER stress was demonstrated to activate the NLRP3 inflammasome in both peripheral and central immune cells. Studies in monocytes indicate that ER stress-induced NLRP3 inflammasome activation occurs by a Ca2+-dependent and ROS-independent mechanism, which is coupled with upregulation of MAMs-resident chaperones, closer ER-mitochondria contacts, as well as mitochondrial depolarization and impaired dynamics. Moreover, enhanced ER stress-induced NLRP3 inflammasome activation in the immune system was found associated with pathological conditions since it was observed in monocytes derived from bipolar disorder (BD) patients, supporting a pro-inflammatory status in BD. In conclusion, by demonstrating that ER-mitochondria communication plays a key role in the response of the innate immune cells to ER stress, this work contributes to elucidate the molecular mechanisms underlying NLRP3 inflammasome activation under stress conditions, and to disclose novel potential therapeutic targets for diseases associated with sterile inflammation.
Collapse
Affiliation(s)
- Ana Catarina Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Jessica De Pascale
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
| | - Rosa Resende
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Susana Cardoso
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Isabel Ferreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University Coimbra, Coimbra, Portugal
| | - Bruno Miguel Neves
- iBiMED-Department of Medical Sciences and Institute for Biomedicine, University Aveiro, Aveiro, Portugal
| | - Mylène A Carrascal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Tecnimede Group, Sintra, Portugal
| | - Mónica Zuzarte
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- iCBR-Institute for Clinical and Biomedical Research, University Coimbra, Coimbra, Portugal
| | - Nuno Madeira
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- CIBIT-Coimbra Institute for Biomedical Imaging and Translational Research, University Coimbra, Coimbra, Portugal
- Department of Psychiatry, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sofia Morais
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Department of Psychiatry, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - António Macedo
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Department of Psychiatry, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Anália do Carmo
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Department of Clinical Pathology, CHUC-UC-Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- Faculty of Medicine, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Maria Teresa Cruz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University Coimbra, Coimbra, Portugal
| | - Cláudia F Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University Coimbra, Coimbra, Portugal.
- Faculty of Medicine, University Coimbra, Coimbra, Portugal.
- CACC-Clinical Academic Center of Coimbra, Coimbra, Portugal.
- , Coimbra, Portugal.
| |
Collapse
|
20
|
Gao H, He C, Hua R, Guo Y, Wang B, Liang C, Gao L, Shang H, Xu JD. Endoplasmic Reticulum Stress of Gut Enterocyte and Intestinal Diseases. Front Mol Biosci 2022; 9:817392. [PMID: 35402506 PMCID: PMC8988245 DOI: 10.3389/fmolb.2022.817392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum, a vast reticular membranous network from the nuclear envelope to the plasma membrane responsible for the synthesis, maturation, and trafficking of a wide range of proteins, is considerably sensitive to changes in its luminal homeostasis. The loss of ER luminal homeostasis leads to abnormalities referred to as endoplasmic reticulum (ER) stress. Thus, the cell activates an adaptive response known as the unfolded protein response (UPR), a mechanism to stabilize ER homeostasis under severe environmental conditions. ER stress has recently been postulated as a disease research breakthrough due to its significant role in multiple vital cellular functions. This has caused numerous reports that ER stress-induced cell dysfunction has been implicated as an essential contributor to the occurrence and development of many diseases, resulting in them targeting the relief of ER stress. This review aims to outline the multiple molecular mechanisms of ER stress that can elucidate ER as an expansive, membrane-enclosed organelle playing a crucial role in numerous cellular functions with evident changes of several cells encountering ER stress. Alongside, we mainly focused on the therapeutic potential of ER stress inhibition in gastrointestinal diseases such as inflammatory bowel disease (IBD) and colorectal cancer. To conclude, we reviewed advanced research and highlighted future treatment strategies of ER stress-associated conditions.
Collapse
Affiliation(s)
- Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuexin Guo
- Department of Oral Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- Undergraduate Student of 2018 Eight Program of Clinical Medicine, Peking University Health Science Center, Beijing, China
| | - Chen Liang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Hongwei Shang
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, China
| | - Jing-Dong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- *Correspondence: Jing-Dong Xu,
| |
Collapse
|
21
|
Abstract
Increasing evidence has demonstrated that obesity impairs female fertility and negatively affects human reproductive outcome following medically assisted reproduction (MAR) treatment. In the United States, 36.5% of women of reproductive age are obese. Obesity results not only in metabolic disorders including type II diabetes and cardiovascular disease, but might also be responsible for chronic inflammation and oxidative stress. Several studies have demonstrated that inflammation and reactive oxygen species (ROS) in the ovary modify steroidogenesis and might induce anovulation, as well as affecting oocyte meiotic maturation, leading to impaired oocyte quality and embryo developmental competence. Although the adverse effect of female obesity on human reproduction has been an object of debate in the past, there is growing evidence showing a link between female obesity and increased risk of infertility. However, further studies need to clarify some gaps in knowledge. We reviewed the recent evidence on the association between female obesity and infertility. In particular, we highlight the association between fat distribution and reproductive outcome, and how the inflammation and oxidative stress mechanisms might reduce ovarian function and oocyte quality. Finally, we evaluate the connection between female obesity and endometrial receptivity.
Collapse
|
22
|
Knoell J, Chillappagari S, Knudsen L, Korfei M, Dartsch R, Jonigk D, Kuehnel MP, Hoetzenecker K, Guenther A, Mahavadi P. PACS2-TRPV1 axis is required for ER-mitochondrial tethering during ER stress and lung fibrosis. Cell Mol Life Sci 2022; 79:151. [PMID: 35212819 PMCID: PMC8881280 DOI: 10.1007/s00018-022-04189-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 01/16/2023]
Abstract
Endoplasmic reticulum (ER) and mitochondria (mito) play a vital role in alveolar type II cell (AEC2) homeostasis and are both stressed in patients with idiopathic pulmonary fibrosis (IPF). Up to now, no data are available with regard to ER–mito cross talk and tethering under conditions of IPF. We here demonstrate that ER–mitochondrial tethering is reduced upon experimental ER stress in vitro and in the IPF AECII ex vivo, and this is—at least in part—due to decreased phosphofurin acidic cluster sorting protein 2 (PACS-2, also called PACS2) protein levels. PACS2 levels are influenced by its interaction with the transient receptor potential cation channel subfamily V member 1 (TRPV1) and can be experimentally modified by the TRPV1-modulating drug capsaicin (CPS). Employing alveolar epithelial cells with overexpression of the terminal ER stress signaling factor Chop or the IPF-associated surfactant protein C mutation (SPCΔexon4) in vitro, we observed a restoration of PACS2 levels upon treatment with CPS. Similarly, treatment of precision cut lung slices from IPF patients with CPS ex vivo forwarded similar effects. Importantly, in all models such kind of intervention also greatly reduced the extent of alveolar epithelial apoptosis. We therefore conclude that therapeutic targeting of the PACS2–TRPV1 axis represents an interesting novel, epithelial-protective approach in IPF.
Collapse
Affiliation(s)
- Jessica Knoell
- Department of Internal Medicine, Justus-Liebig University (JLU), Gaffkystraße 11, 35392, Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Shashi Chillappagari
- Department of Internal Medicine, Justus-Liebig University (JLU), Gaffkystraße 11, 35392, Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany.,Department of Biochemistry, Faculty of Medicine, JLU, Giessen, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany
| | - Martina Korfei
- Department of Internal Medicine, Justus-Liebig University (JLU), Gaffkystraße 11, 35392, Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Ruth Dartsch
- Department of Internal Medicine, Justus-Liebig University (JLU), Gaffkystraße 11, 35392, Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Mark P Kuehnel
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Vienna General Hospital, Vienna, Austria.,European IPF/ILD Registry and Biobank, Giessen, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig University (JLU), Gaffkystraße 11, 35392, Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany.,European IPF/ILD Registry and Biobank, Giessen, Germany.,Member of the Cardio-Pulmonary Institute (CPI), JLU, Giessen, Germany.,Lung Clinic, Agaplesion Evangelisches Krankenhaus Mittelhessen, Giessen, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig University (JLU), Gaffkystraße 11, 35392, Giessen, Germany. .,Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
23
|
Wang Y, Zhang X, Wen Y, Li S, Lu X, Xu R, Li C. Endoplasmic Reticulum-Mitochondria Contacts: A Potential Therapy Target for Cardiovascular Remodeling-Associated Diseases. Front Cell Dev Biol 2021; 9:774989. [PMID: 34858991 PMCID: PMC8631538 DOI: 10.3389/fcell.2021.774989] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular remodeling occurs in cardiomyocytes, collagen meshes, and vascular beds in the progress of cardiac insufficiency caused by a variety of cardiac diseases such as chronic ischemic heart disease, chronic overload heart disease, myocarditis, and myocardial infarction. The morphological changes that occur as a result of remodeling are the critical pathological basis for the occurrence and development of serious diseases and also determine morbidity and mortality. Therefore, the inhibition of remodeling is an important approach to prevent and treat heart failure and other related diseases. The endoplasmic reticulum (ER) and mitochondria are tightly linked by ER-mitochondria contacts (ERMCs). ERMCs play a vital role in different signaling pathways and provide a satisfactory structural platform for the ER and mitochondria to interact and maintain the normal function of cells, mainly by involving various cellular life processes such as lipid metabolism, calcium homeostasis, mitochondrial function, ER stress, and autophagy. Studies have shown that abnormal ERMCs may promote the occurrence and development of remodeling and participate in the formation of a variety of cardiovascular remodeling-associated diseases. This review focuses on the structure and function of the ERMCs, and the potential mechanism of ERMCs involved in cardiovascular remodeling, indicating that ERMCs may be a potential target for new therapeutic strategies against cardiovascular remodeling-induced diseases.
Collapse
Affiliation(s)
- Yu Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Emergency Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinrong Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ya Wen
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Sixuan Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaohui Lu
- Emergency Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ran Xu
- Jinan Tianqiao People's Hospital, Jinan, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
24
|
ER-misfolded proteins become sequestered with mitochondria and impair mitochondrial function. Commun Biol 2021; 4:1350. [PMID: 34857875 PMCID: PMC8640021 DOI: 10.1038/s42003-021-02873-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 11/08/2021] [Indexed: 01/23/2023] Open
Abstract
Proteostasis is a challenge for cellular organisms, as all known protein synthesis machineries are error-prone. Here we show by cell fractionation and microscopy studies that misfolded proteins formed in the endoplasmic reticulum can become associated with and partly transported into mitochondria, resulting in impaired mitochondrial function. Blocking the endoplasmic reticulum-mitochondria encounter structure (ERMES), but not the mitochondrial sorting and assembly machinery (SAM) or the mitochondrial surveillance pathway components Msp1 and Vms1, abrogated mitochondrial sequestration of ER-misfolded proteins. We term this mitochondria-associated proteostatic mechanism for ER-misfolded proteins ERAMS (ER-associated mitochondrial sequestration). We testify to the relevance of this pathway by using mutant α-1-antitrypsin as an example of a human disease-related misfolded ER protein, and we hypothesize that ERAMS plays a role in pathological features such as mitochondrial dysfunction.
Collapse
|
25
|
Kan S, Duan M, Liu Y, Wang C, Xie J. Role of Mitochondria in Physiology of Chondrocytes and Diseases of Osteoarthritis and Rheumatoid Arthritis. Cartilage 2021; 13:1102S-1121S. [PMID: 34894777 PMCID: PMC8804744 DOI: 10.1177/19476035211063858] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE OF REVIEW Mitochondria are recognized to be one of the most important organelles in chondrocytes for their role in triphosphate (ATP) generation through aerobic phosphorylation. Mitochondria also participate in many intracellular processes involving modulating reactive oxygen species (ROS), responding to instantaneous hypoxia stress, regulating cytoplasmic transport of calcium ion, and directing mitophagy to maintain the homeostasis of individual chondrocytes. DESIGNS To summarize the specific role of mitochondria in chondrocytes, we screened related papers in PubMed database and the search strategy is ((mitochondria) AND (chondrocyte)) AND (English [Language]). The articles published in the past 5 years were included and 130 papers were studied. RESULTS In recent years, the integrity of mitochondrial structure has been regarded as a prerequisite for normal chondrocyte survival and defect in mitochondrial function has been found in cartilage-related diseases, such as osteoarthritis (OA) and rheumatoid arthritis (RA). However, the understanding of mitochondria in cartilage is still largely limited. The mechanism on how the changes in mitochondrial structure and function directly lead to the occurrence and development of cartilage-related diseases remains to be elusive. CONCLUSION This review aims to summarize the role of mitochondria in chondrocytes under the physiological and pathological changes from ATP generation, calcium homeostasis, redox regulation, mitophagy modulation, mitochondria biogenesis to immune response activation. The enhanced understanding of molecular mechanisms in mitochondria might offer some new cues for cartilage remodeling and pathological intervention.
Collapse
Affiliation(s)
- Shiyi Kan
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chunli Wang
- “111” Project Laboratory of
Biomechanics and Tissue Repair, Bioengineering College, Chongqing University,
Chongqing, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China,“111” Project Laboratory of
Biomechanics and Tissue Repair, Bioengineering College, Chongqing University,
Chongqing, China,Lab of Bone & Joint Disease, State
Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan
University, Chengdu, China,Jing Xie, Lab of Bone & Joint Disease,
State Key Laboratory of Oral Diseases, West China Hospital of Stomatology,
Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
26
|
Zaitceva V, Kopeina GS, Zhivotovsky B. Anastasis: Return Journey from Cell Death. Cancers (Basel) 2021; 13:3671. [PMID: 34359573 PMCID: PMC8345212 DOI: 10.3390/cancers13153671] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
For over 20 years, it has been a dogma that once the integrity of mitochondria is disrupted and proapoptotic proteins that are normally located in the intermembrane space of mitochondria appeared in the cytoplasm, the process of cell death becomes inevitable. However, it has been recently shown that upon removal of the death signal, even at the stage of disturbance in the mitochondria, cells can recover and continue to grow. This phenomenon was named anastasis. Here, we will critically discuss the present knowledge concerning the mechanisms of cell death reversal, or development of anastasis, methods for its detection, and what role signaling from different intracellular compartments plays in anastasis stimulation.
Collapse
Affiliation(s)
- Victoria Zaitceva
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; (V.Z.); (G.S.K.)
| | - Gelina S. Kopeina
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; (V.Z.); (G.S.K.)
| | - Boris Zhivotovsky
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; (V.Z.); (G.S.K.)
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden
| |
Collapse
|
27
|
He J, Gong M, Wang Z, Liu D, Xie B, Luo C, Li G, Tse G, Liu T. Cardiac abnormalities after induction of endoplasmic reticulum stress are associated with mitochondrial dysfunction and connexin43 expression. Clin Exp Pharmacol Physiol 2021; 48:1371-1381. [PMID: 34133785 DOI: 10.1111/1440-1681.13541] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/30/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER) is responsible for protein synthesis and calcium storage. ER stress, reflected by protein unfolding and calcium handling abnormalities, has been studied as a pathogenic factor in cardiovascular diseases. The aim of this study is to examine the effects of ER stress on mechanical and electrophysiological functions in the heart and explore the underlying molecular mechanisms. A total of 30 rats were randomly divided into control, ER stress inducer (tunicamycin[TN]) and ER stress inhibitor (tunicamycin+4-phenylbutyric acid [4-PBA]) groups. ER stress induction led to significantly systolic and diastolic dysfunction as reflected by maximal increasing/decreasing rate of left intraventricular pressure (±dp/dt), left ventricular peaksystolic pressure(LVSP) and LV end-diastolic pressure(LVEDP). Epicardial mapping performed in vivo revealed reduced conduction velocity and increased conduction heterogeneity associated with the development of spontaneous ventricular tachycardia. Masson's trichrome staining revealed marked fibrosis in the myocardial interstitial and sub-pericardial regions, and thickened epicardium. Western blot analysis revealed increased pro-fibrotic factor transforming growth factor-β1 (TGF-β1), decreased mitochondrial biogenesis protein peroxlsome proliferator-activated receptor-γ coactlvator-1α (PGC-1a), and decreased mitochondrial fusion protein mitofusin-2 (MFN2). These changes were associated with mitochondria dysfunction and connexin 43(CX43)translocation to mitochondria. These abnormalities can be partially prevented by the ER stress inhibitor 4-PBA. Our study shows that ER stress induction can produce cardiac electrical and mechanism dysfunction as well as structural remodelling. Mitochondrial function alterations are contributed by CX43 transposition to mitochondria. These abnormalities can be partially prevented by the ER stress inhibitor 4-PBA.
Collapse
Affiliation(s)
- Jinli He
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Mengqi Gong
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China.,Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zaojia Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Daiqi Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bingxin Xie
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Cunjin Luo
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, UK
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.,Kent and Medway Medical School, Canterbury, UK
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
28
|
Loussouarn C, Pers YM, Bony C, Jorgensen C, Noël D. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Regulate the Mitochondrial Metabolism via Transfer of miRNAs. Front Immunol 2021; 12:623973. [PMID: 33796099 PMCID: PMC8007981 DOI: 10.3389/fimmu.2021.623973] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are the most commonly tested adult progenitor cells in regenerative medicine. They stimulate tissue repair primarily through the secretion of immune-regulatory and pro-regenerative factors. There is increasing evidence that most of these factors are carried on extracellular vesicles (EVs) that are released by MSCs, either spontaneously or after activation. Exosomes and microvesicles are the most investigated types of EVs that act through uptake by target cells and cargo release inside the cytoplasm or through interactions with receptors expressed on target cells to stimulate downstream intracellular pathways. They convey different types of molecules, including proteins, lipids and acid nucleics among which, miRNAs are the most widely studied. The cargo of EVs can be impacted by the culture or environmental conditions that MSCs encounter and by changes in the energy metabolism that regulate the functional properties of MSCs. On the other hand, MSC-derived EVs are also reported to impact the metabolism of target cells. In the present review, we discuss the role of MSC-EVs in the regulation of the energy metabolism and oxidative stress of target cells and tissues with a focus on the role of miRNAs.
Collapse
Affiliation(s)
- Claire Loussouarn
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Yves-Marie Pers
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, Lapeyronie University Hospital, Montpellier, France
| | - Claire Bony
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, Lapeyronie University Hospital, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.,Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Department of Rheumatology, Lapeyronie University Hospital, Montpellier, France
| |
Collapse
|
29
|
Ma RH, Ni ZJ, Thakur K, Zhang F, Zhang YY, Zhang JG, Wei ZJ. Natural Compounds Play Therapeutic Roles in Various Human Pathologies via Regulating Endoplasmic Reticulum Pathway. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
30
|
Victor P, Sarada D, Ramkumar KM. Crosstalk between endoplasmic reticulum stress and oxidative stress: Focus on protein disulfide isomerase and endoplasmic reticulum oxidase 1. Eur J Pharmacol 2020; 892:173749. [PMID: 33245896 DOI: 10.1016/j.ejphar.2020.173749] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022]
Abstract
Cellular stress and inflammation, establishing as disease pathology, have reached great heights in the last few decades. Stress conditions such as hyperglycemia, hyperlipidemia and lipoproteins are known to disturb proteostasis resulting in the accumulation of unfolded or misfolded proteins, alteration in calcium homeostasis culminating in unfolded protein response. Protein disulfide isomerase and endoplasmic reticulum oxidase-1 are the key players in protein folding. The protein folding process assisted by endoplasmic reticulum oxidase-1 results in the production of reactive oxygen species in the lumen of the endoplasmic reticulum. Production of reactive oxygen species beyond the quenching capacity of the antioxidant systems perturbs ER homeostasis. Endoplasmic reticulum stress also induces the production of cytokines leading to inflammatory responses. This has been proven to be the major causative factor for various pathophysiological states compared to other cellular triggers in diseases, which further manifests to increased oxidative stress, mitochondrial dysfunction, and altered inflammatory responses, deleterious to cellular physiology and homeostasis. Numerous studies have drawn correlations between the progression of several diseases in association with endoplasmic reticulum stress, redox protein folding, oxidative stress and inflammatory responses. This review aims to provide an insight into the role of protein disulfide isomerase and endoplasmic reticulum oxidase-1 in endoplasmic reticulum stress, unfolded protein response, mitochondrial dysfunction, and inflammatory responses, which exacerbate the progression of various diseases.
Collapse
Affiliation(s)
- Paul Victor
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India
| | - Dronamraju Sarada
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bio-engineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India; Life Science Division, SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, Tamil Nadu, India.
| |
Collapse
|
31
|
Ho G, Takamatsu Y, Wada R, Sugama S, Waragai M, Takenouchi T, Masliah E, Hashimoto M. Connecting Alzheimer's Disease With Diabetes Mellitus Through Amyloidogenic Evolvability. Front Aging Neurosci 2020; 12:576192. [PMID: 33192467 PMCID: PMC7655535 DOI: 10.3389/fnagi.2020.576192] [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: 06/25/2020] [Accepted: 08/28/2020] [Indexed: 01/26/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) has been clearlylinked to oxidative stress and amylin amyloidosis in pancreatic β-cells. Yet despite extensive investigation, the biological significance of this is not fully understood. Recently, we proposed that Alzheimer's disease (AD)-relevant amyloidogenic proteins (APs), such as amyloid-β (Aβ) and tau, might be involved in evolvability against diverse stressors in the brain. Given the analogous cellular stress environments shared by both T2DM and AD, the objective of this study is to explore T2DM pathogenesis from the viewpoint of amyloidogenic evolvability. Similar to AD-related APs, protofibrillar amylin might confer resistance against the multiple stressors in β-cells and be transmitted to offspring to deliver stress information, in the absence of which, type 1 DM (T1DM) in offspring might develop. On the contrary, T2DM may be manifested through an antagonistic pleiotropy mechanism during parental aging. Such evolvability-associated processes might be affected by parental diabetic conditions, including T1DM and T2DM. Furthermore, the T2DM-mediated increase in AD risk during aging might be attributed to an interaction of amylin with AD-related APs through evolvability, in which amylin protofibrillar formation presumably caused by adiponectin (APN) resistance could increase protofibril formation of AD-related APs in evolvability and subsequently lead to T2DM promotion of AD through antagonistic pleiotropy in aging. This suggests that targeting APN combined with an anti-T2DM agent might be therapeutic against neurodegeneration. Collectively, T1DM and T2DM might be linked through amylin evolvability, and a better understanding of amyloidogenic evolvability might also reveal clues to therapeutic interventions for AD comorbid with T2DM.
Collapse
Affiliation(s)
- Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, United States
| | | | - Ryoko Wada
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Masaaki Waragai
- PCND Neuroscience Research Institute, Poway, CA, United States
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Eliezer Masliah
- Division of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, United States
| | | |
Collapse
|
32
|
Li R, Wang Y, Chen P, Meng J, Zhang H. Inhibiting endoplasmic reticulum stress by activation of G-protein-coupled estrogen receptor to protect retinal astrocytes under hyperoxia. J Biochem Mol Toxicol 2020; 35:e22641. [PMID: 32996202 DOI: 10.1002/jbt.22641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/27/2020] [Accepted: 09/16/2020] [Indexed: 12/30/2022]
Abstract
Retinal vascularization is arrested at the early (hyperoxia) stage in retinopathy of prematurity (ROP), a leading cause of blindness in children. Estrogen was reported to alleviate ROP by inhibiting reactive oxygen species, the upstream signaling molecules of endoplasmic reticulum stress (ERS). Astrocytes have long been proposed to guide angiogenesis, because they form a reticular network that provides a substrate for migrating endothelial cells. However, the factors that control the vascularization of the immature retina and the therapeutic mechanism of estrogen in early ROP remain poorly understood. This study aimed to investigate the role of G-protein-coupled estrogen receptor (GPER), an estrogen receptor distributed in the endoplasmic reticulum (ER), in protecting retinal astrocytes under hyperoxia and the association with ERS. The results showed that GPER was widely expressed in retinal astrocytes. GPER activation increases cell viability, decreases apoptosis, and autophagy of retinal astrocytes, decreases inositol-1,4,5-triphosphate receptor activity, and increases Ca2+ concentration in ER of astrocytes under hyperoxia. GPER blockade reversed all of these changes. Together, our findings indicate that GPER can protect the survival of retinal astrocytes by inhibiting ERS under hyperoxia.
Collapse
Affiliation(s)
- Rong Li
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China
| | - Yao Wang
- Department of Ophthalmology, Eye Institute of Shaanxi Province and Xi'an First Hospital, Xi'an, Shaanxi, China
| | - Pei Chen
- Department of Ophthalmology, Eye Institute of Shaanxi Province and Xi'an First Hospital, Xi'an, Shaanxi, China
| | - Jiamin Meng
- Bioengineering Specialty, School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Hongbing Zhang
- Department of Ophthalmology, Eye Institute of Shaanxi Province and Xi'an First Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
33
|
G-protein coupled estrogen receptor activation protects the viability of hyperoxia-treated primary murine retinal microglia by reducing ER stress. Aging (Albany NY) 2020; 12:17367-17379. [PMID: 32920550 PMCID: PMC7521534 DOI: 10.18632/aging.103733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/19/2020] [Indexed: 01/24/2023]
Abstract
In this study, we investigated the effects of G-protein coupled estrogen receptor (GPER) activation in the early phase of retinopathy of prematurity (ROP) and its association with endoplasmic reticulum (ER) stress using primary murine retinal microglia as an experimental model. Fluorescence microscopy results show that the CD11c-positive primary retinal microglia in vitro cultured for 14 days were GPER-positive. GPER activation using GPER-agonist G-1 reduced autophagy and increased the viability of the hyperoxia-treated primary murine retinal microglia. Furthermore, GPER activation reduced the expression of ER stress-related proteins, IRE1α, PERK and ATF6 in the hyperoxia-treated primary murine retinal microglia compared to the corresponding controls. GPER activation significantly reduced a time-dependent increase in IP3R-dependent calcium release from the ER, thereby maintaining higher calcium levels in the ER of hyperoxia-treated primary retinal microglia. However, the protective effects of G-1 on the hyperoxia-treated primary retinal microglia were eliminated by inactivation of GPER using the GPER-antagonist, G-15. In conclusion, our study demonstrates that GPER activation enhances the survival of hyperoxia-treated primary retinal microglia by reducing ER stress. Our study demonstrates the therapeutic potential of GPER agonists such as G-1 in the early phase of ROP.
Collapse
|
34
|
Ren J, Sun M, Zhou H, Ajoolabady A, Zhou Y, Tao J, Sowers JR, Zhang Y. FUNDC1 interacts with FBXL2 to govern mitochondrial integrity and cardiac function through an IP3R3-dependent manner in obesity. SCIENCE ADVANCES 2020; 6:6/38/eabc8561. [PMID: 32938669 PMCID: PMC7494344 DOI: 10.1126/sciadv.abc8561] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/03/2020] [Indexed: 05/13/2023]
Abstract
Defective mitophagy is causally linked to obesity complications. Here, we identified an interaction between mitophagy protein FUNDC1 (FUN14 domain containing 1) and receptor subunit of human SCF (SKP1/cullin/F-box protein) ubiquitin ligase complex FBXL2 as a gatekeeper for mitochondrial Ca2+ homeostasis through degradation of IP3R3 (inositol 1,4,5-trisphosphate receptor type 3). Loss of FUNDC1 in FUNDC1-/- mice accentuated high-fat diet-induced cardiac remodeling, functional and mitochondrial anomalies, cell death, rise in IP3R3, and Ca2+ overload. Mass spectrometry and co-immunoprecipitation analyses revealed an interaction between FUNDC1 and FBXL2. Truncated mutants of Fbox (Delta-F-box) disengaged FBXL2 interaction with FUNDC1. Activation or transfection of FBXL2, inhibition of IP3R3 alleviated, whereas disruption of FBXL2 localization sensitized lipotoxicity-induced cardiac damage. FUNDC1 deficiency accelerated and decelerated palmitic acid-induced degradation of FBXL2 and IP3R3, respectively. Our data suggest an essential role for interaction between FUNDC1 and FBXL2 in preserving mitochondrial Ca2+ homeostasis and cardiac function in obese hearts.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Pathology, University of Washington Seattle, Seattle, WA 98195, USA
| | - Mingming Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Hao Zhou
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Amir Ajoolabady
- University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5185715179, Iran
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510000, China
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri Columbia, Columbia, MO 65212, USA
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
35
|
Andhavarapu S, Katuri A, Bryant J, Patel V, Gupta U, Asemu G, Makar TK. Intersecting roles of ER stress, mitochondrial dysfunction, autophagy, and calcium homeostasis in HIV-associated neurocognitive disorder. J Neurovirol 2020; 26:664-675. [PMID: 32804309 DOI: 10.1007/s13365-020-00861-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/22/2020] [Accepted: 05/19/2020] [Indexed: 01/04/2023]
Abstract
HIV-associated neurocognitive disorder (HAND) is a collective term describing the spectrum of neurocognitive deficits that arise from HIV infection. Although the introduction to highly active antiretroviral therapy (HAART) has prolonged the lifespan of HIV patients, neurocognitive impairments remain prevalent, as patients are left perpetually with HIV. Currently, physicians face a challenge in treating HAND patients, so a greater understanding of the mechanisms underlying HAND pathology has been a growing focus in HIV research. Recent research has revealed the role disrupted calcium homeostasis in HIV-mediated neurotoxicity. Calcium plays a well-established role in the crosstalk between the mitochondrion and ER as well as in regulating autophagy, and ER stress, mitochondrial dysfunction, and impaired autophagic activity are considered hallmarks in several neurodegenerative and neurocognitive disorders. Therefore, it is paramount that the intricate inter-organelle signaling in relation to calcium homeostasis during HIV infection and the development of HAND is elucidated. This review consolidates current knowledge regarding the neuropathology of neurocognitive disorders and HIV infection with a focus on the underlying role of calcium during ER stress, mitochondrial dysfunction, and autophagy associated with the progression of HAND. The details of this intricate crosstalk during HAND remain relatively unknown; further research in this field can potentially aid in the development of improved therapy for patients suffering from HAND.
Collapse
Affiliation(s)
- Sanketh Andhavarapu
- Institute of Human Virology, University of Maryland, 725 W Lombard St, Baltimore, MD, 21201, USA
| | - Akhil Katuri
- Institute of Human Virology, University of Maryland, 725 W Lombard St, Baltimore, MD, 21201, USA
| | - Joseph Bryant
- Institute of Human Virology, University of Maryland, 725 W Lombard St, Baltimore, MD, 21201, USA
| | - Vivek Patel
- Institute of Human Virology, University of Maryland, 725 W Lombard St, Baltimore, MD, 21201, USA
| | - Udit Gupta
- Institute of Human Virology, University of Maryland, 725 W Lombard St, Baltimore, MD, 21201, USA
| | - Girma Asemu
- Institute of Human Virology, University of Maryland, 725 W Lombard St, Baltimore, MD, 21201, USA
| | - Tapas K Makar
- Institute of Human Virology, University of Maryland, 725 W Lombard St, Baltimore, MD, 21201, USA. .,VA Medical Center, Baltimore, MD, 21201, USA.
| |
Collapse
|
36
|
Endoplasmic Reticulum Stress (ER Stress) and Unfolded Protein Response (UPR) Occur in a Rat Varicocele Testis Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5909306. [PMID: 32802266 PMCID: PMC7411497 DOI: 10.1155/2020/5909306] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/14/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Using a surgically induced varicocele rat model, we show here strong evidence that the misfolded/unfolded protein response that is part of the stress response of the endoplasmic reticulum (ER) is activated in the varicocele testis (VCL), leading to the induction of apoptosis. To support this hypothesis, it is observed that the spliced variant of the X-box protein 1 (XBP1s), resulting from the activation of the inositol-requiring enzyme 1 (IRE1) membrane sensor, is significantly more represented in VCL testicular extracts. The activation of the IRE1/XBP1s pathway is also supported by the observation that the VCL testes show an increase phosphorylation of the c-Jun-kinase (JNK) known to be one intermediate of this pathway and an increased level of caspase-3, the terminal apoptotic effector, partly explaining the apoptotic status of the VCL testis.
Collapse
|
37
|
Di Meo S, Venditti P. Evolution of the Knowledge of Free Radicals and Other Oxidants. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9829176. [PMID: 32411336 PMCID: PMC7201853 DOI: 10.1155/2020/9829176] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
Free radicals are chemical species (atoms, molecules, or ions) containing one or more unpaired electrons in their external orbitals and generally display a remarkable reactivity. The evidence of their existence was obtained only at the beginning of the 20th century. Chemists gradually ascertained the involvement of free radicals in organic reactions and, in the middle of the 20th century, their production in biological systems. For several decades, free radicals were thought to cause exclusively damaging effects . This idea was mainly supported by the finding that oxygen free radicals readily react with all biological macromolecules inducing their oxidative modification and loss of function. Moreover, evidence was obtained that when, in the living organism, free radicals are not neutralized by systems of biochemical defences, many pathological conditions develop. However, after some time, it became clear that the living systems not only had adapted to the coexistence with free radicals but also developed methods to turn these toxic substances to their advantage by using them in critical physiological processes. Therefore, free radicals play a dual role in living systems: they are toxic by-products of aerobic metabolism, causing oxidative damage and tissue dysfunction, and serve as molecular signals activating beneficial stress responses. This discovery also changed the way we consider antioxidants. Their use is usually regarded as helpful to counteract the damaging effects of free radicals but sometimes is harmful as it can block adaptive responses induced by low levels of radicals.
Collapse
Affiliation(s)
- Sergio Di Meo
- Università degli Studi di Napoli Federico II Dipartimento di Biologia, Complesso, Universitario Monte Sant'Angelo, Via Cinthia, I-80126 Napoli, Italy
| | - Paola Venditti
- Università degli Studi di Napoli Federico II Dipartimento di Biologia, Complesso, Universitario Monte Sant'Angelo, Via Cinthia, I-80126 Napoli, Italy
| |
Collapse
|
38
|
Park HB, Park YR, Kim MJ, Jung BD, Park CK, Cheong HT. Endoplasmic Reticulum (ER) Stress Inhibitor or Antioxidant Treatments during Micromanipulation Can Inhibit Both ER and Oxidative Stresses in Porcine SCNT Embryos. Dev Reprod 2020; 24:31-41. [PMID: 32411916 PMCID: PMC7201064 DOI: 10.12717/dr.2020.24.1.31] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/17/2020] [Accepted: 02/01/2020] [Indexed: 12/21/2022]
Abstract
We investigated the effects of endoplasmic reticulum (ER) stress inhibitor and
antioxidant treatments during the micromanipulation of somatic cell nuclear
transfer (SCNT) on in vitro development of SCNT embryos.
Tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor and vitamin C (Vit.
C), an antioxidant, were treated by alone or in combination, then, the level of
X-box binding protein 1 (Xbp1) splicing and the expressions of
ER stress-associated genes, oxidative stress-related genes, and apoptotic genes
were confirmed in the 1-cell and blastocyst stages. In the 1-cell stage, the
levels of Xbp1 splicing were significantly decreased in TUDCA
and Vit. C treatment groups compared to the control
(p<0.05). In addition, the expression levels of most ER
stress-associated genes and oxidative stress-related genes were significantly
lower in all treatment groups than the control (p<0.05),
and the transcript levels of apoptotic genes were also significantly lower in
all treatment groups than the control (p<0.05). In the
blastocyst stage, decreased expression of ER stress-, oxidative stress-, and
apoptosis-related genes were observed only in some treatments. However, the
blastocyst formation rates in TUDCA and Vit. C treatment groups (24.8%
and 22.0%, respectively) and mean blastocyst cell number in all treatment
groups (59.7±4.3 to 63.5±3.3) were significantly higher
(p<0.05) than those of control. The results showed
that the TUDCA or Vit. C treatment during micromanipulation inhibited both ER
and oxidative stresses in the early stage of SCNT embryos, thereby reducing cell
damage and promoting in vitro development.
Collapse
Affiliation(s)
- Hye-Bin Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Yeo-Reum Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Mi-Jeong Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Bae-Dong Jung
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Choon-Keun Park
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Hee-Tae Cheong
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
39
|
Peroxiredoxin 5 deficiency exacerbates iron overload-induced neuronal death via ER-mediated mitochondrial fission in mouse hippocampus. Cell Death Dis 2020; 11:204. [PMID: 32205843 PMCID: PMC7090063 DOI: 10.1038/s41419-020-2402-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 01/01/2023]
Abstract
Iron is an essential element for cellular functions, including those of neuronal cells. However, an imbalance of iron homeostasis, such as iron overload, has been observed in several neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. Iron overload causes neuronal toxicity through mitochondrial fission, dysregulation of Ca2+, ER-stress, and ROS production. Nevertheless, the precise mechanisms between iron-induced oxidative stress and iron toxicity related to mitochondria and endoplasmic reticulum (ER) in vivo are not fully understood. Here, we demonstrate the role of peroxiredoxin 5 (Prx5) in iron overload-induced neurotoxicity using Prx5-deficient mice. Iron concentrations and ROS levels in mice fed a high iron diet were significantly higher in Prx5−/− mice than wildtype (WT) mice. Prx5 deficiency also exacerbated ER-stress and ER-mediated mitochondrial fission via Ca2+/calcineurin-mediated dephosphorylation of Drp1 at Serine 637. Moreover, immunoreactive levels of cleaved caspase3 in the CA3 region of the hippocampus were higher in iron-loaded Prx5−/− mice than WT mice. Furthermore, treatment with N-acetyl-cysteine, a reactive oxygen species (ROS) scavenger, attenuated iron overload-induced hippocampal damage by inhibiting ROS production, ER-stress, and mitochondrial fission in iron-loaded Prx5−/− mice. Therefore, we suggest that iron overload-induced oxidative stress and ER-mediated mitochondrial fission may be essential for understanding iron-mediated neuronal cell death in the hippocampus and that Prx5 may be useful as a novel therapeutic target in the treatment of iron overload-mediated diseases and neurodegenerative diseases.
Collapse
|
40
|
Bursch F, Kalmbach N, Naujock M, Staege S, Eggenschwiler R, Abo-Rady M, Japtok J, Guo W, Hensel N, Reinhardt P, Boeckers TM, Cantz T, Sterneckert J, Van Den Bosch L, Hermann A, Petri S, Wegner F. Altered calcium dynamics and glutamate receptor properties in iPSC-derived motor neurons from ALS patients with C9orf72, FUS, SOD1 or TDP43 mutations. Hum Mol Genet 2020; 28:2835-2850. [PMID: 31108504 DOI: 10.1093/hmg/ddz107] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
The fatal neurodegenerative disease amyotrophic lateral sclerosis (ALS) is characterized by a profound loss of motor neurons (MNs). Until now only riluzole minimally extends life expectancy in ALS, presumably by inhibiting glutamatergic neurotransmission and calcium overload of MNs. Therefore, the aim of this study was to investigate the glutamate receptor properties and key aspects of intracellular calcium dynamics in induced pluripotent stem cell (iPSC)-derived MNs from ALS patients with C9orf72 (n = 4 cell lines), fused in sarcoma (FUS) (n = 9), superoxide dismutase 1 (SOD1) (n = 3) or transactive response DNA-binding protein 43 (TDP43) (n = 3) mutations as well as healthy (n = 7 cell lines) and isogenic controls (n = 3). Using calcium imaging, we most frequently observed spontaneous transients in mutant C9orf72 MNs. Basal intracellular calcium levels and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)-induced signal amplitudes were elevated in mutant TDP43 MNs. Besides, a majority of mutant TDP43 MNs responded to 3.5-dihydroxyphenylglycine as metabotropic glutamate receptor agonist. Quantitative real-time PCR demonstrated significantly increased expression levels of AMPA and kainate receptors in mutant FUS cells compared to healthy and isogenic controls. Furthermore, the expression of kainate receptors and voltage gated calcium channels in mutant C9orf72 MNs as well as metabotropic glutamate receptors in mutant SOD1 cells was markedly elevated compared to controls. Our data of iPSC-derived MNs from familial ALS patients revealed several mutation-specific alterations in glutamate receptor properties and calcium dynamics that could play a role in ALS pathogenesis and may lead to future translational strategies with individual stratification of neuroprotective ALS treatments.
Collapse
Affiliation(s)
- Franziska Bursch
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Norman Kalmbach
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Maximilian Naujock
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany
| | - Selma Staege
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Reto Eggenschwiler
- Research Group Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany
| | | | - Julia Japtok
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Wenting Guo
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, BE-3000 Leuven, Belgium.,Laboratory of Neurobiology, VIB-Center for Brain & Disease Research, BE-3000 Leuven, Belgium
| | - Niko Hensel
- Institute of Neuroanatomy, Hannover Medical School, 30625 Hanover, Germany
| | | | - Tobias M Boeckers
- Institute of Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Tobias Cantz
- Research Group Translational Hepatology and Stem Cell Biology, Cluster of Excellence REBIRTH, Hannover Medical School, 30625 Hannover, Germany
| | | | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, BE-3000 Leuven, Belgium.,Laboratory of Neurobiology, VIB-Center for Brain & Disease Research, BE-3000 Leuven, Belgium
| | - Andreas Hermann
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| |
Collapse
|
41
|
Kim SH, Kwon D, Lee S, Son SW, Kwon JT, Kim PJ, Lee YH, Jung YS. Concentration- and Time-Dependent Effects of Benzalkonium Chloride in Human Lung Epithelial Cells: Necrosis, Apoptosis, or Epithelial Mesenchymal Transition. TOXICS 2020; 8:toxics8010017. [PMID: 32121658 PMCID: PMC7151738 DOI: 10.3390/toxics8010017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 02/06/2023]
Abstract
Benzalkonium chloride (BAC), an antimicrobial agent in inhalable medications and household sprays, has been reported to be toxic to pulmonary organs. Although cell membrane damage has been considered as the main cytotoxic mechanism of BAC, its concentration- and time-dependent cellular effects on lung epithelium have not been fully understood. In the present study, human lung epithelial (H358) cells were exposed to 0.2–40 μg/mL of BAC for 30 min or 21 days. Cell membranes were rapidly disrupted by 30 min exposure, but 24 h incubation of BAC (4–40 μg/mL) predominantly caused apoptosis rather than necrosis. BAC (2–4 μg/mL) induced mitochondrial depolarization, which may be associated with increased expression of pro-apoptotic proteins (caspase-3, PARP, Bax, p53, and p21), and decreased levels of the anti-apoptotic protein Bcl-2. The protein expression levels of IRE1α, BiP, CHOP, and p-JNK were also elevated by BAC (2–4 μg/mL) suggesting the possible involvement of endoplasmic reticulum stress in inducing apoptosis. Long-term (7–21 days) incubation with BAC (0.2–0.6 μg/mL) did not affect cell viability but led to epithelial-mesenchymal transition (EMT) as shown by the decrease of E-cadherin and the increase of N-cadherin, fibronectin, and vimentin, caused by the upregulation of EMT transcription factors, such as Snail, Slug, Twist1, Zeb1, and Zeb2. Therefore, we conclude that apoptosis could be an important mechanism of acute BAC cytotoxicity in lung epithelial cells, and chronic exposure to BAC even at sub-lethal doses can promote pulmonary EMT.
Collapse
Affiliation(s)
- Sou Hyun Kim
- Lab of Molecular Toxicology, College of Pharmacy, Pusan National University, Busan 46241, Korea
| | - Doyoung Kwon
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Seunghyun Lee
- Lab of Molecular Toxicology, College of Pharmacy, Pusan National University, Busan 46241, Korea
| | - Seung Won Son
- Lab of Molecular Toxicology, College of Pharmacy, Pusan National University, Busan 46241, Korea
| | - Jung-Taek Kwon
- Risk Assessment Division, Environmental Health Research Department, National Institute of Environmental Research, Incheon 22689, Korea
| | - Pil-Je Kim
- Risk Assessment Division, Environmental Health Research Department, National Institute of Environmental Research, Incheon 22689, Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
- Correspondence: (Y.-H.L.); (Y.-S.J.); Tel.: +82-2-880-2139 (Y.-H.L.); 82-51-510-2816 (Y.-S.J.)
| | - Young-Suk Jung
- Lab of Molecular Toxicology, College of Pharmacy, Pusan National University, Busan 46241, Korea
- Correspondence: (Y.-H.L.); (Y.-S.J.); Tel.: +82-2-880-2139 (Y.-H.L.); 82-51-510-2816 (Y.-S.J.)
| |
Collapse
|
42
|
Weaning Mice and Adult Mice Exhibit Differential Carbon Tetrachloride-Induced Acute Hepatotoxicity. Antioxidants (Basel) 2020; 9:antiox9030201. [PMID: 32121508 PMCID: PMC7139353 DOI: 10.3390/antiox9030201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/13/2020] [Accepted: 02/16/2020] [Indexed: 02/06/2023] Open
Abstract
Age is a risk factor for drug-induced liver injury (DILI). However, there is a limited understanding of pediatric DILI. Here, 2-week-old weaning and 8-week-old adult male ICR mice were intraperitoneally injected with CCl4 (0.1 mmol/kg equal to 15.4 mg/kg) to comparatively evaluate the time-dependent liver damage and cellular events. CCl4 significantly enhanced the serum alanine aminotransferase/aspartate aminotransferase levels and hepatic centrilobular necrosis in the weaning mice, whereas it induced mild liver injury in the adult mice. CCl4-treated weaning mice exhibited higher hepatic levels of pro-apoptotic proteins (Bax, cleaved caspase-3, -7, and -9), activated MAPKs (p-JNK and p-Erk), and endoplasmic reticulum stress indicators (ATF6 and CHOP) and lower hepatic anti-apoptotic Bcl-2 levels than the adult mice. The weaning mice exhibited enhanced basal hepatic glutathione (GSH) levels due to high glutamate cysteine ligase (GCL) and low anti-cysteine dioxygenase (CDO) enzyme levels. However, CCl4 markedly reduced the hepatic GSH levels only in the weaning mice. Furthermore, higher hepatic levels of oxidative stress-induced malondialdehyde, 4-hydroxynonenal, nitrotyrosine-protein adducts, and oxidized proteins were observed in CCl4-treated weaning mice than in CCl4-treated adult mice. The enhanced levels of hepatic cytochrome P450 (CYP) 2E1 and CYP3A, and decreased hepatic GSH S-transferase (GST)-π and GSH reductase (GR) levels in the weaning mice may contribute to their enhanced susceptibility to liver damage.
Collapse
|
43
|
Eser Faki H, Tras B, Uney K. Alpha lipoic acid and vitamin E improve atorvastatin-induced mitochondrial dysfunctions in rats. Mitochondrion 2020; 52:83-88. [PMID: 32119925 DOI: 10.1016/j.mito.2020.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/12/2019] [Accepted: 02/27/2020] [Indexed: 01/01/2023]
Abstract
To determine the effects of alpha lipoic acid (ALA) and vitamin E (Vit E) on mitochondrial dysfunction caused by statins. A total of 38 Wistar Albino rats were used in this study. The control group received dimethyl sulfoxide. The atorvastatin (A) group received atorvastatin (10 mg/kg). The A + ALA group received atorvastatin (10 mg/kg) and ALA (100 mg/kg). The A + Vit E group was administered atorvastatin (10 mg/kg) and Vit E (100 mg/kg). The A + ALA + Vit E group was administered atorvastatin (10 mg/kg), ALA (100 mg/kg) and Vit E (100 mg/kg). All applications were administered simultaneously by gavage for 20 days. ATP level and complex I activity were measured from liver, muscle, heart, kidney and brain. Atorvastatin significantly decreased the ATP levels in heart and kidney, while a slight decrease was seen in liver, muscle and brain. Atorvastatin caused an insignificant decrease in the complex I activity in all tissues examined. ALA administration significantly improved the ATP levels in the liver, heart and kidney, while Vit E improved the ATP levels in all tissues except the muscle compared to Atorvastatin group. Single administration of both ALA and vit E ameliorated complex I activity in the muscle, heart, kidney and brain. The combination of ALA and Vit E significantly improved the ATP levels in the liver, heart, kidney and brain and also provided significant improvements the complex I activity in all tissues. The undesirable effects of Atorvastatin on mitochondrial functions in this study ameliorated by using ALA and/or Vit E alone and in combination.
Collapse
Affiliation(s)
- Hatice Eser Faki
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031 Konya, Turkey.
| | - Bunyamin Tras
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031 Konya, Turkey
| | - Kamil Uney
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, 42031 Konya, Turkey
| |
Collapse
|
44
|
Delmotte P, Sieck GC. Endoplasmic Reticulum Stress and Mitochondrial Function in Airway Smooth Muscle. Front Cell Dev Biol 2020; 7:374. [PMID: 32010691 PMCID: PMC6974519 DOI: 10.3389/fcell.2019.00374] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/16/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammatory airway diseases such as asthma affect more than 300 million people world-wide. Inflammation triggers pathophysiology via such as tumor necrosis factor α (TNFα) and interleukins (e.g., IL-13). Hypercontraction of airway smooth muscle (ASM) and ASM cell proliferation are major contributors to the exaggerated airway narrowing that occurs during agonist stimulation. An emergent theme in this context is the role of inflammation-induced endoplasmic reticulum (ER) stress and altered mitochondrial function including an increase in the formation of reactive oxygen species (ROS). This may establish a vicious cycle as excess ROS generation leads to further ER stress. Yet, it is unclear whether inflammation-induced ROS is the major mechanism leading to ER stress or the consequence of ER stress. In various diseases, inflammation leads to an increase in mitochondrial fission (fragmentation), associated with reduced levels of mitochondrial fusion proteins, such as mitofusin 2 (Mfn2). Mitochondrial fragmentation may be a homeostatic response since it is generally coupled with mitochondrial biogenesis and mitochondrial volume density thereby reducing demand on individual mitochondrion. ER stress is triggered by the accumulation of unfolded proteins, which induces a homeostatic response to alter protein balance via effects on protein synthesis and degradation. In addition, the ER stress response promotes protein folding via increased expression of molecular chaperone proteins. Reduced Mfn2 and altered mitochondrial dynamics may not only be downstream to ER stress but also upstream such that a reduction in Mfn2 triggers further ER stress. In this review, we summarize the current understanding of the link between inflammation-induced ER stress and mitochondrial function and the role played in the pathophysiology of inflammatory airway diseases.
Collapse
Affiliation(s)
- Philippe Delmotte
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
45
|
Yap J, Chen X, Delmotte P, Sieck GC. TNFα selectively activates the IRE1α/XBP1 endoplasmic reticulum stress pathway in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2020; 318:L483-L493. [PMID: 31940218 DOI: 10.1152/ajplung.00212.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Airway inflammation is a key aspect of diseases such as asthma. Proinflammatory cytokines such as TNFα mediate the inflammatory response. In various diseases, inflammation leads to endoplasmic reticulum (ER) stress, the accumulation of unfolded proteins, which triggers homeostatic responses to restore normal cellular function. We hypothesized that TNFα triggers ER stress through an increase in reactive oxygen species generation in human airway smooth muscle (hASM) with a downstream effect on mitofusin 2 (Mfn2). In hASM cells isolated from lung specimens incidental to patient surgery, dose- and time-dependent effects of TNFα exposure were assessed. Exposure of hASM to tunicamycin was used as a positive control. Tempol (500 μM) was used as superoxide scavenger. Activation of three ER stress pathways were evaluated by Western blotting: 1) autophosphorylation of inositol-requiring enzyme1 (IRE1α) leading to splicing of X-box binding protein 1 (XBP1); 2) autophosphorylation of protein kinase RNA-like endoplasmic reticulum kinase (PERK) leading to phosphorylation of eukaryotic initiation factor 2α; and 3) translocation and cleavage of activating transcription factor 6 (ATF6). We found that exposure of hASM cells to tunicamycin activated all three ER stress pathways. In contrast, TNFα selectively activated the IRE1α/XBP1 pathway in a dose- and time-dependent fashion. Our results indicate that TNFα does not activate the PERK and ATF6 pathways. Exposure of hASM cells to TNFα also decreased Mfn2 protein expression. Concurrent exposure to TNFα and tempol reversed the effect of TNFα on IRE1α phosphorylation and Mfn2 protein expression. Selective activation of the IRE1α/XBP1 pathway in hASM cells after exposure to TNFα may reflect a unique homeostatic role of this pathway in the inflammatory response of hASM cells.
Collapse
Affiliation(s)
- John Yap
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Xujiao Chen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Philippe Delmotte
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
46
|
Ide Y, Horie T, Saito N, Watanabe S, Otani C, Miyasaka Y, Kuwabara Y, Nishino T, Nakao T, Nishiga M, Nishi H, Nakashima Y, Nakazeki F, Koyama S, Kimura M, Tsuji S, Rodriguez RR, Xu S, Yamasaki T, Watanabe T, Yamamoto M, Yanagita M, Kimura T, Kakizuka A, Ono K. Cardioprotective Effects of VCP Modulator KUS121 in Murine and Porcine Models of Myocardial Infarction. JACC Basic Transl Sci 2019; 4:701-714. [PMID: 31709319 PMCID: PMC6834964 DOI: 10.1016/j.jacbts.2019.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 12/23/2022]
Abstract
No effective treatment is yet available to reduce infarct size and improve clinical outcomes after acute myocardial infarction by enhancing early reperfusion therapy using primary percutaneous coronary intervention. The study showed that Kyoto University Substance 121 (KUS121) reduced endoplasmic reticulum stress, maintained adenosine triphosphate levels, and ameliorated the infarct size in a murine cardiac ischemia and reperfusion injury model. The study confirmed the cardioprotective effect of KUS121 in a porcine ischemia and reperfusion injury model. These findings confirmed that KUS121 is a promising novel therapeutic agent for myocardial infarction in conjunction with primary percutaneous coronary intervention.
Collapse
Key Words
- AAR, area at risk
- ATP
- ATP, adenosine triphosphate
- ATPase, adenosine triphosphatase
- BiP, immunoglobulin heavy chain-binding protein
- CHOP, C/EBP homologous protein
- CMR, cardiac magnetic resonance
- EF, ejection fraction
- ER stress
- ER, endoplasmic reticulum
- FRET, fluorescence resonance energy transfer
- FS, fractional shortening
- H2O2, hydrogen peroxide
- HF, heart failure
- I/R, ischemia and reperfusion
- IBMPFD, inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia
- IHD, ischemic heart disease
- KUS121
- KUS121, Kyoto University Substance 121
- LAD, left anterior descending artery
- LV, left ventricular/ventricle
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- TTC, triphenyltetrazolium chloride
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick-end labeling
- VCP, valosin-containing protein
- myocardial infarction
Collapse
Affiliation(s)
- Yuya Ide
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahiro Horie
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naritatsu Saito
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shin Watanabe
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chiharu Otani
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yui Miyasaka
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuhide Kuwabara
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomohiro Nishino
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tetsushi Nakao
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Nishiga
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hitoo Nishi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuhiro Nakashima
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumiko Nakazeki
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoshi Koyama
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuhei Tsuji
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Randolph Ruiz Rodriguez
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sijia Xu
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomohiro Yamasaki
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshimitsu Watanabe
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masamichi Yamamoto
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akira Kakizuka
- Laboratory of Functional Biology, Kyoto University Graduate School of Biostudies and Solution Oriented Research for Science and Technology, Kyoto, Japan
| | - Koh Ono
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
47
|
Lebeau J, Saunders JM, Moraes VWR, Madhavan A, Madrazo N, Anthony MC, Wiseman RL. The PERK Arm of the Unfolded Protein Response Regulates Mitochondrial Morphology during Acute Endoplasmic Reticulum Stress. Cell Rep 2019. [PMID: 29539413 PMCID: PMC5870888 DOI: 10.1016/j.celrep.2018.02.055] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is transmitted to mitochondria and is associated with pathologic mitochondrial dysfunction in diverse diseases. The PERK arm of the unfolded protein response (UPR) protects mitochondria during ER stress through the transcriptional and translational remodeling of mitochondrial molecular quality control pathways. Here, we show that ER stress also induces dynamic remodeling of mitochondrial morphology by promoting protective stress-induced mitochondrial hyperfusion (SIMH). ER-stress-associated SIMH is regulated by the PERK arm of the UPR and activated by eIF2α phosphorylation-dependent translation attenuation. We show that PERK-regulated SIMH is a protective mechanism to prevent pathologic mitochondrial fragmentation and promote mitochondrial metabolism in response to ER stress. These results identify PERK-dependent SIMH as a protective stress-responsive mechanism that regulates mitochondrial morphology during ER stress. Furthermore, our results show that PERK integrates transcriptional and translational signaling to coordinate mitochondrial molecular and organellar quality control in response to pathologic ER insults.
Collapse
Affiliation(s)
- Justine Lebeau
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jaclyn M Saunders
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vivian W R Moraes
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Aparajita Madhavan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicole Madrazo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mary C Anthony
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
48
|
Kim SH, Kwon D, Lee S, Ki SH, Jeong HG, Hong JT, Lee YH, Jung YS. Polyhexamethyleneguanidine Phosphate-Induced Cytotoxicity in Liver Cells Is Alleviated by Tauroursodeoxycholic Acid (TUDCA) via a Reduction in Endoplasmic Reticulum Stress. Cells 2019; 8:cells8091023. [PMID: 31484321 PMCID: PMC6770470 DOI: 10.3390/cells8091023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/26/2019] [Accepted: 08/30/2019] [Indexed: 02/06/2023] Open
Abstract
Polyhexamethyleneguanidine phosphate (PHMG-P) is a widely used polymeric antimicrobial agent known to induce significant pulmonary toxicity. Several studies have reported that the liver also can be a target organ of polyhexamethyleneguanidine (PHMG) toxicity, but the exact effect of this compound on liver cells is not well understood. To identify the mechanism of PHMG hepatotoxicity, HepG2 cells were exposed to PHMG-P for 72 h. The cell viability was significantly decreased by PHMG-P in a time- and concentration-dependent manner. The mitochondrial membrane potential was markedly reduced by PHMG-P and the apoptotic signaling cascade was activated. The increases observed in C/EBP homologous protein (CHOP), p-IRE, and p-JNK levels in PHMG-P-treated cells indicated the induction of endoplasmic reticulum stress. To verify the role of ER stress in PHMG-P-induced cytotoxicity, HepG2 cells were pretreated with the chemical chaperone, tauroursodeoxycholic acid (TUDCA) and then co-treated with TUDCA and PHMG-P for 24 h. Interestingly, TUDCA inhibited PHMG-P-induced ER stress and cytotoxicity in a dose-dependent manner. The apoptotic cell death and mitochondrial depolarization were also prevented by TUDCA. The proteins involved in the apoptotic pathway were all normalized to their control levels in TUDCA-treated cells. In conclusion, the results suggest that PHMG-P induced significant cytotoxicity in liver cells and ER stress-mediated apoptosis, which may be an important mechanism mediating this hepatotoxicity.
Collapse
Affiliation(s)
- Sou Hyun Kim
- Lab of Molecular Toxicology, College of Pharmacy, Pusan National University, Busan 46241, Korea.
| | - Doyoung Kwon
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158-2280, USA.
| | - Seunghyun Lee
- Lab of Molecular Toxicology, College of Pharmacy, Pusan National University, Busan 46241, Korea.
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju 61452, Korea.
| | - Hye Gwang Jeong
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea.
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea.
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.
| | - Young-Suk Jung
- Lab of Molecular Toxicology, College of Pharmacy, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
49
|
Lanzillotta C, Tramutola A, Meier S, Schmitt F, Barone E, Perluigi M, Di Domenico F, Abisambra JF. Early and Selective Activation and Subsequent Alterations to the Unfolded Protein Response in Down Syndrome Mouse Models. J Alzheimers Dis 2019; 62:347-359. [PMID: 29439332 DOI: 10.3233/jad-170617] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Down syndrome (DS) is the most common chromosomal disorder and the leading genetic cause of intellectual disability in humans, which results from the triplication of chromosome 21. DS individuals have an increased risk of developing Alzheimer's disease (AD)-like pathology and dementia by the age of 40 due to the triplication of several genes involved in the formation of amyloid plaques and tau tangles. Further, DS and AD are characterized by the aberrant accumulation of unfolded/misfolded proteins resulting from over-burdened protein quality control systems. The accumulation of misfolded proteins in the endoplasmic reticulum (ER) triggers a cellular stress response called the unfolded protein response (UPR). Long-term activation of the UPR mediates neuronal dysfunction in AD. We hypothesized that the UPR is impacted in a mouse model of DS. To test this, we performed gene and protein expression analysis of ER stress markers in the Ts65Dn mouse model of DS at 3, 9, and 18 months. We identified activation of the PERK pathway in Ts65Dn DS mice at 3 months of age compared to euploid controls. We also determined that the early and overt UPR activation decreased with age, the UPR signal was significantly reduced by 18 months. Our data suggest that UPR activation in DS mouse models occurs early before consistent brain neurodegeneration and might be an essential contributor to dys-proteostasis.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy.,Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Shelby Meier
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Frederick Schmitt
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA.,Epilepsy Center (EpiC) and Spinal Cord and Brain Injury Research Center (SCoBIRC), College of Medicine, University of Kentucky, Lexington, KY, USA.,Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy.,Universidad Autónoma de Chile, Instituto de Ciencias Biomédicas, Facultad de alud, Avenida Pedro de Valdivia 425, Providencia, Santiago, Chile
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Jose F Abisambra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA.,Epilepsy Center (EpiC) and Spinal Cord and Brain Injury Research Center (SCoBIRC), College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
50
|
Zhang X, Shao S, Zhao L, Yang R, Zhao M, Fang L, Li M, Chen W, Song Y, Xu C, Zhou X, Zhao J, Gao L. ER stress contributes to high-fat diet-induced decrease of thyroglobulin and hypothyroidism. Am J Physiol Endocrinol Metab 2019; 316:E510-E518. [PMID: 30620634 DOI: 10.1152/ajpendo.00194.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Recent studies revealed the emerging role of excess uptake of lipids in the development of hypothyroidism. However, the underlying mechanism is largely unknown. We investigated the effect of high-fat diet (HFD) on thyroid function and the role of endoplasmic reticulum (ER) in HFD-induced hypothyroidism. Male Sprague-Dawley rats were fed with HFD or control diet for 18 wk. HFD rats showed an impaired thyroid function, with decreased thyroglobulin (Tg) level. We found the ER stress was triggered in HFD rat thyroid glands and palmitate-treated thyrocytes. Luminal swelling of ER in thyroid epithelial cells of HFD rats was also observed. The rate of Tg degradation increased in palmitate-treated thyrocytes. In addition, applying 4-phenyl butyric acid to alleviate ER stress in HFD rats improved the decrease of Tg and thyroid function. Withdrawal of the HFD improved thyroid function . In conclusion, we demonstrate that ER stress mediates the HFD-induced hypothyroidism, probably by impairing the production of Tg, and attenuation of ER stress improves thyroid function. Our study provides the understanding of how HFD induces hypothyroidism.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Shanshan Shao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Lifang Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Rui Yang
- Experimental Animal Center, Shandong Provincial Hospital Affiliated to Shandong University, Ji-nan, Shandong , China
| | - Meng Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Li Fang
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Mengzhu Li
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Wenbin Chen
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Ji-nan, Shandong , China
| | - Yongfeng Song
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Chao Xu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Xiaoming Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University , Shandong , China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
| | - Ling Gao
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Ji-nan, Shandong , China
- Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Ji-nan, Shandong , China
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Ji-nan, Shandong , China
| |
Collapse
|