51
|
Dong Y, Lee Y, Cui K, He M, Wang B, Bhattacharjee S, Zhu B, Yago T, Zhang K, Deng L, Ouyang K, Wen A, Cowan DB, Song K, Yu L, Brophy ML, Liu X, Wylie-Sears J, Wu H, Wong S, Cui G, Kawashima Y, Matsumoto H, Kodera Y, Wojcikiewicz RJH, Srivastava S, Bischoff J, Wang DZ, Ley K, Chen H. Epsin-mediated degradation of IP3R1 fuels atherosclerosis. Nat Commun 2020; 11:3984. [PMID: 32770009 PMCID: PMC7414107 DOI: 10.1038/s41467-020-17848-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 07/15/2020] [Indexed: 12/18/2022] Open
Abstract
The epsin family of endocytic adapter proteins are widely expressed, and interact with both proteins and lipids to regulate a variety of cell functions. However, the role of epsins in atherosclerosis is poorly understood. Here, we show that deletion of endothelial epsin proteins reduces inflammation and attenuates atherosclerosis using both cell culture and mouse models of this disease. In atherogenic cholesterol-treated murine aortic endothelial cells, epsins interact with the ubiquitinated endoplasmic reticulum protein inositol 1,4,5-trisphosphate receptor type 1 (IP3R1), which triggers proteasomal degradation of this calcium release channel. Epsins potentiate its degradation via this interaction. Genetic reduction of endothelial IP3R1 accelerates atherosclerosis, whereas deletion of endothelial epsins stabilizes IP3R1 and mitigates inflammation. Reduction of IP3R1 in epsin-deficient mice restores atherosclerotic progression. Taken together, epsin-mediated degradation of IP3R1 represents a previously undiscovered biological role for epsin proteins and may provide new therapeutic targets for the treatment of atherosclerosis and other diseases. Endothelial cell (EC) dysfunction and inflammation contribute to plaque destabilization in atherosclerosis, increasing the risk of thrombotic events. Here, the authors show that epsin promotes EC inflammation via a mechanism involving IP3R1 degradation, and that deletion of epsin in the endothelium prevents EC dysfunctoin and atherosclerosis in mice.
Collapse
Affiliation(s)
- Yunzhou Dong
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yang Lee
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kui Cui
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ming He
- Department of Medicine, University of California, San Diego, San Diego, CA, 92093, USA
| | - Beibei Wang
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sudarshan Bhattacharjee
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Tadayuki Yago
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Kun Zhang
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lin Deng
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Kunfu Ouyang
- Department of Medicine, University of California, San Diego, San Diego, CA, 92093, USA
| | - Aiyun Wen
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Douglas B Cowan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kai Song
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lili Yu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Megan L Brophy
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaolei Liu
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jill Wylie-Sears
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Guanglin Cui
- Department of Nutrition and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Yusuke Kawashima
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.,Center for Disease Proteomics, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Hiroyuki Matsumoto
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yoshio Kodera
- Center for Disease Proteomics, Kitasato University School of Science, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | | | - Sanjay Srivastava
- Department of Medicine, Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Joyce Bischoff
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Klaus Ley
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
52
|
Perrotta I. The microscopic anatomy of endothelial cells in human atherosclerosis: Focus on ER and mitochondria. J Anat 2020; 237:1015-1025. [PMID: 32735733 DOI: 10.1111/joa.13281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022] Open
Abstract
Once regarded merely as a bland lipid storage disease consequence of aging, atherosclerosis is currently considered a slow and continuous inflammatory process (partially controllable by treatment) with complex etiology involving a multitude of genetic and environmental risk factors which ultimately result in the formation of the plaque. The vascular endothelium, a monolayer of endothelial cells (ECs), is an important regulatory "organ" critical for cardiovascular homeostasis in health which also contributes significantly to the pathomechanisms of several disease states, including atherosclerosis. Over the years, there has been evidence highlighting the central role of endoplasmic reticulum (ER) in the maintenance of endothelial function and perturbations in ER biology have been proposed to adversely affect a diverse range of endothelial functions. Of particular interest is the evidence that under certain pathophysiological circumstances, abnormal ER ultrastructure correlates with altered ER function and signaling and can contribute to cell injury and apoptosis. Therefore, the ultrastructural traits of ER membranes can have important implications not only for their functional bearings but also for the etiology and pathophysiology of diverse human disorders. With regard to atherosclerosis, the focus of ER research has been centered on the molecular signals originated from the ER to manage conditions of stress, leaving the fine structure of this organelle an almost unexplored (but promising) area of studies. There is, also, increasing evidence that mitochondrial dysfunction plays a critical role in promoting cell apoptosis, inflammation, and oxidative stress, thereby contributing to atheroma growth. It is within this context that the present study has been undertaken to investigate the microscopic architecture of ECs in human atherosclerosis and to determine whether the potential structural abnormalities of ER and mitochondria may play a central pathogenic role in atherogenesis or may merely reflect the condition of a tissue whose integrity has already been disturbed or destroyed. For this purpose, transmission electron microscopy (TEM) remains a powerful technique that can not only provide information about the ultrastructural state of cell organelles but also allow the correlation between different subcellular alterations indicative of a certain pathophysiological condition and cellular response. The present study expands the spectrum of ultrastructural defects known to exist in human atherosclerosis and suggests that ER alterations may be of great importance in the pathogenesis of the disease. The architectural changes of ER may be considered early pathological events that precede any overt histologic abnormalities in the vascular endothelium and its subcellular organelles, primarily the mitochondrial pool.
Collapse
Affiliation(s)
- Ida Perrotta
- Centre for Microscopy and Microanalysis, Transmission Electron Microscopy Laboratory, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
53
|
Ito H, Yamashita Y, Tanaka T, Takaki M, Le MN, Yoshida LM, Morimoto K. Cigarette smoke induces endoplasmic reticulum stress and suppresses efferocytosis through the activation of RhoA. Sci Rep 2020; 10:12620. [PMID: 32724133 PMCID: PMC7387437 DOI: 10.1038/s41598-020-69610-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 07/15/2020] [Indexed: 12/22/2022] Open
Abstract
Impaired efferocytosis is a key mechanism of inflammatory lung diseases, including chronic obstructive pulmonary disease and cystic fibrosis. Cigarette smoking activates RhoA and impairs efferocytosis in alveolar macrophages, but the mechanism has not been fully elucidated. We investigated the role of endoplasmic reticulum (ER) stress induced by cigarette smoking in the disruption of efferocytosis. Both tunicamycin (10 μg/ml) and thapsigargin (0.1 and 1 μM), which are ER stress inducers, suppressed efferocytosis in J774 cells, and a Rho-associated coiled-coil-forming kinase (ROCK) inhibitor (Y27632) reversed this effect. We validated the effect of tunicamycin on efferocytosis in experiments using RAW264.7 cells. Then, we investigated the role of the unfolded protein response (UPR) in efferocytosis impaired by ER stress. A PERK inhibitor (GSK2606414) restored the efferocytosis that had been impaired by TM, and an eIF2α dephosphorylation inhibitor (salubrinal) suppressed efferocytosis. Cigarette smoke extract (CSE) induced ER stress in J774 macrophages and RhoA activation in J774 cells, and the CSE-induced ROCK activity was successfully reversed by GSK2606414 and tauroursodeoxycholic acid. Finally, we confirmed that ER stress suppresses efferocytosis in murine alveolar macrophages and that GSK2606414 could rescue this process. These data suggest that cigarette smoke-induced ER stress and the UPR play crucial roles in RhoA activation and suppression of efferocytosis in the lung.
Collapse
Affiliation(s)
- Hiroyuki Ito
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan.,Department of Clinical Tropical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshiro Yamashita
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan
| | - Takeshi Tanaka
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan
| | - Masahiro Takaki
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan
| | - Minh Nhat Le
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Lay-Myint Yoshida
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Konosuke Morimoto
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki City, Nagasaki, 852-8523, Japan. .,Department of Clinical Tropical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| |
Collapse
|
54
|
Sukhorukov VN, Khotina VA, Bagheri Ekta M, Ivanova EA, Sobenin IA, Orekhov AN. Endoplasmic Reticulum Stress in Macrophages: The Vicious Circle of Lipid Accumulation and Pro-Inflammatory Response. Biomedicines 2020; 8:biomedicines8070210. [PMID: 32668733 PMCID: PMC7400097 DOI: 10.3390/biomedicines8070210] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/08/2023] Open
Abstract
The endoplasmic reticulum (ER) stress is an important event in the pathogenesis of different human disorders, including atherosclerosis. ER stress leads to disturbance of cellular homeostasis, apoptosis, and in the case of macrophages, to foam cell formation and pro-inflammatory cytokines production. In atherosclerosis, several cell types can be affected by ER stress, including endothelial cells, vascular smooth muscular cells, and macrophages. Modified low-density lipoproteins (LDL) and cytokines, in turn, can provoke ER stress through different processes. The signaling cascades involved in ER stress initiation are complex and linked to other cellular processes, such as lysosomal biogenesis and functioning, autophagy, mitochondrial homeostasis, and energy production. In this review, we discuss the underlying mechanisms of ER stress formation and the interplay of lipid accumulation and pro-inflammatory response. We will specifically focus on macrophages, which are the key players in maintaining chronic inflammatory milieu in atherosclerotic lesions, and also a major source of lipid-accumulating foam cells.
Collapse
Affiliation(s)
- Vasily N. Sukhorukov
- Research Institute of Human Morphology, Laboratory of Cellular and Molecular Pathology of Cardiovascular System, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (M.B.E.); (I.A.S.); (A.N.O.)
- Correspondence: (V.N.S.); (E.A.I.)
| | - Victoria A. Khotina
- Research Institute of Human Morphology, Laboratory of Cellular and Molecular Pathology of Cardiovascular System, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (M.B.E.); (I.A.S.); (A.N.O.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Str., 125315 Moscow, Russia
| | - Mariam Bagheri Ekta
- Research Institute of Human Morphology, Laboratory of Cellular and Molecular Pathology of Cardiovascular System, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (M.B.E.); (I.A.S.); (A.N.O.)
| | - Ekaterina A. Ivanova
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Correspondence: (V.N.S.); (E.A.I.)
| | - Igor A. Sobenin
- Research Institute of Human Morphology, Laboratory of Cellular and Molecular Pathology of Cardiovascular System, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (M.B.E.); (I.A.S.); (A.N.O.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, Institute of Experimental Cardiology, 15-a 3-rd Cherepkovskaya Str., 121552 Moscow, Russia
| | - Alexander N. Orekhov
- Research Institute of Human Morphology, Laboratory of Cellular and Molecular Pathology of Cardiovascular System, 3 Tsyurupy Str., 117418 Moscow, Russia; (V.A.K.); (M.B.E.); (I.A.S.); (A.N.O.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Str., 125315 Moscow, Russia
| |
Collapse
|
55
|
Bozaykut P, Ekren R, Sezerman OU, Gladyshev VN, Ozer NK. High-throughput profiling reveals perturbation of endoplasmic reticulum stress-related genes in atherosclerosis induced by high-cholesterol diet and the protective role of vitamin E. Biofactors 2020; 46:653-664. [PMID: 32384218 DOI: 10.1002/biof.1635] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 12/30/2022]
Abstract
Formation of atherosclerotic plaques, called atherogenesis, is a complex process affected by genetic and environmental factors. It was proposed that endoplasmic reticulum (ER) stress is an important factor in the pathogenesis of atherosclerosis and that vitamin E affects atherosclerotic plaque formation via its antioxidant properties. Here, we investigated ER stress-related molecular mechanisms in high-cholesterol diet (HCD, 2%)-induced atherosclerosis model and the role of vitamin E supplementation in it, beyond its antioxidant properties. The consequences of HCD and vitamin E supplementation were examined by determining protein levels of ER stress markers in aortic tissues. As vitamin E supplementation acts on several unfolded protein response (UPR) factors, it decreased ER stress induced by HCD. To elucidate the associated pathways, gene expression profiling was performed, revealing differentially expressed genes enriched in ER stress-related pathways such as the proteasome and the apoptosis pathways. We further assessed the proteasomal activity impaired by HCD in the aorta and showed that vitamin E reversed it to that of control animals. Overall, the study characterized the effects of HCD and vitamin E on ER stress-related gene expression, revealing the role of proteolytic systems during atherogenesis.
Collapse
Affiliation(s)
- Perinur Bozaykut
- Department of Biochemistry, Faculty of Medicine, Marmara University, Istanbul, Turkey
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ruchan Ekren
- Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Osman Ugur Sezerman
- Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Nesrin Kartal Ozer
- Department of Biochemistry, Faculty of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
56
|
Rana SVS. Endoplasmic Reticulum Stress Induced by Toxic Elements-a Review of Recent Developments. Biol Trace Elem Res 2020; 196:10-19. [PMID: 31686395 DOI: 10.1007/s12011-019-01903-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum of all eukaryotic cells is a membrane-bound organelle. Under electron microscope it appears as parallel arrays of "rough membranes" and a maze of "smooth vesicles" respectively. It performs various functions in cell, i.e., synthesis of proteins to degradation of xenobiotics. Bioaccumulation of drugs/chemicals/xenobiotics in the cytosol can trigger ER stress. It is recognized by the accumulation of unfolded or misfolded proteins in the lumen of ER. Present review summarizes the present status of knowledge on ER stress caused by toxic elements, viz arsenic, cadmium, lead, mercury, copper, chromium, and nickel. While inorganic arsenic may induce various glucose-related proteins, i.e., GRP78, GRP94 and CHOP, XBP1, and calpains, cadmium upregulates GRP78. Antioxidants like ascorbic acid, NAC, and Se inhibit the expression of UPR. Exposure to lead also changes ER stress related genes, i.e., GRP 78, GRP 94, ATF4, and ATF6. Mercury too upregulates these genes. Nickel, a carcinogenic element upregulates the expression of Bak, cytochrome C, caspase-3, caspase-9, caspase-12, and GADD 153. Much is not known on ER stress caused by nanoparticles. The review describes inter-organelle association between mitochondria and ER. It also discusses the interdependence between oxidative stress and ER stress. A cross talk amongst different cellular components appears essential to disturb pathways leading to cell death. However, these molecular switches within the signaling network used by toxic elements need to be identified. Nevertheless, ER stress especially caused by toxic elements still remains to be an engaging issue.
Collapse
Affiliation(s)
- S V S Rana
- Department of Toxicology, Ch. Charan Singh University, Meerut, 250 004, India.
| |
Collapse
|
57
|
Sun S, Ji Z, Fu J, Wang XF, Zhang LS. Endosulfan induces endothelial inflammation and dysfunction via IRE1α/NF-κB signaling pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:26163-26171. [PMID: 32361974 DOI: 10.1007/s11356-020-09023-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/22/2020] [Indexed: 05/23/2023]
Abstract
Cardiovascular diseases are related to vascular endothelial cell injury; our previous studies showed that endosulfan could cause hypercoagulation of blood by inducing endothelial cell injury. To clarify the mechanism of it, we treated human umbilical vein endothelial cells (HUVECs) with 0, 1, 5, and 10 μg/mL endosulfan, while in the inhibition groups, reactive oxygen species (ROS) inhibitor N-acetylcysteine (NAC, 3 mmol) and endoplasmic reticulum (ER) stress inhibitor (STF-083010, 10 μmol) were incubated prior to endosulfan. The results showed that endosulfan could induce inflammatory response and dysfunction by increasing the release of inflammatory cytokines such as interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and adhesion molecules such as vascular cell adhesion molecule 1 (VCAM-1) and endothelin-1 (ET-1), and inducing ROS production in HUVECs. We also found that endosulfan could cause ER damage, remarkably increase the expressions of inositol-requiring enzyme 1α (IRE1α), phosphorylated IRE1α (p-IRE1α), GRP78, XBP1, nuclear factor-kappa B (NF-κB), and phosphorylated NF-κB (p-NF-κB) in HUVECs. The presence of NAC antagonized the ROS production, expressions of IRE1α and p-IRE1α; however, STF-083010 could decrease the expression levels of GRP78, XBP1, NF-κB, and p-NF-κB and attenuate IL-1β, IL-6, TNF-α, VCAM-1, and ET-1 release induced by endosulfan. These results demonstrated that endosulfan-induced endothelial inflammation and dysfunction through the IRE1α/NF-κB signaling pathway may be triggered by oxidative stress. The study provided experimental basis for the correlation between environmental pollutants (endosulfan) and cardiovascular diseases.
Collapse
Affiliation(s)
- ShiTian Sun
- College of Clinical Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - ZhengGuo Ji
- Department of Urology, Beijing Friendship Hospital, Capital medical University, Beijing, China
| | - JiaRong Fu
- College of Clinical Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Xi-Feng Wang
- Department of Critical Care Medicine, Yu Huang Ding Hospital, Qingdao University, Yantai, 264000, People's Republic of China.
| | - Lian-Shuang Zhang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| |
Collapse
|
58
|
Golonka RM, Xiao X, Abokor AA, Joe B, Vijay-Kumar M. Altered nutrient status reprograms host inflammation and metabolic health via gut microbiota. J Nutr Biochem 2020; 80:108360. [PMID: 32163821 PMCID: PMC7242157 DOI: 10.1016/j.jnutbio.2020.108360] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 02/07/2023]
Abstract
The metabolism of macro- and micronutrients is a complex and highly regulated biological process. An imbalance in the metabolites and their signaling networks can lead to nonresolving inflammation and consequently to the development of chronic inflammatory-associated diseases. Therefore, identifying the accumulated metabolites and altered pathways during inflammatory disorders would not only serve as "real-time" markers but also help in the development of nutritional therapeutics. In this review, we explore recent research that has delved into elucidating the effects of carbohydrate/calorie restriction, protein malnutrition, lipid emulsions and micronutrient deficiencies on metabolic health and inflammation. Moreover, we describe the integrated stress response in terms of amino acid starvation and lipemia and how this modulates new age diseases such as inflammatory bowel disease and atherosclerosis. Lastly, we explain the latest research on metaflammation and inflammaging. This review focuses on multiple signaling pathways, including, but not limited to, the FGF21-β-hydroxybutryate-NLRP3 axis, the GCN2-eIF2α-ATF4 pathway, the von Hippel-Lindau/hypoxia-inducible transcription factor pathway and the TMAO-PERK-FoxO1 axis. Additionally, throughout the review, we explain how the gut microbiota responds to altered nutrient status and also how antimicrobial peptides generated from nutrient-based signaling pathways can modulate the gut microbiota. Collectively, it must be emphasized that metabolic starvation and inflammation are strongly regulated by both environmental (i.e., nutrition, gut microbiome) and nonenvironmental (i.e., genetics) factors, which can influence the susceptibility to inflammatory disorders.
Collapse
Affiliation(s)
- Rachel M Golonka
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Xia Xiao
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ahmed A Abokor
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Bina Joe
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614
| | - Matam Vijay-Kumar
- UT Microbiome Consortium, Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614.
| |
Collapse
|
59
|
Sun JL, Abd El-Aty AM, Jeong JH, Jung TW. Ginsenoside Rb2 Ameliorates LPS-Induced Inflammation and ER Stress in HUVECs and THP-1 Cells via the AMPK-Mediated Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:967-985. [PMID: 32431178 DOI: 10.1142/s0192415x20500469] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammation and endoplasmic reticulum (ER) stress have been documented to contribute to the development of atherosclerosis. Ginsenoside Rb2 has been reported to exhibit antidiabetic effects. However, the effects of Rb2 on atherosclerotic responses such as inflammation and ER stress in endothelial cells and monocytes remain unclear. In this study, the expression of inflammation and ER stress markers was determined using a Western blotting method. Concentrations of tumor necrosis factor alpha (TNF[Formula: see text]) and monocyte chemoattractant protein-1 (MCP-1) in culture media were assessed by enzyme-linked immunosorbent assay (ELISA) and apoptosis was evaluated by a cell viability assay and a caspase-3 activity measurement kit. We found that exposure of HUVECs and THP-1 monocytes to Rb2 attenuated inflammation and ER stress, resulting in amelioration of apoptosis and THP-1 cell adhesion to HUVECs under lipopolysaccharide (LPS) condition. Increased AMPK phosphorylation and heme oxygenase (HO)-1 expression, including GPR120 expression were observed in Rb2-treated HUVECs and THP-1 monocytes. Downregulation of both, AMPK phosphorylation and HO-1expression rescued these observed changes. Furthermore, GPR120 siRNA mitigated Rb2-induced AMPK phosphorylation. These results suggest that Rb2 inhibits LPS-mediated apoptosis and THP-1 cell adhesion to HUVECs by GPR120/AMPK/HO-1-associated attenuating inflammation and ER stress. Therefore, Rb2 can be used as a potential therapeutic molecule for treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jaw Long Sun
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey.,Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
60
|
Di Pasquale E, Condorelli G. Endoplasmic reticulum stress at the crossroads of progeria and atherosclerosis. EMBO Mol Med 2020; 11:emmm.201910360. [PMID: 30902910 PMCID: PMC6460347 DOI: 10.15252/emmm.201910360] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare pathology caused by a specific mutation (c.1824C>T; p.G608G) in the LMNA gene (Eriksson et al, 2003). In healthy conditions, LMNA encodes lamins A and C, two major structural nuclear proteins. The mutation creates a splice site in exon 11, resulting in ubiquitous expression of progerin, an aberrant lamin A precursor. Mutations of LMNA can cause laminopathies, a group of diseases with a wide spectrum of, often overlapping, tissue‐specific phenotypes. HGPS is probably one of the most devastating forms of laminopathy. Affected patients display signs of accelerated aging, such as lack of subcutaneous fat, hair loss, joint contractures, and skin thinning, and usually die prematurely from cardiovascular complications. Atherosclerosis is one of the most severe and clinically relevant features of HGPS, manifesting in the absence of classical risk factors, such as increased low‐density lipoprotein and C‐reactive protein (Gordon et al, 2005). In this issue, Hamczyk et al (2019) describe a mechanism for HGPS‐related atherosclerosis.
Collapse
Affiliation(s)
- Elisa Di Pasquale
- Institute of Genetic and Biomedical Research (IRGB) - Milan Unit, National Research Council, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS Rozzano, Milan, Italy
| | - Gianluigi Condorelli
- Institute of Genetic and Biomedical Research (IRGB) - Milan Unit, National Research Council, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS Rozzano, Milan, Italy.,Humanitas University, Milan, Italy
| |
Collapse
|
61
|
|
62
|
PERK Inhibition Mitigates Restenosis and Thrombosis: A Potential Low-Thrombogenic Antirestenotic Paradigm. JACC Basic Transl Sci 2020; 5:245-263. [PMID: 32215348 PMCID: PMC7091514 DOI: 10.1016/j.jacbts.2019.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 12/13/2019] [Accepted: 12/13/2019] [Indexed: 12/18/2022]
Abstract
Drug-eluting stents impede neointimal smooth muscle cell hyperplasia but exacerbate endothelial cell dysfunction and thrombogenicity. It has been a challenge to identify a common target to inhibit both. Findings in this study suggest PERK as such a target. A PERK inhibitor administered either via an endovascular (in biomimetic nanocarriers) or perivascular (in hydrogel) route effectively mitigated neointimal hyperplasia in rats. Oral gavage of the PERK inhibitor partially preserved the normal blood flow in a mouse model of induced thrombosis. Dampening PERK activity inhibited STAT3 while activating SRF in smooth muscle cells, and also reduced prothrombogenic tissue factor and growth impairment of endothelial cells.
Developing endothelial-protective, nonthrombogenic antirestenotic treatments has been a challenge. A major hurdle to this has been the identification of a common molecular target in both smooth muscle cells and endothelial cells, inhibition of which blocks dysfunction of both cell types. The authors’ findings suggest that the PERK kinase could be such a target. Importantly, PERK inhibition mitigated both restenosis and thrombosis in preclinical models, implicating a low-thrombogenic antirestenotic paradigm.
Collapse
Key Words
- ATF, activating transcription factor
- Ad, adenovirus
- CHOP, CCAAT-enhancer-binding protein homologous protein
- DES, drug-eluting stents
- DMSO, dimethyl sulfoxide
- EC, endothelial cell
- ER, endoplasmic reticulum
- FBS, fetal bovine serum
- GFP, green fluorescent protein
- HA, hemagglutinin
- I/M, intima to media
- IEL, internal elastic lamina
- IH, intimal hyperplasia
- IRE1, inositol-requiring kinase 1
- MRTF-A, myocardin related transcription factor A
- PDGF, platelet-derived growth factor
- PDGF-BB, platelet-derived growth factor with 2 B subunits
- PERK
- PERK, protein kinase RNA-like endoplasmic reticulum kinase
- SMA, smooth muscle actin
- SMC, smooth muscle cell
- SRF, serum response factor
- STAT3, signal transducer and activator of transcription 3
- TNF, tumor necrosis factor
- eIF2, eukaryotic translation initiation factor 2
- endothelial cells
- restenosis
- siRNA, small interfering ribonucleic acid
- smooth muscle cells
- thrombosis
Collapse
|
63
|
Pradas-Juni M, Hansmeier NR, Link JC, Schmidt E, Larsen BD, Klemm P, Meola N, Topel H, Loureiro R, Dhaouadi I, Kiefer CA, Schwarzer R, Khani S, Oliverio M, Awazawa M, Frommolt P, Heeren J, Scheja L, Heine M, Dieterich C, Büning H, Yang L, Cao H, Jesus DFD, Kulkarni RN, Zevnik B, Tröder SE, Knippschild U, Edwards PA, Lee RG, Yamamoto M, Ulitsky I, Fernandez-Rebollo E, Vallim TQDA, Kornfeld JW. A MAFG-lncRNA axis links systemic nutrient abundance to hepatic glucose metabolism. Nat Commun 2020; 11:644. [PMID: 32005828 PMCID: PMC6994702 DOI: 10.1038/s41467-020-14323-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/27/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity and type 2 diabetes mellitus are global emergencies and long noncoding RNAs (lncRNAs) are regulatory transcripts with elusive functions in metabolism. Here we show that a high fraction of lncRNAs, but not protein-coding mRNAs, are repressed during diet-induced obesity (DIO) and refeeding, whilst nutrient deprivation induced lncRNAs in mouse liver. Similarly, lncRNAs are lost in diabetic humans. LncRNA promoter analyses, global cistrome and gain-of-function analyses confirm that increased MAFG signaling during DIO curbs lncRNA expression. Silencing Mafg in mouse hepatocytes and obese mice elicits a fasting-like gene expression profile, improves glucose metabolism, de-represses lncRNAs and impairs mammalian target of rapamycin (mTOR) activation. We find that obesity-repressed LincIRS2 is controlled by MAFG and observe that genetic and RNAi-mediated LincIRS2 loss causes elevated blood glucose, insulin resistance and aberrant glucose output in lean mice. Taken together, we identify a MAFG-lncRNA axis controlling hepatic glucose metabolism in health and metabolic disease. Despite widespread transcription of LncRNA in mammalian systems, their contribution to metabolic homeostasis at the cellular and tissue level remains elusive. Here Pradas-Juni et al. describe a transcription factor–LncRNA pathway that couples hepatocyte nutrient sensing to regulation of glucose metabolism in mice.
Collapse
Affiliation(s)
- Marta Pradas-Juni
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.,Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Nils R Hansmeier
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Jenny C Link
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.,Department of Medicine, Division of Cardiology, UCLA, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Elena Schmidt
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Bjørk Ditlev Larsen
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Paul Klemm
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Nicola Meola
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Hande Topel
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.,Izmir Biomedicine and Genome Center (IBG), Mithatpasa Ave. 58/5, 35340, Izmir, Turkey.,Department of Medical Biology and Genetics, Graduate School of Health Sciences, Dokuz Eylul University, Mithatpasa Ave. 1606, 35330, Izmir, Turkey
| | - Rute Loureiro
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.,Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Ines Dhaouadi
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Christoph A Kiefer
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Robin Schwarzer
- Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Sajjad Khani
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Matteo Oliverio
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Motoharu Awazawa
- Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.,Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Peter Frommolt
- Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, Martinistraße 52, 20246, Hamburg, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, Martinistraße 52, 20246, Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, Martinistraße 52, 20246, Hamburg, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hanover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ling Yang
- Cardiovascular Branch, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA.,Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Haiming Cao
- Cardiovascular Branch, National Heart Lung and Blood Institute, Bethesda, MD, 20892, USA
| | - Dario F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Rohit N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Branko Zevnik
- CECAD in vivo Research Facility, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Simon E Tröder
- CECAD in vivo Research Facility, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, University Hospital Ulm, Albert-Einstein Allee 93, 89081, Ulm, Germany
| | - Peter A Edwards
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.,Department of Medicine, Division of Cardiology, UCLA, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | | | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku Medical Megabank Organization, Sendai, 980-8573, Japan
| | - Igor Ulitsky
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Eduardo Fernandez-Rebollo
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Thomas Q de Aguiar Vallim
- Department of Biological Chemistry, University of California, Los Angeles (UCLA), 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA. .,Department of Medicine, Division of Cardiology, UCLA, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA.
| | - Jan-Wilhelm Kornfeld
- Functional Genomics and Metabolism Unit, Department for Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark. .,Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany. .,Cologne Cluster of Excellence-Cellular Stress Responses in Ageing-associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany.
| |
Collapse
|
64
|
Wei K, Luo J, Cao J, Peng L, Ren L, Zhang F. Adiponectin Protects Obese Rats from Aggravated Acute Lung Injury via Suppression of Endoplasmic Reticulum Stress. Diabetes Metab Syndr Obes 2020; 13:4179-4190. [PMID: 33192080 PMCID: PMC7653273 DOI: 10.2147/dmso.s278684] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Endoplasmic reticulum (ER) stress seems to mediate the obesity-induced susceptibility to acute lung injury (ALI). The present study was designed to evaluate the role of ER stress in adiponectin (APN)-induced lung protection in an obese rat model treated with lipopolysaccharide (LPS). METHODS Four-week-old male Sprague-Dawley rats fed either a normal chow diet or a high-fat diet for 12 weeks were randomly assigned to one of the following groups: lean rats, diet-induced obesity rats, lean rats with ALI, obese rats with ALI, obese rats pretreated with 4-phenylbutyric acid (4-PBA) before ALI or obese rats pretreated with APN before ALI. At 24 h after instillation of LPS into the lungs, cell counts in the bronchoalveolar lavage fluid (BALF) were determined. Lung tissues were separated to assess the degree of inflammation, pulmonary oedema, epithelial apoptosis and the expression of ER stress marker proteins. RESULTS The 78-kDa glucose-regulated protein (GRP78) and C/EBP homologous protein (CHOP) expression in the lung tissues of obese rats was upregulated before ALI, as well as the elevated apoptosis in epithelial cells. During ALI, the expression of ER stress marker proteins was similarly increased in both lean and obese rats, while significant downregulation of Mitofusin 2 (MFN2) was detected in obese epithelial cells. The lung tissues of obese rats showed higher concentrations of tumor necrosis factor-alpha (TNF-α), Interleukin 6 (IL-6) and IL-10, enhanced neutrophil counts and elevated wet/dry weight ratios. APN and 4-PBA decreased the degree of ER stress and suppressed LPS-induced lung inflammation, pulmonary oedema and epithelial apoptosis. CONCLUSION APN may exert protective effects against the exacerbated lung injuries in obese rats by attenuating ER stress, which operates as a key molecular pathway in the progression of ALI.
Collapse
Affiliation(s)
- Ke Wei
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Correspondence: Ke Wei Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1# Youyi Road, Yuzhong District, Chongqing, People’s Republic of ChinaTel +86 23 89011069Fax +86 23 89011062 Email
| | - Jie Luo
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jun Cao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Lihua Peng
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Li Ren
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Fan Zhang
- Department of Anesthesiology, Jianyang People’s Hospital, Jianyang, Sichuan641400, People’s Republic of China
| |
Collapse
|
65
|
Guerrini V, Gennaro ML. Foam Cells: One Size Doesn't Fit All. Trends Immunol 2019; 40:1163-1179. [PMID: 31732284 DOI: 10.1016/j.it.2019.10.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/10/2019] [Accepted: 10/12/2019] [Indexed: 02/07/2023]
Abstract
Chronic inflammation in many infectious and metabolic diseases, and some cancers, is accompanied by the presence of foam cells. These cells form when the intracellular lipid content of macrophages exceeds their capacity to maintain lipid homeostasis. Concurrently, critical macrophage immune functions are diminished. Current paradigms of foam cell formation derive from studies of atherosclerosis. However, recent studies indicate that the mechanisms of foam cell biogenesis during tuberculosis differ from those operating during atherogenesis. Here, we review how foam cell formation and function vary with disease context. Since foam cells are therapeutic targets in atherosclerosis, further research on the disease-specific mechanisms of foam cell biogenesis and function is needed to explore the therapeutic consequences of targeting these cells in other diseases.
Collapse
Affiliation(s)
- Valentina Guerrini
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Maria Laura Gennaro
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|
66
|
Zahid MDK, Rogowski M, Ponce C, Choudhury M, Moustaid-Moussa N, Rahman SM. CCAAT/enhancer-binding protein beta (C/EBPβ) knockdown reduces inflammation, ER stress, and apoptosis, and promotes autophagy in oxLDL-treated RAW264.7 macrophage cells. Mol Cell Biochem 2019; 463:211-223. [PMID: 31686316 DOI: 10.1007/s11010-019-03642-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is associated with deregulated cholesterol metabolism and formation of macrophage foam cells. CCAAT/enhancer-binding protein beta (C/EBPβ) is a transcription factor, and its inhibition has recently been shown to prevent atherosclerosis development and foam cell formation. However, whether C/EBPβ regulates inflammation, endoplasmic reticulum (ER) stress, and apoptosis, in macrophage foam cells and its underlying molecular mechanism remains unknown. Here, we investigated the effect of C/EBPβ knockdown on proteins and genes implicated in inflammation, ER stress, apoptosis, and autophagy in macrophage foam cells. RAW264.7 macrophage cells were transfected with control and C/EBPβ-siRNA and then treated with nLDL and oxLDL. Key proteins and genes involved in inflammation, ER stress, apoptosis, and autophagy were analyzed by western blot and qPCR. We found that short interfering RNA (siRNA)-mediated knockdown of C/EBPβ attenuated atherogenic lipid-mediated induction of proteins and genes implicated in inflammation (P-NFkB-p65, NFkB-p65, and TNFα), ER stress (ATF4 and ATF6), and apoptosis (CHOP, caspase 1, 3, and 12). Interestingly, C/EBPβ knockdown upregulated the expression of autophagy proteins (LC3A/B-II, ATG5) and genes (LC3B, ATG5) but decreased the mammalian target of rapamycin (mTOR) protein phosphorylation and mTORC1 gene expression in oxLDL-loaded RAW264.7 macrophage cells. More importantly, treatment with rapamycin (inhibitor of mTOR) increased expression of proteins implicated in autophagy and cholesterol efflux in oxLDL-loaded RAW 264.7 macrophage cells. The present results suggest that C/EBPβ inactivation regulates macrophage foam cell formation in atherogenesis by reducing inflammation, ER stress, and apoptosis and by promoting autophagy and inactivating mTOR.
Collapse
Affiliation(s)
- M D Khurshidul Zahid
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX, 79409-1270, USA
| | - Michael Rogowski
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX, 79409-1270, USA
| | - Christopher Ponce
- Department of Mathematics, Texas Tech University, Lubbock, TX, 79409-1270, USA
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M, Health Sciences Center, College Station, TX, 78363, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX, 79409-1270, USA
| | - Shaikh M Rahman
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX, 79409-1270, USA.
| |
Collapse
|
67
|
Enhanced cellular cholesterol efflux by naringenin is mediated through inhibiting endoplasmic reticulum stress - ATF6 activity in macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1472-1482. [DOI: 10.1016/j.bbalip.2019.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/28/2019] [Accepted: 06/03/2019] [Indexed: 11/22/2022]
|
68
|
Hsieh CL, Huang HS, Chen KC, Saka T, Chiang CY, Chung LWK, Sung SY. A Novel Salicylanilide Derivative Induces Autophagy Cell Death in Castration-Resistant Prostate Cancer via ER Stress-Activated PERK Signaling Pathway. Mol Cancer Ther 2019; 19:101-111. [PMID: 31530650 DOI: 10.1158/1535-7163.mct-19-0387] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/24/2019] [Accepted: 09/12/2019] [Indexed: 11/16/2022]
Abstract
Metastatic castration-resistant prostate cancer (CRPC) is currently incurable. Cancer growth and progression is intimately affected by its interaction with host microenvironment. Cotargeting of the stroma and prostate cancer is therefore an emerging therapeutic strategy for metastatic CRPC. Cancer-induced osteoclastogenesis is known to contribute to CRPC bone metastasis. This study is to extend pharmacologic value of our synthesized LCC03, a derivative of 5-(2',4'-difluorophenyl)-salicylanilide that has previously testified for its osteoclastogenesis activity, by exploring its additional cytotoxic properties and underlying mechanism in CRPC cells. LCC03 was chemically synthesized and examined for cell growth inhibition in a serial of CRPC cell lines. We demonstrated that LCC03 dose-dependently suppressed proliferation and retarded cell-cycle progression in CRPC cells. The classical autophagy features, including autophagosome formation and LC3-II conversion, were dramatically shown in LCC03-treated CRPC cells, and it was associated with the suppressed AKT/mTOR signaling pathways, a major negative regulator of autophagy. Moreover, an expanded morphology of the endoplasmic reticulum (ER), increased expression of the ER stress markers GRP78 and PERK, and eIF2α phosphorylation were observed. Blockage of autophagy and PERK pathways using small molecule inhibitors or shRNA knockdown reversed LCC03-induced autophagy and cell death, thus indicating that the PERK-eIF2α pathway contributed to the LCC03-induced autophagy. Furthermore, treatment of tumor-bearing mice with intraperitoneal administered LCC03 suppressed the growth of CRPC xenografts in mouse bone without systemic toxicity. The dual action of 5-(2',4'-difluorophenyl)-salicylanilide on targeting both the osteoclasts and the tumor cells strongly indicates that LCC03 is a promising anticancer candidate for preventing and treating metastatic CRPC.
Collapse
Affiliation(s)
- Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsu-Shan Huang
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Chou Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Urology, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Urology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Teigi Saka
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ying Chiang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Leland W K Chung
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shian-Ying Sung
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan. .,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Joint Clinical Research Center, Office of Human Research, Taipei Medical University, Taipei, Taiwan.,Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
69
|
Chung YP, Yen CC, Tang FC, Lee KI, Liu SH, Wu CC, Hsieh SS, Su CC, Kuo CY, Chen YW. Methylmercury exposure induces ROS/Akt inactivation-triggered endoplasmic reticulum stress-regulated neuronal cell apoptosis. Toxicology 2019; 425:152245. [PMID: 31330229 DOI: 10.1016/j.tox.2019.152245] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/06/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
Epidemiological studies have positively linked mercury exposure and neurodegenerative diseases (ND). Methylmercury (MeHg), an organic form of mercury, is a ubiquitous and potent environmental neurotoxicant that easily crosses the blood-brain barrier and causes irreversible injury to the central nervous system (CNS). However, the molecular mechanisms underlying MeHg-induced neurotoxicity remain unclear. Here, the present study found that Neuro-2a cells underwent apoptosis in response to MeHg (1-5 μM), which was accompanied by increased phosphatidylserine (PS) exposure on the outer cellular membrane leaflets, caspase-3 activity, and the activation of caspase cascades and poly (ADP-ribose) polymerase (PARP). Exposure of Neuro-2a cells to MeHg also triggered endoplasmic reticulum (ER) stress, which was identified via several key molecules (including: glucose-regulated protein (GRP)78, GRP94, C/EBP homologous protein (CHOP) X-box binding protein(XBP)-1, protein kinase R-like ER kinase (PERK), eukaryotic initiation factor 2α (eIF2α), inositol-requiring enzyme(IRE)-1, activation transcription factor(AFT)4, and ATF6. Transfection with GRP78-, GRP94-, CHOP-, and XBP-1-specific small interfering (si)RNA significantly suppressed the expression of these proteins, and attenuated cytotoxicity and caspase-12, -7, and -3 activation in MeHg-exposed cells. Furthermore, MeHg dramatically decreased Akt phosphorylation, and the overexpression of activation of Akt1 (myr-Akt1) could significantly prevent MeHg-induced Akt inactivation, as well as apoptotic and ER stress-related signals. Pretreatment with the antioxidant N-acetylcysteine (NAC) effectively prevented MeHg-induced neuronal cell reactive oxygen species (ROS) generation, apoptotic and ER stress-related signals, and Akt inactivation. Collectively, these results indicate that MeHg exerts its cytotoxicity in neurons by inducing ROS-mediated Akt inactivation up-regulated ER stress, which induces apoptosis and ultimately leads to cell death.
Collapse
Affiliation(s)
- Yao-Pang Chung
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Department of Occupational Safety and Health, College of Health Care and Management, Chung Shan Medical University, Taichung, 402, Taiwan
| | - Cheng-Chieh Yen
- Department of Occupational Safety and Health, College of Health Care and Management, Chung Shan Medical University, Taichung, 402, Taiwan; Department of Occupational Medicine, Chung Shan Medical University Hospital, Taichung, 402, Taiwan
| | - Feng-Cheng Tang
- Department of Occupational Medicine, Changhua Christian Hospital, Changhua County, 500, Taiwan; Department of Leisure Services Management, Chaoyang University of Technology, Taichung, 413, Taiwan
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taichung, 427, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chin-Ching Wu
- Department of Public Health, China Medical University, Taichung, 404, Taiwan
| | - Shang-Shu Hsieh
- Department of Emergency, Taichung Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, Taichung, 427, Taiwan
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
| | - Chun-Ying Kuo
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County 500, Taiwan
| | - Ya-Wen Chen
- Department of Physiology and Graduate Institute of Basic Medical Science, School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
70
|
Enhanced endoplasmic reticulum and mitochondrial stress in abdominal aortic aneurysm. Clin Sci (Lond) 2019; 133:1421-1438. [PMID: 31239294 DOI: 10.1042/cs20190399] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative vascular disease with a complex aetiology that remains to be fully elucidated. Clinical management of AAA is limited to surgical repair, while an effective pharmacotherapy is still awaited. Endoplasmic reticulum (ER) stress and mitochondrial dysfunction have been involved in the pathogenesis of cardiovascular diseases (CVDs), although their contribution to AAA development is uncertain. Therefore, we aimed to determine their implication in AAA and investigated the profile of oxysterols in plasma, specifically 7-ketocholesterol (7-KC), as an ER stress inducer.In the present study, we determined aortic ER stress activation in a large cohort of AAA patients compared with healthy donors. Higher gene expression of activating transcription factor (ATF) 6 (ATF6), IRE-1, X-binding protein 1 (XBP-1), C/EBP-homologous protein (CHOP), CRELD2 and suppressor/enhancer of Lin-12-like (SEL1L) and greater protein levels of active ATF6, active XBP1 and of the pro-apoptotic protein CHOP were detected in human aneurysmatic samples. This was accompanied by an exacerbated apoptosis, higher reactive oxygen species (ROS) production and by a reduction in mitochondrial biogenesis in the vascular wall of AAA. The quantification of oxysterols, performed by liquid chromatography-(atmospheric pressure chemical ionization (APCI))-mass spectrometry, showed that levels of 7-KC were significantly higher while those of 7α-hydroxycholesterol (HC), 24-HC and 27-HC were lower in AAA patients compared with healthy donors. Interestingly, the levels of 7-KC correlate with the expression of ER stress markers.Our results evidence an induction of ER stress in the vascular wall of AAA patients associated with an increase in circulating 7-KC levels and a reduction in mitochondrial biogenesis suggesting their implication in the pathophysiology of this disease.
Collapse
|
71
|
Kudyba HM, Cobb DW, Fierro MA, Florentin A, Ljolje D, Singh B, Lucchi NW, Muralidharan V. The endoplasmic reticulum chaperone PfGRP170 is essential for asexual development and is linked to stress response in malaria parasites. Cell Microbiol 2019; 21:e13042. [PMID: 31087747 PMCID: PMC6699899 DOI: 10.1111/cmi.13042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/09/2019] [Accepted: 05/09/2019] [Indexed: 12/24/2022]
Abstract
The vast majority of malaria mortality is attributed to one parasite species: Plasmodium falciparum. Asexual replication of the parasite within the red blood cell is responsible for the pathology of the disease. In Plasmodium, the endoplasmic reticulum (ER) is a central hub for protein folding and trafficking as well as stress response pathways. In this study, we tested the role of an uncharacterised ER protein, PfGRP170, in regulating these key functions by generating conditional mutants. Our data show that PfGRP170 localises to the ER and is essential for asexual growth, specifically required for proper development of schizonts. PfGRP170 is essential for surviving heat shock, suggesting a critical role in cellular stress response. The data demonstrate that PfGRP170 interacts with the Plasmodium orthologue of the ER chaperone, BiP. Finally, we found that loss of PfGRP170 function leads to the activation of the Plasmodium eIF2α kinase, PK4, suggesting a specific role for this protein in this parasite stress response pathway.
Collapse
Affiliation(s)
- Heather M Kudyba
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - David W Cobb
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Manuel A Fierro
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Anat Florentin
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Dragan Ljolje
- Malaria Branch and Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Balwan Singh
- Malaria Branch and Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Naomi W Lucchi
- Malaria Branch and Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Vasant Muralidharan
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Cellular Biology, University of Georgia, Athens, Georgia
| |
Collapse
|
72
|
Yap J, Cabrera-Fuentes HA, Irei J, Hausenloy DJ, Boisvert WA. Role of Macrophages in Cardioprotection. Int J Mol Sci 2019; 20:E2474. [PMID: 31109146 PMCID: PMC6566352 DOI: 10.3390/ijms20102474] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/14/2019] [Accepted: 05/17/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality worldwide. It is widely known that non-resolving inflammation results in atherosclerotic conditions, which are responsible for a host of downstream pathologies including thrombosis, myocardial infarction (MI), and neurovascular events. Macrophages, as part of the innate immune response, are among the most important cell types in every stage of atherosclerosis. In this review we discuss the principles governing macrophage function in the healthy and infarcted heart. More specifically, how cardiac macrophages participate in myocardial infarction as well as cardiac repair and remodeling. The intricate balance between phenotypically heterogeneous populations of macrophages in the heart have profound and highly orchestrated effects during different phases of myocardial infarction. In the early "inflammatory" stage of MI, resident cardiac macrophages are replaced by classically activated macrophages derived from the bone marrow and spleen. And while the macrophage population shifts towards an alternatively activated phenotype, the inflammatory response subsides giving way to the "reparative/proliferative" phase. Lastly, we describe the therapeutic potential of cardiac macrophages in the context of cell-mediated cardio-protection. Promising results demonstrate innovative concepts; one employing a subset of yolk sac-derived, cardiac macrophages that have complete restorative capacity in the injured myocardium of neonatal mice, and in another example, post-conditioning of cardiac macrophages with cardiosphere-derived cells significantly improved patient's post-MI diagnoses.
Collapse
Affiliation(s)
- Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| | - Hector A Cabrera-Fuentes
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL 264610, Mexico.
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia.
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Yong Loo Lin School of Medicine, National University Singapore, Singapore 117597, Singapore.
- The Hatter Cardiovascular Institute, University College London, London WC1E 6HX, UK.
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, London W1T 7DN, UK.
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia.
| |
Collapse
|
73
|
Kurshakova MM, Nabirochkina EN, Georgieva SG, Kopytova DV. TRF4, the novel TBP-related protein of Drosophila melanogaster, is concentrated at the endoplasmic reticulum and copurifies with proteins participating in the processes associated with endoplasmic reticulum. J Cell Biochem 2019; 120:7927-7939. [PMID: 30426565 DOI: 10.1002/jcb.28070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
Understanding the functions of TBP-related factors is essential for studying chromatin assembly and transcription regulation in higher eukaryotes. The novel TBP-related protein-coding gene, trf4, was described in Drosophila melanogaster. trf4 is found only in Drosophila and has likely originated in Drosophila common ancestor. TRF4 protein has a distant homology with TBP and TRF2 in the region of TBP-like domain and is evolutionarily conserved among distinct Drosophila species, which indicates its functional significance. TRF4 is widely expressed in D. melanogaster with high levels of its expression being observed in testes. Interestingly enough, TRF4 has become a cytoplasmic protein having lost nuclear localization signal sequence. TRF4 is concentrated at the endoplasmic reticulum (ER) and copurifies with the proteins participating in the ER-associated processes. We suggest that trf4 gene is an example of homolog neofunctionalization by protein subcellular relocalization pathway, where the subcellular relocalization of gene product of duplicated gene leads to the new functions in ER-associated processes.
Collapse
Affiliation(s)
- Maria M Kurshakova
- Department of transcription factors of eukaryotes, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Elena N Nabirochkina
- Department of transcription factors of eukaryotes, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sofia G Georgieva
- Department of transcription factors of eukaryotes, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Daria V Kopytova
- Department of transcription factors of eukaryotes, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
74
|
Lipid-regulating properties of butyric acid and 4-phenylbutyric acid: Molecular mechanisms and therapeutic applications. Pharmacol Res 2019; 144:116-131. [PMID: 30954630 DOI: 10.1016/j.phrs.2019.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/08/2019] [Accepted: 04/01/2019] [Indexed: 12/30/2022]
Abstract
In the past two decades, significant advances have been made in the etiology of lipid disorders. Concomitantly, the discovery of liporegulatory functions of certain short-chain fatty acids has generated interest in their clinical applications. In particular, butyric acid (BA) and its derivative, 4-phenylbutyric acid (PBA), which afford health benefits against lipid disorders while being generally well tolerated by animals and humans have been assessed clinically. This review examines the evidence from cell, animal and human studies pertaining to the lipid-regulating effects of BA and PBA, their molecular mechanisms and therapeutic potential. Collectively, the evidence supports the view that intakes of BA and PBA benefit lipid homeostasis across biological systems. We reviewed the evidence that BA and PBA downregulate de novo lipogenesis, ameliorate lipotoxicity, slow down atherosclerosis progression, and stimulate fatty acid β-oxidation. Central to their mode of action, BA appears to function as a histone deacetylase (HDAC) inhibitor while PBA acts as a chemical chaperone and/or a HDAC inhibitor. Areas of further inquiry include the effects of BA and PBA on adipogenesis, lipolysis and apolipoprotein metabolism.
Collapse
|
75
|
Chae CW, Kwon YW. Cell signaling and biological pathway in cardiovascular diseases. Arch Pharm Res 2019; 42:195-205. [PMID: 30877558 DOI: 10.1007/s12272-019-01141-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
Abstract
Currently, coronary artery disease accounts for a large proportion of deaths occurring worldwide. Damage to the heart muscle over a short period of time leads to myocardial infarction (MI). The biological mechanisms of atherosclerosis, one of the causes of MI, have been well studied. Resistin, a type of adipokine, is closely associated with intravascular level of low-density lipoprotein cholesterol and augmentation of the expression of adhesion molecules in endothelial cells. Therefore, resistin, which is highly associated with inflammation, can progress into coronary artery disease. Adenylyl cyclase associated protein 1, a binding partner of resistin, also plays an important role in inducing pro-inflammatory cytokines. The induction of these cytokines can aggravate atherosclerosis by promoting severe plaque rupture of the lesion site. Recently, drugs, such as statins that can inhibit inflammation have been extensively studied. The development of effective new drugs that can directly or indirectly block pro-inflammatory cytokines may have a great potential in the treatment of coronary artery disease in the future.
Collapse
Affiliation(s)
- Cheong-Whan Chae
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, 110-744, Republic of Korea
| | - Yoo-Wook Kwon
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, 110-744, Republic of Korea. .,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea. .,Department of Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
76
|
Xia MM, Wang M, Jiang H, Liu Y, Ma L, Lu C, Zhang W. Association of Angiotensin-Converting Enzyme Insertion/Deletion Polymorphism with the Risk of Atherosclerosis. J Stroke Cerebrovasc Dis 2019; 28:1732-1743. [PMID: 30878369 DOI: 10.1016/j.jstrokecerebrovasdis.2019.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/17/2019] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
AIMS The objective of this study was to perform a meta-analysis to evaluate the association between angiotensin-converting enzyme (ACE) gene insertion/deletion (I/D) polymorphism and susceptibility to atherosclerosis (AS). METHODS MEDLINE, EMBASE, and the ISI Web of Science were searched for all eligible published studies concerning the relationship of ACE gene polymorphism with AS without language restrictions. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to evaluate this relationship under different genetic models using meta-analytic methods. RESULTS A total of 15 articles (16 studies) were involved in this meta-analysis. The D allele of the ACE gene had a nonsignificant increase in the risk of AS (D versus I: OR = 1.23, 95% CI, .98-1.53, P = .07; I2 = 87.2%, Pheterogeneity < .01). Compared with the II genotype, the DI (relative risk [RR]: 1.35, 95% CI: 1.09, 1.67, P < .01; I2 = 47.8%, Pheterogeneity = .017) and (DD + DI) (RR = 1.38, 95% CI: 1.04, 1.82, P = .02; I2 = 73.3%, Pheterogeneity < .01) genotype of ACE was associated with higher risk of AS, respectively. Subjects with the DD genotype showed a statistically nonsignificant trend toward greater risk of AS (RR = 1.53, 95% CI: .97, 2.43, P = .07; I2 = 88.6%, Pheterogeneity < .01). Further subgroup analyses showed that significant relationships were only found in Europeans under different gene polymorphism or different genotype models rather than Asians. CONCLUSIONS The present meta-analysis indicated that the D allele in the ACE gene was associated with the risk of AS, especially in Europeans. Furthermore, increased copy number of D allele was significantly associated with increased AS risk in a dose-dependent manner.
Collapse
Affiliation(s)
- Man-Man Xia
- The First Affiliated Hospital, Xi'an Jiaotong University College of medicine, Xi'an, China; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Mingxu Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Hong Jiang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yan Liu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China.
| | - Le Ma
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Chao Lu
- Xi'an Honghui Hospital, Xi'an, China.
| | - Wei Zhang
- The First Affiliated Hospital, Xi'an Jiaotong University College of medicine, Xi'an, China.
| |
Collapse
|
77
|
Sun D, Zhang M, Li Y, Mei S, Qin J, Yan J. c‑Jun/Ap‑1 is upregulated in an Ang II‑induced abdominal aortic aneurysm formation model and mediates Chop expression in mouse aortic smooth muscle cells. Mol Med Rep 2019; 19:3459-3468. [PMID: 30864718 PMCID: PMC6472129 DOI: 10.3892/mmr.2019.10017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 02/08/2019] [Indexed: 01/30/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is an asymptomatic, potentially lethal disease whose ruptures have a high mortality rate. An effective pharmacological approach to decrease expansion or prevent the rupture of AAAs in humans remains lacking. Previous studies have suggested that activator protein 1 (c-Jun/AP-1) and C/EBP homologous protein (Chop) are involved in the development of AAA. The purpose of the present study was to investigate whether c-Jun/AP-1 mediates Chop overexpression in AAA. c-Jun/AP-1 and Chop protein levels were determined in an angiotensin II (Ang II)-induced AAA model using apolipoprotein E-deficient mice. Additionally, mouse aortic smooth muscle cells (MOVAS cell line) were treated with Ang II. Apoptosis was evaluated via TUNEL assay, MOVAS cell migration ability was assessed by monolayer wound healing assay and the levels of c-Jun/AP-1 and Chop were determined by western blotting, immunofluorescence and immunocytochemical assays. Following c-Jun silencing using c-Jun-specific small interfering (si)RNA, Chop expression was evaluated. Furthermore, chromatin immunoprecipitation (ChIP) was used to investigate whether c-Jun/Ap-1 binds directly to the DNA damage-inducible transcript 3 protein (Ddit3) promoter. It was observed that c-Jun/AP-1 and Chop were synchronously overexpressed in Ang II-induced AAA and Ang II-treated cells, and that apoptosis and migration were induced by Ang II. In addition, Chop was suppressed when c-Jun was silenced by targeted siRNA. Notably, the ChIP assay demonstrated that the DNA fragments pulled down by primary antibodies against c-Jun/Ap-1 were able to be amplified by (Ddit3) promoter-specific primers. c-Jun/AP-1 may therefore mediate Chop expression in MOVAS cells via Ddit3. These results suggested that c-Jun/AP-1 may be a novel target for AAA therapy.
Collapse
Affiliation(s)
- Dating Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Mingxi Zhang
- Division of Cardiology, Department of Internal Medicine, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430030, P.R. China
| | - Yuanyuan Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shuai Mei
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jin Qin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
78
|
Gold K, Gaharwar AK, Jain A. Emerging trends in multiscale modeling of vascular pathophysiology: Organ-on-a-chip and 3D printing. Biomaterials 2019; 196:2-17. [PMID: 30072038 PMCID: PMC6344330 DOI: 10.1016/j.biomaterials.2018.07.029] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/13/2018] [Accepted: 07/18/2018] [Indexed: 01/17/2023]
Abstract
Most biomedical and pharmaceutical research of the human vascular system aims to unravel the complex mechanisms that drive disease progression from molecular to organ levels. The knowledge gained can then be used to innovate diagnostic and treatment strategies which can ultimately be determined precisely for patients. Despite major advancements, current modeling strategies are often limited at identifying, quantifying, and dissecting specific cellular and molecular targets that regulate human vascular diseases. Therefore, development of multiscale modeling approaches are needed that can advance our knowledge and facilitate the design of next-generation therapeutic approaches in vascular diseases. This article critically reviews animal models, static in vitro systems, and dynamic in vitro culture systems currently used to model vascular diseases. A leading emphasis on the potential of emerging approaches, specifically organ-on-a-chip and three-dimensional (3D) printing, to recapitulate the innate human vascular physiology and anatomy is described. The applications of these approaches and future outlook in designing and screening novel therapeutics are also presented.
Collapse
Affiliation(s)
- Karli Gold
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA; Department of Material Sciences, Texas A&M University, College Station, TX, 77843, USA; Center for Remote Health and Technologies and Systems, Texas A&M University, College Station, TX, 77843, USA.
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
79
|
β-Asarone Regulates ER Stress and Autophagy Via Inhibition of the PERK/CHOP/Bcl-2/Beclin-1 Pathway in 6-OHDA-Induced Parkinsonian Rats. Neurochem Res 2019; 44:1159-1166. [DOI: 10.1007/s11064-019-02757-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/15/2019] [Accepted: 02/17/2019] [Indexed: 12/20/2022]
|
80
|
Leonard A, Grose V, Paton AW, Paton JC, Yule DI, Rahman A, Fazal F. Selective Inactivation of Intracellular BiP/GRP78 Attenuates Endothelial Inflammation and Permeability in Acute Lung Injury. Sci Rep 2019; 9:2096. [PMID: 30765717 PMCID: PMC6376000 DOI: 10.1038/s41598-018-38312-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022] Open
Abstract
The role of Endoplasmic Reticulum Chaperone and Signaling Regulator BiP/GRP78 in acute inflammatory injury, particularly in the context of lung endothelium, is poorly defined. In his study, we monitored the effect of SubAB, a holoenzyme that cleaves and specifically inactivates BiP/GRP78 and its inactive mutant SubAA272B on lung inflammatory injury in an aerosolized LPS inhalation mouse model of acute lung injury (ALI). Analysis of lung homogenates and bronchoalveolar lavage (BAL) fluid showed that LPS-induced lung inflammation and injury were significantly inhibited in SubAB- but not in SubAA272B-treated mice. SubAB-treated mice were also protected from LPS-induced decrease in lung compliance. Gene transfer of dominant negative mutant of BiP in the lung endothelium protected against LPS-induced lung inflammatory responses. Consistent with this, stimulation of endothelial cells (EC) with thrombin caused an increase in BiP/GRP78 levels and inhibition of ER stress with 4-phenylbutyric acid (4-PBA) prevented this response as well as increase in VCAM-1, ICAM-1, IL-6, and IL-8 levels. Importantly, thrombin-induced Ca2+ signaling and EC permeability were also prevented upon BiP/GRP78 inactivation. The above EC responses are mediated by intracellular BiP/GRP78 and not by cell surface BiP/GRP78. Together, these data identify intracellular BiP/GRP78 as a novel regulator of endothelial dysfunction associated with ALI.
Collapse
Affiliation(s)
- Antony Leonard
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA
| | - Valerie Grose
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - David I Yule
- Department of Pharmacology & Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA
| | - Arshad Rahman
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA
| | - Fabeha Fazal
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, New York, 14642, USA.
| |
Collapse
|
81
|
Miyoshi A, Koyama S, Sasagawa-Monden M, Kadoya M, Konishi K, Shoji T, Inaba M, Yamamoto Y, Koyama H. JNK and ATF4 as two important platforms for tumor necrosis factor-α-stimulated shedding of receptor for advanced glycation end products. FASEB J 2018; 33:3575-3589. [PMID: 30452882 DOI: 10.1096/fj.201701553rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Soluble receptor for advanced glycation end products (sRAGE), shed from cell surfaces, is found in human circulation and has been implicated in cardiovascular disease. Its pathophysiological regulation and underlying mechanisms are scarcely understood. In endothelium-specific human RAGE transgenic mice, human sRAGE was detected in circulation, whereas its level was markedly increased after LPS treatment. That increase was preceded by a rapid rise in TNF-α level. Treatment with TNF-α also significantly increased serum sRAGE. In human microvascular endothelial cells or human umbilical vein endothelial cells with RAGE overexpression, TNF-α markedly induced RAGE shedding, which was dependent on MMP9 and ADAM10. TNF-α-stimulated MMP9 expression was completely dependent on JNK activation, with its inhibition partially effective in suppressing TNF-α-induced RAGE shedding. In contrast, TNF-α transiently induced activation transcription factor (ATF)4, a major component in unfolded protein response (UPR), whereas knockdown of ATF4 abrogated TNF-α-stimulated RAGE shedding. Protein levels of the pro and activated forms of ADAM10 were also decreased by ATF4 knockdown, whereas inhibition of other components of UPR, including XBP1 and ATF6, failed to block TNF-α-stimulated RAGE shedding. Although the endoplasmic reticulum stressors thapsigargin and tunicamycin induced markedly and sustained expression of ATF4 and XBP-1, they did not induce RAGE shedding to the same level as TNF-α, suggesting that ATF4 is necessary but not sufficient alone for TNF-α-mediated RAGE shedding. ATF4 inhibition did not affect TNF-α-stimulated MMP9 expression, whereas inhibition of JNK activity did not influence ADAM10 activation. Thus, inflammatory cascades including TNF-α induced RAGE shedding in endothelial cells in vivo and in vitro. JNK and ATF4 may be 2 platforms for regulation of TNF-α-stimulated RAGE shedding.-Miyoshi, A., Koyama, S., Sasagawa-Monden, M., Kadoya, M., Konishi, K., Shoji, T., Inaba, M., Yamamoto, Y., Koyama, H. JNK and ATF4 as two important platforms for tumor necrosis factor-α-stimulated shedding of receptor for advanced glycation end products.
Collapse
Affiliation(s)
- Akio Miyoshi
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Sachie Koyama
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masayo Sasagawa-Monden
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan.,Department of Endocrinology, Metabolism, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan; and
| | - Manabu Kadoya
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Kosuke Konishi
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takuhito Shoji
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masaaki Inaba
- Department of Endocrinology, Metabolism, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan; and
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hidenori Koyama
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
82
|
Guo X, Wang L, Xia X, Wang P, Li X. Effects of atorvastatin and/or probucol on recovery of atherosclerosis in high-fat-diet-fed apolipoprotein E-deficient mice. Biomed Pharmacother 2018; 109:1445-1453. [PMID: 30551396 DOI: 10.1016/j.biopha.2018.10.184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/27/2018] [Accepted: 10/31/2018] [Indexed: 10/27/2022] Open
Abstract
INTRODUCTION We have investigated the possible effects and mechanism of atorvastatin, a statin, and/or probucol, a powerful antioxidant used to lower cholesterol before 1995, on the atherosclerosis development. METHODS Apolipoprotein-E-deficient (ApoE-/-) mice fed with the high fat diet were randomly divided into 3 groups (n = 10/each group): Placebo, Atorvastatin (10 mg/ kg/d), and atorvastatin (10 mg/kg/d) plus probucol (10 mg/kg/d) groups. C57BL/6 J mice were fed with normal diet as the control group (n = 10). Animals were sacrificed 10 weeks after the intervention. To evaluate the experimental atherosclerosis, blood tests were used for measuring serum lipoprotein profile, Western blots for endoplasmic reticulum (ER) stress protein expression, H&E staining for plaque lesions, immunohistology for macrophages, inflammatory cytokines, innate immune receptor TLR-4, transcription factor NF-κB, and atherosclerosis plaques. RESULTS Compared with the control group, ApoE-/- mice in the placebo group showed with the significantly (p < 0.05) higher levels of serum total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL) and oxidized low density lipoprotein (ox-LDL), PERK, GRP78, CHOP, IL-1β, TNF-α and NF-κB, but with the lower levels of high-density lipoprotein cholesterol (HDL) and TLR-4, and also the increase in macrophages and the aortic media collagen, and the decrease in the elastic fibers (p < 0.01). Treatment with atorvastatin recovered all these features (p < 0.05 or p < 0.01) near to the levels in the control group. In addition, the combination of atorvastatin and probucol has shown the slightly stronger effect than the use of atorvastatin alone without statistical significances when comparing most bio-markers of atherosclerosis, but with significant differences in the reduction of the plaque lesion areas and macrophages (p < 0.05). CONCLUSIONS Atorvastatin and/or probucol suppresses ER stress and increase the level of TLR-4, which lowers NF-κB, resulting in the recovery of atherosclerosis in the ApoE-/- mouse model.
Collapse
Affiliation(s)
- Xiaokun Guo
- Department of Geratology, The Second Hospital of Tianjin Medical University, PR China
| | - Lin Wang
- Department of Geratology, The Second Hospital of Tianjin Medical University, PR China
| | - Xiaoshuang Xia
- Department of Neurology, The Second Hospital of Tianjin Medical University, PR China
| | - Peilu Wang
- Department of Neurology, The Second Hospital of Tianjin Medical University, PR China
| | - Xin Li
- Department of Geratology, The Second Hospital of Tianjin Medical University, PR China.
| |
Collapse
|
83
|
Kamiya T, Watanabe M, Hara H, Mitsugi Y, Yamaguchi E, Itoh A, Adachi T. Induction of Human-Lung-Cancer-A549-Cell Apoptosis by 4-Hydroperoxy-2-decenoic Acid Ethyl Ester through Intracellular ROS Accumulation and the Induction of Proapoptotic CHOP Expression. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:10741-10747. [PMID: 30296076 DOI: 10.1021/acs.jafc.8b04424] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Royal jelly, a natural product secreted by honeybees, contains several fatty acids, such as 10-hydroxy-2-decenoic acid (DE), and shows anti- and pro-apoptotic properties. 4-Hydroperoxy-2-decenoic acid ethyl ester (HPO-DAEE), a DE derivative, exhibits potent antioxidative activity; however, it currently remains unclear whether HPO-DAEE induces cancer-cell death. In the present study, treatment with HPO-DAEE induced human-lung-cancer-A549-cell death (52.7 ± 10.2%) that was accompanied by DNA fragmentation. Moreover, the accumulation of intracellular reactive oxygen species (ROS, 2.38 ± 0.1-fold) and the induction of proapoptotic CCAAT-enhancer-binding-protein-homologous-protein (CHOP) expression (18.4 ± 4.0-fold) were observed in HPO-DAEE-treated cells. HPO-DAEE-elicited CHOP expression and cell death were markedly suppressed by pretreatment with N-acetylcysteine (NAC), an antioxidant, by 2.40 ± 1.57-fold and 5.7 ± 1.6%, respectively. Pretreatment with 4-phenylbutyric acid (PBA), an inhibitor of endoplasmic reticulum stress, also suppressed A549-cell death (38.4 ± 1.1%). Furthermore, we demonstrated the involvement of extracellular-signal-regulated protein kinase (ERK) and p38-related signaling in HPO-DAEE-elicited cell-death events. Overall, we concluded that HPO-DAEE induces A549-cell apoptosis through the ROS-ERK-p38 pathway and, at least in part, the CHOP pathway.
Collapse
|
84
|
Testa G, Rossin D, Poli G, Biasi F, Leonarduzzi G. Implication of oxysterols in chronic inflammatory human diseases. Biochimie 2018; 153:220-231. [DOI: 10.1016/j.biochi.2018.06.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/07/2018] [Indexed: 12/18/2022]
|
85
|
LincRNA H19 protects from dietary obesity by constraining expression of monoallelic genes in brown fat. Nat Commun 2018; 9:3622. [PMID: 30190464 PMCID: PMC6127097 DOI: 10.1038/s41467-018-05933-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 07/31/2018] [Indexed: 01/22/2023] Open
Abstract
Increasing brown adipose tissue (BAT) thermogenesis in mice and humans improves metabolic health and understanding BAT function is of interest for novel approaches to counteract obesity. The role of long noncoding RNAs (lncRNAs) in these processes remains elusive. We observed maternally expressed, imprinted lncRNA H19 increased upon cold-activation and decreased in obesity in BAT. Inverse correlations of H19 with BMI were also observed in humans. H19 overexpression promoted, while silencing of H19 impaired adipogenesis, oxidative metabolism and mitochondrial respiration in brown but not white adipocytes. In vivo, H19 overexpression protected against DIO, improved insulin sensitivity and mitochondrial biogenesis, whereas fat H19 loss sensitized towards HFD weight gains. Strikingly, paternally expressed genes (PEG) were largely absent from BAT and we demonstrated that H19 recruits PEG-inactivating H19-MBD1 complexes and acts as BAT-selective PEG gatekeeper. This has implications for our understanding how monoallelic gene expression affects metabolism in rodents and, potentially, humans. Brown adipose tissue (BAT) thermogenesis counteracts obesity and promotes metabolic health. The role of long non-coding RNAs (lncRNAs) in the regulation of this process is not well understood. Here the authors identify a maternally expressed lncRNA, H19, that increases BAT oxidative metabolism and energy expenditure.
Collapse
|
86
|
So JS. Roles of Endoplasmic Reticulum Stress in Immune Responses. Mol Cells 2018; 41:705-716. [PMID: 30078231 PMCID: PMC6125421 DOI: 10.14348/molcells.2018.0241] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/03/2018] [Accepted: 07/19/2018] [Indexed: 02/06/2023] Open
Abstract
The endoplasmic reticulum (ER) is a critical organelle for protein synthesis, folding and modification, and lipid synthesis and calcium storage. Dysregulation of ER functions leads to the accumulation of misfolded- or unfolded-protein in the ER lumen, and this triggers the unfolded protein response (UPR), which restores ER homeostasis. The UPR is characterized by three distinct downstream signaling pathways that promote cell survival or apoptosis depending on the stressor, the intensity and duration of ER stress, and the cell type. Mammalian cells express the UPR transducers IRE1, PERK, and ATF6, which control transcriptional and translational responses to ER stress. Direct links between ER stress and immune responses are also evident, but the mechanisms by which UPR signaling cascades are coordinated with immunity remain unclear. This review discusses recent investigations of the roles of ER stress in immune responses that lead to differentiation, maturation, and cytokine expression in immune cells. Further understanding of how ER stress contributes to the pathogenesis of immune disorders will facilitate the development of novel therapies that target UPR pathways.
Collapse
Affiliation(s)
- Jae-Seon So
- Department of Medical Biotechnology, Dongguk University Gyeongju, Gyeongju 38066,
Korea
| |
Collapse
|
87
|
Solano-Gálvez SG, Abadi-Chiriti J, Gutiérrez-Velez L, Rodríguez-Puente E, Konstat-Korzenny E, Álvarez-Hernández DA, Franyuti-Kelly G, Gutiérrez-Kobeh L, Vázquez-López R. Apoptosis: Activation and Inhibition in Health and Disease. Med Sci (Basel) 2018; 6:E54. [PMID: 29973578 PMCID: PMC6163961 DOI: 10.3390/medsci6030054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022] Open
Abstract
There are many types of cell death, each involving multiple and complex molecular events. Cell death can occur accidentally when exposed to extreme physical, chemical, or mechanical conditions, or it can also be regulated, which involves a genetically coded complex machinery to carry out the process. Apoptosis is an example of the latter. Apoptotic cell death can be triggered through different intracellular signalling pathways that lead to morphological changes and eventually cell death. This is a normal and biological process carried out during maturation, remodelling, growth, and development in tissues. To maintain tissue homeostasis, regulatory, and inhibitory mechanisms must control apoptosis. Paradoxically, these same pathways are utilized during infection by distinct intracellular microorganisms to evade recognition by the immune system and therefore survive, reproduce and develop. In cancer, neoplastic cells inhibit apoptosis, thus allowing their survival and increasing their capability to invade different tissues and organs. The purpose of this work is to review the generalities of the molecular mechanisms and signalling pathways involved in apoptosis induction and inhibition. Additionally, we compile the current evidence of apoptosis modulation during cancer and Leishmania infection as a model of apoptosis regulation by an intracellular microorganism.
Collapse
Affiliation(s)
- Sandra Georgina Solano-Gálvez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Jack Abadi-Chiriti
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Luis Gutiérrez-Velez
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Eduardo Rodríguez-Puente
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Enrique Konstat-Korzenny
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Diego-Abelardo Álvarez-Hernández
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| | - Giorgio Franyuti-Kelly
- Medical IMPACT, Infectious Disease Department, Mexico City 53900, Estado de México, Mexico.
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Instituto Nacional de Cardiología, Mexico City, 14080, Mexico.
| | - Rosalino Vázquez-López
- Departamento de Microbiología, Centro de Investigación en Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucán Estado de México 52786, México.
| |
Collapse
|
88
|
Jung TW, Park HS, Choi GH, Kim D, Ahn SH, Kim DS, Lee T, Jeong JH. Maresin 1 attenuates pro-inflammatory reactions and ER stress in HUVECs via PPARα-mediated pathway. Mol Cell Biochem 2018; 448:335-347. [DOI: 10.1007/s11010-018-3392-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
|
89
|
Gibson MS, Domingues N, Vieira OV. Lipid and Non-lipid Factors Affecting Macrophage Dysfunction and Inflammation in Atherosclerosis. Front Physiol 2018; 9:654. [PMID: 29997514 PMCID: PMC6029489 DOI: 10.3389/fphys.2018.00654] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/14/2018] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease and a leading cause of human mortality. The lesional microenvironment contains a complex accumulation of variably oxidized lipids and cytokines. Infiltrating monocytes become polarized in response to these stimuli, resulting in a broad spectrum of macrophage phenotypes. The extent of lipid loading in macrophages influences their phenotype and consequently their inflammatory status. In response to excess atherogenic ligands, many normal cell processes become aberrant following a loss of homeostasis. This can have a direct impact upon the inflammatory response, and conversely inflammation can lead to cell dysfunction. Clear evidence for this exists in the lysosomes, endoplasmic reticulum and mitochondria of atherosclerotic macrophages, the principal lesional cell type. Furthermore, several intrinsic cell processes become dysregulated under lipidotic conditions. Therapeutic strategies aimed at restoring cell function under disease conditions are an ongoing coveted aim. Macrophages play a central role in promoting lesional inflammation, with plaque progression and stability being directly proportional to macrophage abundance. Understanding how mixtures or individual lipid species regulate macrophage biology is therefore a major area of atherosclerosis research. In this review, we will discuss how the myriad of lipid and lipoprotein classes and products used to model atherogenic, proinflammatory immune responses has facilitated a greater understanding of some of the intricacies of chronic inflammation and cell function. Despite this, lipid oxidation produces a complex mixture of products and with no single or standard method of derivatization, there exists some variation in the reported effects of certain oxidized lipids. Likewise, differences in the methods used to generate macrophages in vitro may also lead to variable responses when apparently identical lipid ligands are used. Consequently, the complexity of reported macrophage phenotypes has implications for our understanding of the metabolic pathways, processes and shifts underpinning their activation and inflammatory status. Using oxidized low density lipoproteins and its oxidized cholesteryl esters and phospholipid constituents to stimulate macrophage has been hugely valuable, however there is now an argument that only working with low complexity lipid species can deliver the most useful information to guide therapies aimed at controlling atherosclerosis and cardiovascular complications.
Collapse
Affiliation(s)
- Mark S Gibson
- Lysosomes in Chronic Human Pathologies and Infection, Faculdade de Ciências Médicas, Centro de Estudos de Doenças Crónicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Neuza Domingues
- Lysosomes in Chronic Human Pathologies and Infection, Faculdade de Ciências Médicas, Centro de Estudos de Doenças Crónicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Otilia V Vieira
- Lysosomes in Chronic Human Pathologies and Infection, Faculdade de Ciências Médicas, Centro de Estudos de Doenças Crónicas, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
90
|
Ardic S, Gumrukcu A, Gonenc Cekic O, Erdem M, Reis Kose GD, Demir S, Kose B, Yulug E, Mentese A, Turedi S. The value of endoplasmic reticulum stress markers (GRP78 and CHOP) in the diagnosis of acute mesenteric ischemia. Am J Emerg Med 2018; 37:596-602. [PMID: 29958740 DOI: 10.1016/j.ajem.2018.06.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022] Open
Abstract
AIM To evaluate levels of the endoplasmic reticulum (ER) stress markers GRP78 and CHOP in acute mesenteric ischemia (AMI) and to examine relations with degrees of AMI-related intestinal injury. MATERIALS AND METHODS Twenty-four rats were divided into four groups. Group I and Group III represented the control groups, from which blood and tissue specimens were collected 2 and 6 h after laparotomy without superior mesenteric artery (SMA) ligation. Group II and Group IV constituted the ischemia groups, from which blood and tissue specimens were collected 2 and 6 h after SMA ligation. The ER stress markers GRP78 and CHOP, total oxidant status (TOS), total antioxidant status (TAS), and the oxidative stress index (OSI) were investigated in each group. Ileum specimens were assessed in terms of ischemic injury, and appropriate comparisons were performed. RESULTS Significantly higher GRP78, CHOP, TOS, and TAS values were determined in the ischemia groups (groups II and IV) compared to the control groups (groups I and III). This elevation was greater in the 6 h ischemia group, the group exposed to the greatest ischemic injury (Group IV). Significant and powerful correlation was present between histopathological damage and levels of the ER stress markers and oxidative markers. CONCLUSION According to our results, ER stress markers (GRP78 and CHOP) increase significantly following ischemic injury. This elevation has the potential to be used diagnostically and also in prognostic terms due to the powerful correlation it exhibits with AMI-related ischemic injury.
Collapse
Affiliation(s)
- Senol Ardic
- University of Health Science, Faculty of Medicine, Department of Emergency Medicine, Trabzon, Turkey
| | - Aysegul Gumrukcu
- University of Health Science, Faculty of Medicine, Department of Emergency Medicine, Trabzon, Turkey
| | - Ozgen Gonenc Cekic
- University of Health Science, Faculty of Medicine, Department of Emergency Medicine, Trabzon, Turkey
| | - Mehmet Erdem
- Karadeniz Technical University, Faculty of Medicine, Department of Medical Biochemistry, Trabzon, Turkey
| | - Goksen Derya Reis Kose
- Karadeniz Technical University, Faculty of Medicine, Department of Histology and Embryology, Trabzon, Turkey
| | - Selim Demir
- Karadeniz Technical University, Faculty of Health Sciences, Department of Nutrition and Dietetics, Trabzon, Turkey
| | - Bestami Kose
- University of Health Science, Faculty of Medicine, Department of Emergency Medicine, Trabzon, Turkey
| | - Esin Yulug
- Karadeniz Technical University, Faculty of Medicine, Department of Histology and Embryology, Trabzon, Turkey
| | - Ahmet Mentese
- Karadeniz Technical University, Vocational School of Health Sciences, Program of Medical Laboratory Techniques, Trabzon, Turkey
| | - Suleyman Turedi
- Karadeniz Technical University, Faculty of Medicine, Department of Emergency Medicine, Trabzon, Turkey.
| |
Collapse
|
91
|
Varghese JF, Patel R, Yadav UCS. Novel Insights in the Metabolic Syndrome-induced Oxidative Stress and Inflammation-mediated Atherosclerosis. Curr Cardiol Rev 2018; 14:4-14. [PMID: 28990536 PMCID: PMC5872260 DOI: 10.2174/1573403x13666171009112250] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/09/2017] [Accepted: 09/28/2017] [Indexed: 02/06/2023] Open
Abstract
Context: Atherosclerosis is a progressive pathological process and a leading cause of mor-tality worldwide. Clinical research and epidemiological studies state that atherosclerosis is caused by an amalgamation of metabolic and inflammatory deregulation involving three important pathological events including Endothelial Dysfunction (ED), Foam Cell Formation (FCF), and Vascular Smooth Muscle Cells (VSMCs) proliferation and migration. Objectives: Research in recent years has identified Metabolic Syndrome (MS), which involves factors such as obesity, insulin resistance, dyslipidemia and diabetes, to be responsible for the pathophysiol-ogy of atherosclerosis. These factors elevate oxidative stress and inflammation-induced key signalling molecules and various microRNAs (miRs). In present study, we have reviewed recently identified molecular targets in the pathophysiology of atherosclerosis. Methods: Scientific literature obtained from databases such as university library, PubMed and Google along with evidences from published experimental work in relevant journals has been sum-marized in this review article. Results: The molecular events and cell signalling implicated in atherogenic processes of ED, FCF and VSMCs hyperplasia are sequential and progressive, and involve cross talks at many levels. Specific molecules such as transcription factors, inflammatory cytokines and chemokines and miRs have been identified playing crucial role in most of the events leading to atherosclerosis. Conclusion: Studies associated with MS induced oxidative stress- and inflammation- mediated sig-nalling pathways along with critical miRs help in better understanding of the pathophysiology of ath-erosclerosis. Several key molecules discussed in this review could be potent target for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Johnna F Varghese
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat - 382030, India
| | - Rohit Patel
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat - 382030, India
| | - Umesh C S Yadav
- School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat - 382030, India
| |
Collapse
|
92
|
Leznicki P, Natarajan J, Bader G, Spevak W, Schlattl A, Abdul Rehman SA, Pathak D, Weidlich S, Zoephel A, Bordone MC, Barbosa-Morais NL, Boehmelt G, Kulathu Y. Expansion of DUB functionality generated by alternative isoforms - USP35, a case study. J Cell Sci 2018; 131:jcs.212753. [PMID: 29685892 DOI: 10.1242/jcs.212753] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/01/2018] [Indexed: 12/12/2022] Open
Abstract
Protein ubiquitylation is a dynamic post-translational modification that can be reversed by deubiquitylating enzymes (DUBs). It is unclear how the small number (∼100) of DUBs present in mammalian cells regulate the thousands of different ubiquitylation events. Here, we analysed annotated transcripts of human DUBs and found ∼300 ribosome-associated transcripts annotated as protein coding, which thus increases the total number of DUBs. By using USP35, a poorly studied DUB, as a case study, we provide evidence that alternative isoforms contribute to the functional expansion of DUBs. We show that there are two different USP35 isoforms that localise to different intracellular compartments and have distinct functions. Our results reveal that isoform 1 is an anti-apoptotic factor that inhibits staurosporine- and TNF-related apoptosis-inducing ligand (TRAIL; also known as TNFSF10)-induced apoptosis. In contrast, USP35 isoform 2 is an integral membrane protein of the endoplasmic reticulum (ER) that is also present at lipid droplets. Manipulations of isoform 2 levels cause rapid ER stress, likely through deregulation of lipid homeostasis, and lead to cell death. Our work highlights how alternative isoforms provide functional expansion of DUBs and sets directions for future research.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Pawel Leznicki
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Jayaprakash Natarajan
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Gerd Bader
- Boehringer Ingelheim RCV GmbH & Co KG, Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Walter Spevak
- Boehringer Ingelheim RCV GmbH & Co KG, Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Andreas Schlattl
- Boehringer Ingelheim RCV GmbH & Co KG, Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Syed Arif Abdul Rehman
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Deepika Pathak
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Simone Weidlich
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Andreas Zoephel
- Boehringer Ingelheim RCV GmbH & Co KG, Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Marie C Bordone
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Nuno L Barbosa-Morais
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Guido Boehmelt
- Boehringer Ingelheim RCV GmbH & Co KG, Dr. Boehringer Gasse 5-11, 1120 Vienna, Austria
| | - Yogesh Kulathu
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
93
|
Emerging roles of endoplasmic reticulum-resident selenoproteins in the regulation of cellular stress responses and the implications for metabolic disease. Biochem J 2018; 475:1037-1057. [PMID: 29559580 DOI: 10.1042/bcj20170920] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 12/25/2022]
Abstract
Chronic metabolic stress leads to cellular dysfunction, characterized by excessive reactive oxygen species, endoplasmic reticulum (ER) stress and inflammation, which has been implicated in the pathogenesis of obesity, type 2 diabetes and cardiovascular disease. The ER is gaining recognition as a key organelle in integrating cellular stress responses. ER homeostasis is tightly regulated by a complex antioxidant system, which includes the seven ER-resident selenoproteins - 15 kDa selenoprotein, type 2 iodothyronine deiodinase and selenoproteins S, N, K, M and T. Here, the findings from biochemical, cell-based and mouse studies investigating the function of ER-resident selenoproteins are reviewed. Human experimental and genetic studies are drawn upon to highlight the relevance of these selenoproteins to the pathogenesis of metabolic disease. ER-resident selenoproteins have discrete roles in the regulation of oxidative, ER and inflammatory stress responses, as well as intracellular calcium homeostasis. To date, only two of these ER-resident selenoproteins, selenoproteins S and N have been implicated in human disease. Nonetheless, the potential of all seven ER-resident selenoproteins to ameliorate metabolic dysfunction warrants further investigation.
Collapse
|
94
|
Shibasaki Y, Horikawa M, Ikegami K, Kiuchi R, Takeda M, Hiraide T, Morita Y, Konno H, Takeuchi H, Setou M, Sakaguchi T. Stearate-to-palmitate ratio modulates endoplasmic reticulum stress and cell apoptosis in non-B non-C hepatoma cells. Cancer Sci 2018; 109:1110-1120. [PMID: 29427339 PMCID: PMC5891190 DOI: 10.1111/cas.13529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/25/2018] [Accepted: 02/02/2018] [Indexed: 12/29/2022] Open
Abstract
The increased prevalence of hepatocellular carcinoma (HCC) without viral infection, namely, NHCC, is a major public health issue worldwide. NHCC is frequently derived from non‐alcoholic fatty liver (NAFL) and non‐alcoholic steatohepatitis, which exhibit dysregulated fatty acid (FA) metabolism. This raises the possibility that NHCC evolves intracellular machineries to adapt to dysregulated FA metabolism. We herein aim to identify NHCC‐specifically altered FA and key molecules to achieve the adaptation. To analyze FA, imaging mass spectrometry (IMS) was performed on 15 HCC specimens. The composition of saturated FA (SFA) in NHCC was altered from that in typical HCC. The stearate‐to‐palmitate ratio (SPR) was significantly increased in NHCC. Associated with the SPR increase, the ELOVL6 protein level was upregulated in NHCC. The knockdown of ELOVL6 reduced SPR, and enhanced endoplasmic reticulum stress, inducing apoptosis of Huh7 and HepG2 cells. In conclusion, NHCC appears to adapt to an FA‐rich environment by modulating SPR through ELOVL6.
Collapse
Affiliation(s)
- Yasushi Shibasaki
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Makoto Horikawa
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.,International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Koji Ikegami
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.,International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Ryota Kiuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Makoto Takeda
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takanori Hiraide
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yoshifumi Morita
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hiroyuki Konno
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hiroya Takeuchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Mitsutoshi Setou
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan.,International Mass Imaging Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takanori Sakaguchi
- Second Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
95
|
Su SA, Xie Y, Fu Z, Wang Y, Wang JA, Xiang M. Emerging role of exosome-mediated intercellular communication in vascular remodeling. Oncotarget 2018; 8:25700-25712. [PMID: 28147325 PMCID: PMC5421963 DOI: 10.18632/oncotarget.14878] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 01/18/2017] [Indexed: 12/17/2022] Open
Abstract
Vascular remodeling refers to the alternations of function and structure in vasculature. A complex autocrine/paracrine set of cellular interaction is involved in vascular remodeling. Exosome, a newly identified natural nanocarrier and intercellular messenger, plays a pivotal role in regulating cell-to-cell communication. Exosome emerges as an important mediator in the process of vascular remodeling, showing the most prognostic and therapeutic potent in vascular diseases. Benefiting from exosomal trafficking, the vasculature can not only maintain its function and structure in physiological condition, but also adapt itself in pathological status. In this review, we will represent the roles of exosomes in angiogenesis, endothelial function and cardiac regeneration. In addition, greatly depending on the pathophysiological status of donor cells and peripheral micro-circumstance, the exosomal content could alter, which makes exosomes exhibit pleiotropic effects in vascular diseases. Hence, the diverse effects of exosomes in vascular diseases including atherosclerosis, neointima formation and vascular repair, primary hypertension, pulmonary artery hypertension, and aortic aneurysm will be discussed. Finally, the translational appliances targeting exosomes will be concluded by providing updated applications of engineered exosomes in clinic.
Collapse
Affiliation(s)
- Sheng-An Su
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Yao Xie
- Cardiovascular Division, King's College London BHF Center, London, United Kingdom
| | - Zurong Fu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Yaping Wang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Jian-An Wang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Meixiang Xiang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| |
Collapse
|
96
|
Park SH, Kang MK, Choi YJ, Kim YH, Antika LD, Kim DY, Lee EJ, Lim SS, Kang YH. α-Asarone blocks 7β-hydroxycholesterol-exposed macrophage injury through blocking elF2α phosphorylation and prompting beclin-1-dependent autophagy. Oncotarget 2018; 8:7370-7383. [PMID: 28088783 PMCID: PMC5352328 DOI: 10.18632/oncotarget.14566] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 01/02/2017] [Indexed: 12/27/2022] Open
Abstract
Macrophage apoptosis is salient in advanced atherosclerotic lesions and is induced by several stimuli including endoplasmic reticulum (ER) stress. This study examined that a-asarone present in purple perilla abrogated macrophage injury caused by oxysterols via ER stress- and autophagy-mediated mechanisms. Nontoxic a-asarone at 1-20 M attenuated 7β-hydroxycholesterol-induced activation of eukaryotic initiation factor 2a in macrophages leading to C/EBP homologous protein (CHOP) expression and apoptosis due to sustained ER stress. The a-asarone treatment increased the formation of autophagolysosomes localizing in perinuclear regions of 7β-hydroxycholesterol-exposed macrophages. Consistently, this compound promoted the induction of the key autophagic proteins of beclin-1, vacuolar protein sorting 34 and p150 responsible for vesicle nucleation, and prompted the conversion of microtubule-associated protein 1A/1B-light chain 3 and the induction of p62, neighbor of BRCA1 and autophagy-related (Atg) 12-Atg5-Atg16L conjugate involved in phagophore expansion and autophagosome formation. Additionally, a-asarone increased ER phosphorylation of bcl-2 facilitating beclin-1 entry to autophagic process. Furthermore, the deletion of Atg5 or beclin-1 gene enhanced apoptotic CHOP induction. Collectively, a-asarone-stimulated autophagy may be potential multi-targeted therapeutic avenues in treating ER stress-associated macrophage apoptosis.
Collapse
Affiliation(s)
- Sin-Hye Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Min-Kyung Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Yean-Jung Choi
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Yun-Ho Kim
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Lucia Dwi Antika
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Dong Yeon Kim
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Eun-Jung Lee
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Soon Sung Lim
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Korea
| |
Collapse
|
97
|
Moura-Assis A, Afonso MS, de Oliveira V, Morari J, dos Santos GA, Koike M, Lottenberg AM, Ramos Catharino R, Velloso LA, Sanchez Ramos da Silva A, de Moura LP, Ropelle ER, Pauli JR, Cintra DEC. Flaxseed oil rich in omega-3 protects aorta against inflammation and endoplasmic reticulum stress partially mediated by GPR120 receptor in obese, diabetic and dyslipidemic mice models. J Nutr Biochem 2018; 53:9-19. [DOI: 10.1016/j.jnutbio.2017.09.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 01/10/2023]
|
98
|
Mohammadi A, Vahabzadeh Z, Jamalzadeh S, Khalili T. Trimethylamine-N-oxide, as a risk factor for atherosclerosis, induces stress in J774A.1 murine macrophages. Adv Med Sci 2018; 63:57-63. [PMID: 28822264 DOI: 10.1016/j.advms.2017.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/17/2017] [Accepted: 06/28/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE Trimethylamine N-oxide (TMAO) is a biomarker for kidney problems. It has also been introduced as a risk factor for atherosclerosis. The classic risk factors for atherosclerosis trigger cellular and humeral immunoreaction in macrophages through induction of heat shock protein expressions and increased levels of GRP94 and HSP70 are associated with increased atherosclerosis risk. The present study evaluated the possible effect(s) of TMAO on the expression of GRP94 and HSP70 at protein levels. METHODS J774A.1 murine macrophages were treated with different micromolar concentrations of TMAO and 4-phenylbutyric acid (PBA), a chemical chaperone, for 8, 18, 24, and 48h intervals. Tunicamycin was also used as a control for induction of endoplasmic reticulum stress. Western blotting was used to evaluate the expression of GRP94 and HSP70 in macrophages at protein levels. RESULT Tunicamycin greatly increased protein levels of GRP94. Similarly, but to a lesser extent compared to tunicamycin, TMAO also increased GRP94. In 24h treated cells, only 300μM of TMAO, and in cells treated for 48h, all doses of TMAO produced a significant increase in relative HSP70 protein levels compared to the control. PBA failed to induce any changes in GRP94 or HSP70 protein levels. CONCLUSION GRP94 and HSP70 are stress-inducible heat shock protein, so the elevation in J774A.1 murine macrophages can clearly define cells under stress and elucidate the contribution of stress induced by TMAO that may have a part in the abnormal activation of macrophages involved in foam cell formation.
Collapse
Affiliation(s)
- Abbas Mohammadi
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Zakaria Vahabzadeh
- Liver & Digestive Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Soran Jamalzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Tahereh Khalili
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
99
|
Xue A, Lin J, Que C, Yu Y, Tu C, Chen H, Liu B, Zhao X, Wang T, Ma K, Li L. Aberrant endoplasmic reticulum stress mediates coronary artery spasm through regulating MLCK/MLC2 pathway. Exp Cell Res 2018; 363:321-331. [DOI: 10.1016/j.yexcr.2018.01.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/02/2018] [Accepted: 01/23/2018] [Indexed: 11/25/2022]
|
100
|
Miragem AA, Homem de Bittencourt PI. Nitric oxide-heat shock protein axis in menopausal hot flushes: neglected metabolic issues of chronic inflammatory diseases associated with deranged heat shock response. Hum Reprod Update 2018; 23:600-628. [PMID: 28903474 DOI: 10.1093/humupd/dmx020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/28/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although some unequivocal underlying mechanisms of menopausal hot flushes have been demonstrated in animal models, the paucity of similar approaches in humans impedes further mechanistic outcomes. Human studies might show some as yet unexpected physiological mechanisms of metabolic adaptation that permeate the phase of decreased oestrogen levels in both symptomatic and asymptomatic women. This is particularly relevant because both the severity and time span of hot flushes are associated with increased risk of chronic inflammatory disease. On the other hand, oestrogen induces the expression of heat shock proteins of the 70 kDa family (HSP70), which are anti-inflammatory and cytoprotective protein chaperones, whose expression is modulated by different types of physiologically stressful situations, including heat stress and exercise. Therefore, lower HSP70 expression secondary to oestrogen deficiency increases cardiovascular risk and predisposes the patient to senescence-associated secretory phenotype (SASP) that culminates in chronic inflammatory diseases, such as obesities, type 2 diabetes, neuromuscular and neurodegenerative diseases. OBJECTIVE AND RATIONALE This review focuses on HSP70 and its accompanying heat shock response (HSR), which is an anti-inflammatory and antisenescent pathway whose intracellular triggering is also oestrogen-dependent via nitric oxide (NO) production. The main goal of the manuscript was to show that the vasomotor symptoms that accompany hot flushes may be a disguised clue for important neuroendocrine alterations linking oestrogen deficiency to the anti-inflammatory HSR. SEARCH METHODS Results from our own group and recent evidence on hypothalamic control of central temperature guided a search on PubMed and Google Scholar websites. OUTCOMES Oestrogen elicits rapid production of the vasodilatory gas NO, a powerful activator of HSP70 expression. Whence, part of the protective effects of oestrogen over cardiovascular and neuroendocrine systems is tied to its capacity of inducing the NO-elicited HSR. The hypothalamic areas involved in thermoregulation (infundibular nucleus in humans and arcuate nucleus in other mammals) and whose neurons are known to have their function altered after long-term oestrogen ablation, particularly kisspeptin-neurokinin B-dynorphin neurons, (KNDy) are the same that drive neuroprotective expression of HSP70 and, in many cases, this response is via NO even in the absence of oestrogen. From thence, it is not illogical that hot flushes might be related to an evolutionary adaptation to re-equip the NO-HSP70 axis during the downfall of circulating oestrogen. WIDER IMPLICATIONS Understanding of HSR could shed light on yet uncovered mechanisms of menopause-associated diseases as well as on possible manipulation of HSR in menopausal women through physiological, pharmacological, nutraceutical and prebiotic interventions. Moreover, decreased HSR indices (that can be clinically determined with ease) in perimenopause could be of prognostic value in predicting the moment and appropriateness of starting a HRT.
Collapse
Affiliation(s)
- Antônio Azambuja Miragem
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil.,Federal Institute of Education, Science and Technology 'Farroupilha', Rua Uruguai 1675, Santa Rosa, RS 98900-000, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology, Department of Physiology, Federal University of Rio Grande do Sul, Rua Sarmento Leite 500, ICBS, 2nd Floor, Suite 350, Porto Alegre, RS 90050-170, Brazil
| |
Collapse
|