1
|
Byun JH, Lebeau PF, Trink J, Uppal N, Lanktree MB, Krepinsky JC, Austin RC. Endoplasmic reticulum stress as a driver and therapeutic target for kidney disease. Nat Rev Nephrol 2025; 21:299-313. [PMID: 39988577 DOI: 10.1038/s41581-025-00938-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
The endoplasmic reticulum (ER) has crucial roles in metabolically active cells, including protein translation, protein folding and quality control, lipid biosynthesis, and calcium homeostasis. Adverse metabolic conditions or pathogenic genetic variants that cause misfolding and accumulation of proteins within the ER of kidney cells initiate an injurious process known as ER stress that contributes to kidney disease and its cardiovascular complications. Initiation of ER stress activates the unfolded protein response (UPR), a cellular defence mechanism that functions to restore ER homeostasis. However, severe or chronic ER stress rewires the UPR to activate deleterious pathways that exacerbate inflammation, apoptosis and fibrosis, resulting in kidney injury. This insidious crosstalk between ER stress, UPR activation, oxidative stress and inflammation forms a vicious cycle that drives kidney disease and vascular damage. Furthermore, genetic variants that disrupt protein-folding mechanisms trigger ER stress, as evidenced in autosomal-dominant tubulointerstitial kidney disease and Fabry disease. Emerging therapeutic strategies that enhance protein-folding capacity and reduce the burden of ER stress have shown promising results in kidney diseases. Thus, integrating knowledge of how genetic variants cause protein misfolding and ER stress into clinical practice will enhance treatment strategies and potentially improve outcomes for various kidney diseases and their vascular complications.
Collapse
Affiliation(s)
- Jae Hyun Byun
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Paul F Lebeau
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Jackie Trink
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Nikhil Uppal
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine and Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Matthew B Lanktree
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine and Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Joan C Krepinsky
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, The Research Institute of St Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada.
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Xu S, Xu Y, Wang Z, Wei Z, Mei Y, Cao Y, Li B, Zhang H, Zhang Z. Endoplasmic reticulum stress causes long bone shortening in P4hb C402R/+ mice: A mouse model exhibiting significant features of cole-carpenter syndrome driven by P4HB mutations. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167663. [PMID: 39778777 DOI: 10.1016/j.bbadis.2025.167663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Cole-Carpenter syndrome (CCS) is a rare autosomal-dominant genetic disease characterized by craniosynostosis, ocular proptosis, hydrocephalus, distinctive facial features, and bone fragility. Previous cases of CCS are associated with genetic variations in P4HB, which encodes the protein disulfide isomerase (PDI), a key enzyme in protein folding. Patients with CCS caused by P4HB mutations often present with short stature, limb deformities, and abnormal epiphyseal plates. However, the underlying mechanisms are largely unknown. To investigate this, a mouse model expressing the P4hbC402R mutation (corresponding to P4HBC400R in humans) was generated. Although the mouse model did not exhibit craniofacial bone defects or brittle bone phenotypes, it did show significantly shortened long bones-a prominent characteristic of P4HB-induced CCS. This was due to impaired proliferation and delayed hypertrophy of growth plate chondrocytes. Mutant PDI was found to accumulate abnormally in the endoplasmic reticulum (ER), and in vitro experiments revealed defects in both the catalytic and chaperone activities of mutant PDI. In addition, we observed enhanced ER stress and activation of the PKR-like ER kinase (PERK) pathway in P4hbC402R/+ chondrocytes. Inhibition of ER stress mitigated PERK activation, alleviated defective chondrocyte proliferation and differentiation, thereby rescuing bone length. Taken together, enhanced ER stress and the activation of the PERK, potentially initiated by the malfunctioning of PDIC402R or its abnormal accumulation within the ER, or both, lead to compromised chondrocyte proliferation and differentiation in mice, and ultimately stunts mice growth. This provides new insights into the pathogenesis of P4HB-dominated CCS and offers potential therapeutic targets.
Collapse
Affiliation(s)
- Shuqin Xu
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Xu
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyuan Wang
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhanying Wei
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yazhao Mei
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangjia Cao
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China.
| | - Hao Zhang
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhenlin Zhang
- Shanghai Clinical Research Center of Bone Disease, Department of Osteoporosis and Bone Diseases, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Rodrigues BL, Pascoal LB, Genaro LM, Warrak LSCA, Rodrigues BAG, Coope A, Camargo MG, Oliveira PDSP, Ayrizono MDLS, Velloso LA, Leal RF. In Vitro Inhibition of Endoplasmic Reticulum Stress: A Promising Therapeutic Strategy for Patients with Crohn's Disease. Cells 2025; 14:270. [PMID: 39996742 PMCID: PMC11853800 DOI: 10.3390/cells14040270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Crohn's disease (CD) is an inflammatory bowel disease marked by an abnormal immune response and excessive pro-inflammatory cytokines, leading to impaired protein processing and endoplasmic reticulum (ER) stress. This stress, caused by the accumulation of misfolded proteins, triggers the unfolded protein response (UPR) through IRE1/Xbp-1, PERK/eIF2α, and ATF6 pathways, which are linked to intestinal inflammation. This study aimed to investigate ER stress in CD patients' intestinal mucosa and evaluate phenylbutyrate (PBA) as an ER stress inhibitor. METHODS Colon biopsies from CD patients and controls were cultured under five conditions, including 4-PBA treatments. Real-time PCR, cytokine level, and immunohistochemistry were performed. RESULTS Immunohistochemistry revealed that ER stress was activated in CD patients' intestinal epithelial cells and lamina propria cells. PERK/eIF2α, but not IRE1/Xbp-1 or ATF6, was upregulated in CD patients compared to controls. UPR-related genes (STC2, CALR, HSPA5, HSP90B1) were also elevated in CD patients. PBA treatment significantly reduced ER stress and UPR markers while decreasing apoptotic markers like DDIT3. Pro-inflammatory cytokines, such as IL-1β, IL-6, IL-17, TNF- α, and sCD40L, were significantly reduced after PBA treatment. CONCLUSION ER stress and UPR pathways are activated in CD colonic mucosa, and PBA reduces these markers, suggesting potential therapeutic benefits for CD-related inflammation.
Collapse
Affiliation(s)
- Bruno Lima Rodrigues
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lívia Bitencourt Pascoal
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lívia Moreira Genaro
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Leonardo Saint Clair Assad Warrak
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Beatriz Alves Guerra Rodrigues
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Andressa Coope
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Michel Gardere Camargo
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Priscilla de Sene Portel Oliveira
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-864, Brazil
| | - Raquel Franco Leal
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| |
Collapse
|
4
|
Yamazaki A, Omura I, Kamikawa Y, Hide M, Tanaka A, Kaneko M, Imaizumi K, Saito A. Unfolded protein response modulates Tyrosinase levels and melanin production during melanogenesis. J Dermatol Sci 2025; 117:36-44. [PMID: 39818444 DOI: 10.1016/j.jdermsci.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/09/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Melanocytes protect the body from ultraviolet radiation by synthesizing melanin. Tyrosinase, a key enzyme in melanin production, accumulates in the endoplasmic reticulum (ER) during melanin synthesis, potentially causing ER stress. However, regulating ER function for melanin synthesis has been less studied than controlling Tyrosinase activity. OBJECTIVE This study investigates the regulatory mechanisms of melanin production, focusing on ER stress and the ER stress-induced response. METHODS B16 mouse melanoma cells induced to undergo melanogenesis were treated with unfolded protein response (UPR) inhibitors or chemical chaperones, and their effects on melanogenesis were analyzed. RESULTS During melanogenesis in B16 cells stimulated by alpha-melanocyte-stimulating hormone (α-MSH), ER stress and UPR activation occurred, accompanied by increased Tyrosinase protein. Reducing IRE1 and ATF6 branch activity lowered melanin levels, while chemical chaperone treatment restored melanin production and increased Tyrosinase levels. CONCLUSION UPR activation, linked to elevated Tyrosinase levels, influences melanin production during melanogenesis. Modulating UPR can regulate melanin synthesis and provides a potential new approach for treating pigmentation disorders.
Collapse
Affiliation(s)
- Akari Yamazaki
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Issei Omura
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Frontier Science and Interdisciplinary Research, Faculty of Medicine, Kanazawa University, Ishikawa, Japan
| | - Yasunao Kamikawa
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Dermatology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masayuki Kaneko
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Atsushi Saito
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Frontier Science and Interdisciplinary Research, Faculty of Medicine, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
5
|
Ma K, Zhang Y, Zhao J, Zhou L, Li M. Endoplasmic reticulum stress: bridging inflammation and obesity-associated adipose tissue. Front Immunol 2024; 15:1381227. [PMID: 38638434 PMCID: PMC11024263 DOI: 10.3389/fimmu.2024.1381227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
Obesity presents a significant global health challenge, increasing the susceptibility to chronic conditions such as diabetes, cardiovascular disease, and hypertension. Within the context of obesity, lipid metabolism, adipose tissue formation, and inflammation are intricately linked to endoplasmic reticulum stress (ERS). ERS modulates metabolism, insulin signaling, inflammation, as well as cell proliferation and death through the unfolded protein response (UPR) pathway. Serving as a crucial nexus, ERS bridges the functionality of adipose tissue and the inflammatory response. In this review, we comprehensively elucidate the mechanisms by which ERS impacts adipose tissue function and inflammation in obesity, aiming to offer insights into targeting ERS for ameliorating metabolic dysregulation in obesity-associated chronic diseases such as hyperlipidemia, hypertension, fatty liver, and type 2 diabetes.
Collapse
Affiliation(s)
| | | | | | | | - Min Li
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Al-Daghestani H, Qaisar R, Al Kawas S, Ghani N, Rani KGA, Azeem M, Hasnan HK, Kassim NK, Samsudin AR. Pharmacological inhibition of endoplasmic reticulum stress mitigates osteoporosis in a mouse model of hindlimb suspension. Sci Rep 2024; 14:4719. [PMID: 38413677 PMCID: PMC10899598 DOI: 10.1038/s41598-024-54944-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/19/2024] [Indexed: 02/29/2024] Open
Abstract
Hindlimb suspension (HLS) mice exhibit osteoporosis of the hindlimb bones and may be an excellent model to test pharmacological interventions. We investigated the effects of inhibiting endoplasmic reticulum (ER) stress with 4-phenyl butyrate (4-PBA) on the morphology, physicochemical properties, and bone turnover markers of hindlimbs in HLS mice. We randomly divided 21 male C57BL/6J mice into three groups, ground-based controls, untreated HLS group and 4-PBA treated group (HLS+4PBA) (100mg/kg/day, intraperitoneal) for 21 days. We investigated histopathology, micro-CT imaging, Raman spectroscopic analysis, and gene expression. Untreated HLS mice exhibited reduced osteocyte density, multinucleated osteoclast-like cells, adipocyte infiltration, and reduced trabecular striations on micro-CT than the control group. Raman spectroscopy revealed higher levels of ER stress, hydroxyproline, non-collagenous proteins, phenylalanine, tyrosine, and CH2Wag as well as a reduction in proteoglycans and adenine. Furthermore, bone alkaline phosphatase and osteocalcin were downregulated, while Cathepsin K, TRAP, and sclerostin were upregulated. Treatment with 4-PBA partially restored normal bone histology, increased collagen crosslinking, and mineralization, promoted anti-inflammatory markers, and downregulated bone resorption markers. Our findings suggest that mitigating ER stress with 4-PBA could be a therapeutic intervention to offset osteoporosis in conditions mimicking hindlimb suspension.
Collapse
Affiliation(s)
- Hiba Al-Daghestani
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE
- Space Medicine Research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Sausan Al Kawas
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, 27272, UAE
| | - Nurhafizah Ghani
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - K G Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Muhammad Azeem
- Department of Mathematical and Physical Sciences, University of Nizwa, Nizwa 33, Sultanate of Oman
| | - Hijaz Kamal Hasnan
- Department of Geology, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Nur Karyatee Kassim
- School of Dental Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia.
| | - A R Samsudin
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah, 27272, UAE.
| |
Collapse
|
7
|
Wi D, Park CY. 1,25-dihydroxyvitamin D 3 affects thapsigargin-induced endoplasmic reticulum stress in 3T3-L1 adipocytes. Nutr Res Pract 2024; 18:1-18. [PMID: 38352211 PMCID: PMC10861344 DOI: 10.4162/nrp.2024.18.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/01/2023] [Accepted: 12/06/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Endoplasmic reticulum (ER) stress in adipose tissue causes an inflammatory response and leads to metabolic diseases. However, the association between vitamin D and adipose ER stress remains poorly understood. In this study, we investigated whether 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) alleviates ER stress in adipocytes. MATERIALS/METHODS 3T3-L1 cells were treated with different concentrations (i.e., 10-100 nM) of 1,25(OH)2D3 after or during differentiation (i.e., on day 0-7, 3-7, or 7). They were then incubated with thapsigargin (TG, 500 nM) for an additional 24 h to induce ER stress. Next, we measured the mRNA and protein levels of genes involved in unfold protein response (UPR) and adipogenesis using real-time polymerase chain reaction and western blotting and quantified the secreted protein levels of pro-inflammatory cytokines. Finally, the mRNA levels of UPR pathway genes were measured in adipocytes transfected with siRNA-targeting Vdr. RESULTS Treatment with 1,25(OH)2D3 during various stages of adipocyte differentiation significantly inhibited ER stress induced by TG. In fully differentiated 3T3-L1 adipocytes, 1,25(OH)2D3 treatment suppressed mRNA levels of Ddit3, sXbp1, and Atf4 and decreased the secretion of monocyte chemoattractant protein-1, interleukin-6, and tumor necrosis factor-α. However, downregulation of the mRNA levels of Ddit3, sXbp1, and Atf4 following 1,25(OH)2D3 administration was not observed in Vdr-knockdown adipocytes. In addition, exposure of 3T3-L1 preadipocytes to 1,25(OH)2D3 inhibited transcription of Ddit3, sXbp1, Atf4, Bip, and Atf6 and reduced the p-alpha subunit of translation initiation factor 2 (eIF2α)/eIF2α and p-protein kinase RNA-like ER kinase (PERK)/PERK protein ratios. Furthermore, 1,25(OH)2D3 treatment before adipocyte differentiation reduced adipogenesis and the mRNA levels of adipogenic genes. CONCLUSIONS Our data suggest that 1,25(OH)2D3 prevents TG-induced ER stress and inflammatory responses in mature adipocytes by downregulating UPR signaling via binding with Vdr. In addition, the inhibition of adipogenesis by vitamin D may contribute to the reduction of ER stress in adipocytes.
Collapse
Affiliation(s)
- Dain Wi
- Department of Food & Nutrition, College of Health Science, The University of Suwon, Hwaseong 18323, Korea
| | - Chan Yoon Park
- Department of Food & Nutrition, College of Health Science, The University of Suwon, Hwaseong 18323, Korea
| |
Collapse
|
8
|
Commey KL, Enaka A, Nakamura R, Yamamoto A, Tsukigawa K, Nishi K, Iohara D, Hirayama F, Otagiri M, Yamasaki K. Development of α-Cyclodextrin-Based Orally Disintegrating Tablets for 4-Phenylbutyrate. Pharmaceutics 2024; 16:82. [PMID: 38258093 PMCID: PMC10818935 DOI: 10.3390/pharmaceutics16010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Despite major improvements brought about by the introduction of taste-masked formulations of 4-phenylbutyrate (PB), poor compliance remains a significant drawback to treatment for some pediatric and dysphagic patients with urea cycle disorders (UCDs). This study reports on the development of a cyclodextrin (CD)-based orally disintegrating tablet (ODT) formulation for PB as an alternative to existing formulations. This is based on previous reports of the PB taste-masking potential of CDs and the suitability of ODTs for improving compliance in pediatric and dysphagic populations. In preliminary studies, the interactions of PB with α and βCD in the solid state were characterized using X-ray diffraction, scanning electron microscopy, dissolution, and accelerated stability studies. Based on these studies, lyophilized PB-CD solid systems were formulated into ODTs after wet granulation. Evaluation of the ODTs showed that they had adequate physical characteristics, including hardness and friability and good storage stability. Notably, the developed αCD-based ODT for PB had a disintegration time of 28 s and achieved a slightly acidic and agreeable pH (≈5.5) in solution, which is suitable for effective PB-CD complexation and taste masking. The developed formulation could be helpful as an alternative to existing PB formulations, especially for pediatric and dysphagic UCD patients.
Collapse
Affiliation(s)
- Kindness L. Commey
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Airi Enaka
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
| | - Ryota Nakamura
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
| | - Asami Yamamoto
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
| | - Kenji Tsukigawa
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Koji Nishi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Daisuke Iohara
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Fumitoshi Hirayama
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.E.); (R.N.); (A.Y.); (K.T.); (K.N.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| |
Collapse
|
9
|
Wang LY, Li J, Gong B, Wang RH, Chen YL, Yin J, Yang C, Lin JT, Liu HZ, Yang Y, Li J, Li C, Yao N. Orosomucoid proteins limit endoplasmic reticulum stress in plants. THE NEW PHYTOLOGIST 2023; 240:1134-1148. [PMID: 37606093 DOI: 10.1111/nph.19200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/17/2023] [Indexed: 08/23/2023]
Abstract
Sphingolipids are cell membrane components and signaling molecules that induce endoplasmic reticulum (ER) stress responses, but the underlying mechanism is unknown. Orosomucoid proteins (ORMs) negatively regulate serine palmitoyltransferase activity, thus helping maintain proper sphingolipid levels in humans, yeast, and plants. In this report, we explored the roles of ORMs in regulating ER stress in Arabidopsis thaliana. Loss of ORM1 and ORM2 function caused constitutive activation of the unfolded protein response (UPR), as did treatment with the ceramide synthase inhibitor Fumonisin B1 (FB1) or ceramides. FB1 treatment induced the transcription factor bZIP28 to relocate from the ER membrane to the nucleus. The transcription factor WRKY75 positively regulates the UPR and physically interacted with bZIP28. We also found that the orm mutants showed impaired ER-associated degradation (ERAD), blocking the degradation of misfolded MILDEW RESISTANCE LOCUS-O 12 (MLO-12). ORM1 and ORM2 bind to EMS-MUTAGENIZED BRI1 SUPPRESSOR 7 (EBS7), a plant-specific component of the Arabidopsis ERAD complex, and regulate its stability. These data strongly suggest that ORMs in the ER membrane play vital roles in the UPR and ERAD pathways to prevent ER stress in Arabidopsis. Our results reveal that ORMs coordinate sphingolipid homeostasis with ER quality control and play a role in stress responses.
Collapse
Affiliation(s)
- Ling-Yan Wang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Jian Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, China
| | - Benqiang Gong
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Rui-Hua Wang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yi-Li Chen
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Jian Yin
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Chang Yang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Jia-Ting Lin
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Hao-Zhuo Liu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yubing Yang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Jianfeng Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Chunyu Li
- Institution of Fruit Tree Research, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Nan Yao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| |
Collapse
|
10
|
Delmotte P, Yap JQ, Dasgupta D, Sieck GC. Chemical Chaperone 4-PBA Mitigates Tumor Necrosis Factor Alpha-Induced Endoplasmic Reticulum Stress in Human Airway Smooth Muscle. Int J Mol Sci 2023; 24:15816. [PMID: 37958799 PMCID: PMC10649207 DOI: 10.3390/ijms242115816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
Airway inflammation and pro-inflammatory cytokines such as tumor necrosis factor alpha (TNFα) underlie the pathophysiology of respiratory diseases, including asthma. Previously, we showed that TNFα activates the inositol-requiring enzyme 1α (IRE1α)/X-box binding protein 1 spliced (XBP1s) endoplasmic reticulum (ER) stress pathway in human airway smooth muscle (hASM) cells. The ER stress pathway is activated by the accumulation of unfolded proteins in the ER. Accordingly, chemical chaperones such as 4-phenylbutyric acid (4-PBA) may reduce ER stress activation. In the present study, we hypothesized that chemical chaperone 4-PBA mitigates TNFα-induced ER stress in hASM cells. hASM cells were isolated from bronchiolar tissue obtained from five patients with no history of smoking or respiratory diseases. The hASM cells' phenotype was confirmed via the expression of alpha-smooth muscle actin and elongated morphology. hASM cells from the same patient sample were then separated into three 12 h treatment groups: (1) TNFα (20 ng/mL), (2) TNFα + 4-PBA (1 μM, 30 min pretreatment), and (3) untreated control. The expressions of total IRE1α and phosphorylated IRE1α (pIRE1αS724) were determined through Western blotting. The splicing of XBP1 mRNA was analyzed using RT-PCR. We found that TNFα induced an increase in pIRE1αS724 phosphorylation, which was mitigated by treatment with chemical chaperone 4-PBA. We also found that TNFα induced an increase in XBP1s mRNA, which was also mitigated by treatment with chemical chaperone 4-PBA. These results support our hypothesis and indicate that chemical chaperone 4-PBA treatment mitigates TNFα-induced ER stress in hASM cells.
Collapse
Affiliation(s)
| | | | | | - Gary C. Sieck
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (P.D.); (J.Q.Y.); (D.D.)
| |
Collapse
|
11
|
Commey KL, Nakatake A, Enaka A, Nakamura R, Nishi K, Tsukigawa K, Ikeda H, Yamaguchi K, Iohara D, Hirayama F, Yamasaki K, Otagiri M. Study of the Structural Chemistry of the Inclusion Complexation of 4-Phenylbutyrate and Related Compounds with Cyclodextrins in Solution: Differences in Inclusion Mode with Cavity Size Dependency. Int J Mol Sci 2023; 24:15091. [PMID: 37894771 PMCID: PMC10606765 DOI: 10.3390/ijms242015091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
4-phenylbutyrate (PB) and structurally related compounds hold promise for treating many diseases, including cancers. However, pharmaceutical limitations, such as an unpleasant taste or poor aqueous solubility, impede their evaluation and clinical use. This study explores cyclodextrin (CD) complexation as a strategy to address these limitations. The structural chemistry of the CD complexes of these compounds was analyzed using phase solubility, nuclear magnetic resonance (NMR) spectroscopic techniques, and molecular modeling to inform the choice of CD for such application. The study revealed that PB and its shorter-chain derivative form 1:1 αCD complexes, while the longer-chain derivatives form 1:2 (guest:host) complexes. αCD includes the alkyl chain of the shorter-chain compounds, depositing the phenyl ring around its secondary rim, whereas two αCD molecules sandwich the phenyl ring in a secondary-to-secondary rim orientation for the longer-chain derivatives. βCD includes each compound to form 1:1 complexes, with their alkyl chains bent to varying degrees within the CD cavity. γCD includes two molecules of each compound to form 2:1 complexes, with both parallel and antiparallel orientations plausible. The study found that αCD is more suitable for overcoming the pharmaceutical drawbacks of PB and its shorter-chain derivative, while βCD is better for the longer-chain derivatives.
Collapse
Affiliation(s)
- Kindness L. Commey
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
| | - Akari Nakatake
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
| | - Airi Enaka
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
| | - Ryota Nakamura
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
| | - Koji Nishi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Kenji Tsukigawa
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Hirohito Ikeda
- Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Jonan-ku, Fukuoka 814-0180, Japan;
| | - Koki Yamaguchi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Daisuke Iohara
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Fumitoshi Hirayama
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; (K.L.C.); (A.N.); (A.E.); (R.N.); (K.N.); (K.T.); (K.Y.); (D.I.); (F.H.)
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| |
Collapse
|
12
|
Chen Q, Thompson J, Hu Y, Lesnefsky EJ. Endoplasmic reticulum stress and alterations of peroxiredoxins in aged hearts. Mech Ageing Dev 2023; 215:111859. [PMID: 37661065 PMCID: PMC11103240 DOI: 10.1016/j.mad.2023.111859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/20/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Aging-related cardiovascular disease is influenced by multiple factors, with oxidative stress being a key contributor. Aging-induced endoplasmic reticulum (ER) stress exacerbates oxidative stress by impairing mitochondrial function. Furthermore, a decline in antioxidants, including peroxiredoxins (PRDXs), augments the oxidative stress during aging. To explore if ER stress leads to PRDX degradation during aging, young adult (3 mo.) and aged (24 mo.) male mice were studied. Treatment with 4-phenylbutyrate (4-PBA) was used to alleviate ER stress in young adult and aged mice. Aged hearts showed elevated oxidative stress levels compared to young hearts. However, treatment with 4-PBA to attenuate ER stress reduced oxidative stress in aged hearts, indicating that ER stress contributes to increased oxidative stress in aging. Moreover, aging resulted in reduced levels of peroxiredoxin 3 (PRDX3) in mitochondria and peroxiredoxin 4 (PRDX4) in myocardium. While 4-PBA treatment improved PRDX3 content in aged hearts, it did not restore PRDX4 content in aged mice. These findings suggest that ER stress not only leads to mitochondrial dysfunction and increased oxidant stress but also impairs a vital antioxidant defense through decreased PRDX3 content. Additionally, the results suggest that PRDX4 may contribute an upstream role in inducing ER stress during aging.
Collapse
Affiliation(s)
- Qun Chen
- Departments of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jeremy Thompson
- Departments of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ying Hu
- Departments of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Edward J Lesnefsky
- Departments of Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, USA; Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA; Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298, USA; Richmond Department of Veterans Affairs Medical Center, Richmond, VA 23249, USA.
| |
Collapse
|
13
|
Yeap JW, Ali IAH, Ibrahim B, Tan ML. Chronic obstructive pulmonary disease and emerging ER stress-related therapeutic targets. Pulm Pharmacol Ther 2023; 81:102218. [PMID: 37201652 DOI: 10.1016/j.pupt.2023.102218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023]
Abstract
COPD pathogenesis is frequently associated with endoplasmic reticulum stress (ER stress) progression. Targeting the major unfolded protein response (UPR) branches in the ER stress pathway may provide pharmacotherapeutic selection strategies for treating COPD and enable relief from its symptoms. In this study, we aimed to systematically review the potential role of the ER stress inhibitors of major UPR branches (IRE1, PERK, and ATF6) in COPD-related studies and determine the current stage of knowledge in this field. The systematic review was carried out adhering to the PRISMA checklist based on published studies obtained from specific keyword searches of three databases, namely PubMed, ScienceDirect and Springer Database. The search was limited to the year 2000-2022 which includes all in vitro studies, in vivo studies and clinical trials related to the application of ER stress inhibitors toward COPD-induced models and disease. The risk of bias was evaluated using the QUIN, SYRCLE, revised Cochrane risk of bias tool for randomized trials (RoB 2.0) and NIH tool respectively. A total of 7828 articles were screened from three databases and a final total of 37 studies were included in the review. The ER stress and UPR pathways are potentially useful to prevent COPD progression and attenuate the exacerbation of COPD and related symptoms. Interestingly, the off-target effects from inhibition of the UPR pathway may be desirable or undesirable depending on context and therapeutic applications. Targeting the UPR pathway could have complex consequences as the production of ER molecules involved in folding may be impaired which could continuously provoke misfolding of proteins. Although several emerging compounds were noted to be potentially useful for targeted therapy against COPD, clinical studies have yet to be thoroughly explored.
Collapse
Affiliation(s)
- Jia Wen Yeap
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Pulau Pinang, Malaysia
| | - Irfhan Ali Hyder Ali
- Respiratory Department, Penang General Hospital, Jalan Residensi, 10990, Pulau Pinang, Malaysia
| | - Baharudin Ibrahim
- Department of Clinical Pharmacy & Pharmacy Practice, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mei Lan Tan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Pulau Pinang, Malaysia; Centre For Global Sustainability Studies (CGSS), Universiti Sains Malaysia, 11800, Pulau Pinang, Malaysia.
| |
Collapse
|
14
|
Ranade A, Khan AA, Gul MT, Suresh S, Qaisar R, Ahmad F, Karim A. Suppression of endoplasmic reticulum stress reverses hindlimb unloading-induced hepatic cellular processes in mice. Biochim Biophys Acta Gen Subj 2023:130422. [PMID: 37406741 DOI: 10.1016/j.bbagen.2023.130422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND The Hindlimb unloaded mouse, an animal model of simulated microgravity demonstrates significant metabolic and hepatic derangements. However, cellular and molecular mechanisms driving liver dysfunction in Hindlimb unloaded mice are poorly characterized. METHODS We investigated the possible contribution of dysregulated protein homeostasis by endoplasmic reticulum, endoplasmic reticulum stress, to liver dysfunction during HU. C57BL/6j male mice were grouped into ground-based controls or Hindlimb unloaded groups treated daily with vehicle or 4-phenylbutyrate (4-PBA), a potent inhibitor of endoplasmic reticulum stress. Following three weeks of HU, mice were sacrificed, and liver tissues were dissected for further analysis. RESULTS Hindlimb unloaded was associated with hepatic atrophy and elevated endoplasmic reticulum stress, which was restored by 4-PBA treatment. The Gene Ontology analysis revealed the downregulation of genes primarily involved in liver metabolic and Wingless-related integration site (WNT) signaling pathways, while those related to cytochrome P450, and liver fibrosis were upregulated. The Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed downregulation of several genes involved in metabolic pathways following treatment with 4-PBA, induced by HU. CONCLUSIONS We report several differential and uniquely expressed genes associated with microgravity-induced elevated ER stress and liver injury. Our data has translational potential in unraveling novel molecular targets for pharmaceutical therapies of liver diseases. GENERAL SIGNIFICANCE Our novel findings show a pathogenic role for elevated ER stress in liver injury in microgravity conditions.
Collapse
Affiliation(s)
- Anu Ranade
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Amir Ali Khan
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Muhammad Tehsil Gul
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Savitha Suresh
- Iron Biology Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates
| | - Asima Karim
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Iron Biology Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
15
|
Zhang Q, Chen Y, Wang Q, Wang Y, Feng W, Chai L, Liu J, Li D, Chen H, Qiu Y, Shen N, Shi X, Xie X, Li M. HMGB1-induced activation of ER stress contributes to pulmonary artery hypertension in vitro and in vivo. Respir Res 2023; 24:149. [PMID: 37268944 DOI: 10.1186/s12931-023-02454-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/18/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND HMGB1 and ER stress have been considered to participate in the progression of pulmonary artery hypertension (PAH). However, the molecular mechanism underlying HMGB1 and ER stress in PAH remains unclear. This study aims to explore whether HMGB1 induces pulmonary artery smooth muscle cells (PASMCs) functions and pulmonary artery remodeling through ER stress activation. METHODS Primary cultured PASMCs and monocrotaline (MCT)-induced PAH rats were applied in this study. Cell proliferation and migration were determined by CCK-8, EdU and transwell assay. Western blotting was conducted to detect the protein levels of protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor-4 (ATF4), seven in absentia homolog 2 (SIAH2) and homeodomain interacting protein kinase 2 (HIPK2). Hemodynamic measurements, immunohistochemistry staining, hematoxylin and eosin staining were used to evaluate the development of PAH. The ultrastructure of ER was observed by transmission electron microscopy. RESULTS In primary cultured PASMCs, HMGB1 reduced HIPK2 expression through upregulation of ER stress-related proteins (PERK and ATF4) and subsequently increased SIAH2 expression, which ultimately led to PASMC proliferation and migration. In MCT-induced PAH rats, interfering with HMGB1 by glycyrrhizin, suppression of ER stress by 4-phenylbutyric acid or targeting SIAH2 by vitamin K3 attenuated the development of PAH. Additionally, tetramethylpyrazine (TMP), as a component of traditional Chinese herbal medicine, reversed hemodynamic deterioration and vascular remodeling by targeting PERK/ATF4/SIAH2/HIPK2 axis. CONCLUSIONS The present study provides a novel insight to understand the pathogenesis of PAH and suggests that targeting HMGB1/PERK/ATF4/SIAH2/HIPK2 cascade might have potential therapeutic value for the prevention and treatment of PAH.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xiangyu Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
16
|
Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, Tian Y, Fu X. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (Beijing) 2023; 4:e283. [PMID: 37303813 PMCID: PMC10248034 DOI: 10.1002/mco2.283] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents one of the fastest growing epidemic metabolic disorders worldwide and is a strong contributor for a broad range of comorbidities, including vascular, visual, neurological, kidney, and liver diseases. Moreover, recent data suggest a mutual interplay between T2DM and Corona Virus Disease 2019 (COVID-19). T2DM is characterized by insulin resistance (IR) and pancreatic β cell dysfunction. Pioneering discoveries throughout the past few decades have established notable links between signaling pathways and T2DM pathogenesis and therapy. Importantly, a number of signaling pathways substantially control the advancement of core pathological changes in T2DM, including IR and β cell dysfunction, as well as additional pathogenic disturbances. Accordingly, an improved understanding of these signaling pathways sheds light on tractable targets and strategies for developing and repurposing critical therapies to treat T2DM and its complications. In this review, we provide a brief overview of the history of T2DM and signaling pathways, and offer a systematic update on the role and mechanism of key signaling pathways underlying the onset, development, and progression of T2DM. In this content, we also summarize current therapeutic drugs/agents associated with signaling pathways for the treatment of T2DM and its complications, and discuss some implications and directions to the future of this field.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Huimin Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yu Zhang
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Geng Liu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Haixia Xu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Guocheng Rao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
17
|
Ajoolabady A, Lebeaupin C, Wu NN, Kaufman RJ, Ren J. ER stress and inflammation crosstalk in obesity. Med Res Rev 2023; 43:5-30. [PMID: 35975736 DOI: 10.1002/med.21921] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/07/2022] [Accepted: 07/20/2022] [Indexed: 02/04/2023]
Abstract
The endoplasmic reticulum (ER) governs the proper folding of polypeptides and proteins through various chaperones and enzymes residing within the ER organelle. Perturbation in the ER folding process ensues when overwhelmed protein folding exceeds the ER handling capacity, leading to the accumulation of misfolded/unfolded proteins in the ER lumen-a state being referred to as ER stress. In turn, ER stress induces a gamut of signaling cascades, termed as the "unfolded protein response" (UPR) that reinstates the ER homeostasis through a panel of gene expression modulation. This type of UPR is usually deemed "adaptive UPR." However, persistent or unresolved ER stress hyperactivates UPR response, which ultimately, triggers cell death and inflammatory pathways, termed as "maladaptive/terminal UPR." A plethora of evidence indicates that crosstalks between ER stress (maladaptive UPR) and inflammation precipitate obesity pathogenesis. In this regard, the acquisition of the mechanisms linking ER stress to inflammation in obesity might unveil potential remedies to tackle this pathological condition. Herein, we aim to elucidate key mechanisms of ER stress-induced inflammation in the context of obesity and summarize potential therapeutic strategies in the management of obesity through maneuvering ER stress and ER stress-associated inflammation.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Cardiology and Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cynthia Lebeaupin
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Ne N Wu
- Department of Cardiology and Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Jun Ren
- Department of Cardiology and Shanghai Institute for Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Epstein-Barr virus-induced gene 3 commits human mesenchymal stem cells to differentiate into chondrocytes via endoplasmic reticulum stress sensor. PLoS One 2022; 17:e0279584. [PMID: 36548354 PMCID: PMC9778607 DOI: 10.1371/journal.pone.0279584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 12/11/2022] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSC) can differentiate into chondrocytes. Epstein-Barr virus-induced gene 3 (EBI3) is differentially expressed during chondrogenic differentiation and can be produced by MSC. EBI3 is also a subunit of interleukin (IL)-27 and IL-35, and it accumulates in the endoplasmic reticulum (ER) when its partners, such as IL-27 p28 and IL-35 p35, are insufficient. ER stress induced by protein accumulation is responsible for chondrogenic differentiation. However, the role of EBI3 and its relevance to the ER stress in chondrogenic differentiation of MSC have never been addressed. Here, we demonstrate that EBI3 protein is expressed in the early stage of chondrogenic differentiation of MSC. Additionally, knockdown, overexpression, or induction of EBI3 through IL-1β inhibits chondrogenesis. We show that EBI3 localizes and accumulates in the ER of MSC after overexpression or induction by IL-1β and TNF-α, whereas ER stress inhibitor 4-phenylbutyric acid decreases its accumulation in MSC. Moreover, EBI3 modulates ER stress sensor inositol-requiring enzyme 1 α (IRE1α) after induced by IL-1β, and MSC-like cells coexpress EBI3 and IRE1α in rheumatoid arthritis (RA) synovial tissue. Altogether, these data demonstrate that intracellular EBI3 commits to chondrogenic differentiation by regulating ER stress sensor IRE1α.
Collapse
|
19
|
4-PBA Attenuates Fat Accumulation in Cultured Spotted Seabass Fed High-Fat-Diet via Regulating Endoplasmic Reticulum Stress. Metabolites 2022; 12:metabo12121197. [PMID: 36557235 PMCID: PMC9784988 DOI: 10.3390/metabo12121197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/13/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Excessive fat accumulation is a common phenomenon in cultured fish, which can cause metabolic disease such as fatty liver. However, the relative regulatory approach remains to be explored. Based on this, two feeding trials were conducted. Firstly, fish were fed either a normal-fat diet (NFD) or a high-fat diet (HFD) for eight weeks and sampled at the 2nd, 4th, 6th, and 8th week after feeding (Experiment I). In the first four weeks, fish fed an HFD grew faster than those fed an NFD. Conversely, the body weight and weight gain were higher in the NFD group at the 6th and 8th weeks. Under light and transmission electron microscopes, fat accumulation of the liver was accompanied by an obvious endoplasmic reticulum (ER) swell. Accordingly, the expressions of atf-6, ire-1, perk, eif-2α, atf-4, grp78, and chop showed that ER stress was activated at the 6th and 8th weeks. In Experiment II, 50 mg/kg 4-PBA (an ERs inhibitor) was supplemented to an HFD; this was named the 4-PBA group. Then, fish was fed with an NFD, an HFD, and a 4-PBA diet for eight weeks. As the result, the excessive fat deposition caused by an HFD was reversed by 4-PBA. The expression of ER stress-related proteins CHOP and GRP78 was down-regulated by 4-PBA, and the transmission electron microscope images also showed that 4-PBA alleviated ER stress induced by the feeding of an HFD. Furthermore, 4-PBA administration down-regulated SREBP-1C/ACC/FAS, the critical pathways of fat synthesis. In conclusion, the results confirmed that ER stress plays a contributor role in the fat deposition by activating the SREBP-1C/ACC/FAS pathway. 4-PBA as an ER stress inhibitor could reduce fat deposition caused by an HFD via regulating ER stress.
Collapse
|
20
|
Zhou R, He M, Fan J, Li R, Zuo Y, Li B, Gao G, Sun T. The role of hypothalamic endoplasmic reticulum stress in schizophrenia and antipsychotic-induced weight gain: A narrative review. Front Neurosci 2022; 16:947295. [PMID: 36188456 PMCID: PMC9523121 DOI: 10.3389/fnins.2022.947295] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022] Open
Abstract
Schizophrenia (SCZ) is a serious mental illness that affects 1% of people worldwide. SCZ is associated with a higher risk of developing metabolic disorders such as obesity. Antipsychotics are the main treatment for SCZ, but their side effects include significant weight gain/obesity. Despite extensive research, the underlying mechanisms by which SCZ and antipsychotic treatment induce weight gain/obesity remain unclear. Hypothalamic endoplasmic reticulum (ER) stress is one of the most important pathways that modulates inflammation, neuronal function, and energy balance. This review aimed to investigate the role of hypothalamic ER stress in SCZ and antipsychotic-induced weight gain/obesity. Preliminary evidence indicates that SCZ is associated with reduced dopamine D2 receptor (DRD2) signaling, which significantly regulates the ER stress pathway, suggesting the importance of ER stress in SCZ and its related metabolic disorders. Antipsychotics such as olanzapine activate ER stress in hypothalamic neurons. These effects may induce decreased proopiomelanocortin (POMC) processing, increased neuropeptide Y (NPY) and agouti-related protein (AgRP) expression, autophagy, and leptin and insulin resistance, resulting in hyperphagia, decreased energy expenditure, and central inflammation, thereby causing weight gain. By activating ER stress, antipsychotics such as olanzapine activate hypothalamic astrocytes and Toll-like receptor 4 signaling, thereby causing inflammation and weight gain/obesity. Moreover, evidence suggests that antipsychotic-induced ER stress may be related to their antagonistic effects on neurotransmitter receptors such as DRD2 and the histamine H1 receptor. Taken together, ER stress inhibitors could be a potential effective intervention against SCZ and antipsychotic-induced weight gain and inflammation.
Collapse
Affiliation(s)
- Ruqin Zhou
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- *Correspondence: Meng He,
| | - Jun Fan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Ruoxi Li
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufeng Zuo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Benben Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
- Guanbin Gao,
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- Taolei Sun,
| |
Collapse
|
21
|
Izadi MS, Eskandari F, Binayi F, Salimi M, Rashidi FS, Hedayati M, Dargahi L, Ghanbarian H, Zardooz H. Oxidative and endoplasmic reticulum stress develop adverse metabolic effects due to the high-fat high-fructose diet consumption from birth to young adulthood. Life Sci 2022; 309:120924. [PMID: 36063978 DOI: 10.1016/j.lfs.2022.120924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 11/27/2022]
Abstract
AIMS The early postnatal dietary intake has been considered a crucial factor affecting the offspring later life metabolic status. Consistently, this study investigated the oxidative and endoplasmic reticulum (ER) stress intervention in the induction of adverse metabolic effects due to the high-fat high-fructose diet (HFHFD) consumption from birth to young adulthood in rat offspring. MATERIALS AND METHODS After delivery, the dams with their pups were randomly allocated into the normal diet (ND) and HFHFD groups. At weaning, the male offspring were divided into ND-None, ND-DMSO, ND-4-phenyl butyric acid (4-PBA), HFHFD-None, HFHFD-DMSO, and HFHFD-4-PBA groups and fed on their respected diets for five weeks. Then, the drug was injected for ten days. Subsequently, glucose and lipid metabolism parameters, oxidative and ER stress markers, and Wolfram syndrome1 (Wfs1) expression were assessed. KEY FINDINGS In the HFHFD group, anthropometrical parameters, plasma high-density lipoprotein (HDL), and glucose-stimulated insulin secretion and content were decreased. Whereas, the levels of plasma leptin, low-density lipoprotein (LDL) and glucose, hypothalamic leptin, pancreatic catalase activity and glutathione (GSH), pancreatic and hypothalamic malondialdehyde (MDA), binding immunoglobulin protein (BIP) and C/EBP homologous protein (CHOP), and pancreatic WFS1 protein were increased. 4-PBA administration in the HFHFD group, decreased the hypothalamic and pancreatic MDA, BIP and CHOP levels. While, increased the Insulin mRNA and glucose-stimulated insulin secretion and content. SIGNIFICANCE HFHFD intake from birth to young adulthood through the development of pancreatic and hypothalamic oxidative and ER stress, increased the pancreatic WFS1 protein and impaired glucose and lipid homeostasis in male rat offspring.
Collapse
Affiliation(s)
- Mina Sadat Izadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Eskandari
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Salimi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadat Rashidi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Turishcheva E, Vildanova M, Onishchenko G, Smirnova E. The Role of Endoplasmic Reticulum Stress in Differentiation of Cells of Mesenchymal Origin. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:916-931. [PMID: 36180988 PMCID: PMC9483250 DOI: 10.1134/s000629792209005x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 05/23/2023]
Abstract
Endoplasmic reticulum (ER) is a multifunctional membrane-enclosed organelle. One of the major ER functions is cotranslational transport and processing of secretory, lysosomal, and transmembrane proteins. Impaired protein processing caused by disturbances in the ER homeostasis results in the ER stress. Restoration of normal ER functioning requires activation of an adaptive mechanism involving cell response to misfolded proteins, the so-called unfolded protein response (UPR). Besides controlling protein folding, UPR plays a key role in other physiological processes, in particular, differentiation of cells of connective, muscle, epithelial, and neural tissues. Cell differentiation is induced by the physiological levels of ER stress, while excessive ER stress suppresses differentiation and can result in cell death. So far, it remains unknown whether UPR activation induces cell differentiation or if UPR is initiated by the upregulated synthesis of secretory proteins during cell differentiation. Cell differentiation is an important stage in the development of multicellular organisms and is tightly controlled. Suppression or excessive activation of this process can lead to the development of various pathologies in an organism. In particular, impairments in the differentiation of connective tissue cells can result in the development of fibrosis, obesity, and osteoporosis. Recently, special attention has been paid to fibrosis as one of the major complications of COVID-19. Therefore, studying the role of UPR in the activation of cell differentiation is of both theoretical and practical interest, as it might result in the identification of molecular targets for selective regulation of cell differentiation stages and as well as the potential to modulate the mechanisms involved in the development of various pathological states.
Collapse
Affiliation(s)
| | - Mariya Vildanova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Galina Onishchenko
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Elena Smirnova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
23
|
Lee ES, Aryal YP, Kim TY, Kim JY, Yamamoto H, An CH, An SY, Lee Y, Sohn WJ, Jung JK, Ha JH, Kim JY. Facilitation of Reparative Dentin Using a Drug Repositioning Approach With 4-Phenylbutric Acid. Front Physiol 2022; 13:885593. [PMID: 35600310 PMCID: PMC9114641 DOI: 10.3389/fphys.2022.885593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
For hard tissue formation, cellular mechanisms, involved in protein folding, processing, and secretion play important roles in the endoplasmic reticulum (ER). In pathological and regeneration conditions, ER stress hinders proper formation and secretion of proteins, and tissue regeneration by unfolded protein synthesis. 4-Phenylbutyric acid (4PBA) is a chemical chaperone that alleviates ER stress through modulation in proteins folding and protein trafficking. However, previous studies about 4PBA only focused on the metabolic diseases rather than on hard tissue formation and regeneration. Herein, we evaluated the function of 4PBA in dentin regeneration using an exposed pulp animal model system via a local delivery method as a drug repositioning strategy. Our results showed altered morphological changes and cellular physiology with histology and immunohistochemistry. The 4PBA treatment modulated the inflammation reaction and resolved ER stress in the early stage of pulp exposure. In addition, 4PBA treatment activated blood vessel formation and TGF-β1 expression in the dentin-pulp complex. Micro-computed tomography and histological examinations confirmed the facilitated formation of the dentin bridge in the 4PBA-treated specimens. These results suggest that proper modulation of ER stress would be an important factor for secretion and patterned formation in dentin regeneration.
Collapse
Affiliation(s)
- Eui-Seon Lee
- Department of Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
| | - Yam Prasad Aryal
- Department of Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
| | - Tae-Young Kim
- Department of Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
| | - Ji-Youn Kim
- Department of Dental Hygiene, Gachon University, Incheon, South Korea
| | - Hitoshi Yamamoto
- Department of Histology and Developmental Biology, Tokyo Dental College, Tokyo, Japan
| | - Chang-Hyeon An
- Department of Oral and Maxillofacial Radiology, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
| | - Seo-Young An
- Department of Oral and Maxillofacial Radiology, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
| | - Youngkyun Lee
- Department of Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
| | - Wern-Joo Sohn
- Pre-Major of Cosmetics and Pharmaceutics, Daegu Haany University, Gyeongsan, South Korea
| | - Jae-Kwang Jung
- Department of Oral Medicine, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
| | - Jung-Hong Ha
- Department of Conservative Dentistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, South Korea
- *Correspondence: Jae-Young Kim, , orcid.org/0000-0002-6752-5683; Jung-Hong Ha, , orcid.org/0000-0002-0469-4324
| | - Jae-Young Kim
- Department of Biochemistry, School of Dentistry, IHBR, Kyungpook National University, Daegu, Korea
- *Correspondence: Jae-Young Kim, , orcid.org/0000-0002-6752-5683; Jung-Hong Ha, , orcid.org/0000-0002-0469-4324
| |
Collapse
|
24
|
Park TJ, Park SY, Lee HJ, Abd El-Aty A, Jeong JH, Jung TW. α-ketoisocaproic acid promotes ER stress through impairment of autophagy, thereby provoking lipid accumulation and insulin resistance in murine preadipocytes. Biochem Biophys Res Commun 2022; 603:109-115. [DOI: 10.1016/j.bbrc.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 01/03/2023]
|
25
|
Chen Q, Thompson J, Hu Y, Lesnefsky EJ. Reversing mitochondrial defects in aged hearts: role of mitochondrial calpain activation. Am J Physiol Cell Physiol 2022; 322:C296-C310. [PMID: 35044856 PMCID: PMC8836732 DOI: 10.1152/ajpcell.00279.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/03/2023]
Abstract
Aging chronically increases endoplasmic reticulum (ER) stress that contributes to mitochondrial dysfunction. Activation of calpain 1 (CPN1) impairs mitochondrial function during acute ER stress. We proposed that aging-induced ER stress led to mitochondrial dysfunction by activating CPN1. We posit that attenuation of the ER stress or direct inhibition of CPN1 in aged hearts can decrease cardiac injury during ischemia-reperfusion by improving mitochondrial function. Male young (3 mo) and aged mice (24 mo) were used in the present study, and 4-phenylbutyrate (4-PBA) was used to decrease the ER stress in aged mice. Subsarcolemmal (SSM) and interfibrillar mitochondria (IFM) were isolated. Chronic 4-PBA treatment for 2 wk decreased CPN1 activation as shown by the decreased cleavage of spectrin in cytosol and apoptosis inducing factor (AIF) and the α1 subunit of pyruvate dehydrogenase (PDH) in mitochondria. Treatment improved oxidative phosphorylation in 24-mo-old SSM and IFM at baseline compared with vehicle. When 4-PBA-treated 24-mo-old hearts were subjected to ischemia-reperfusion, infarct size was decreased. These results support that attenuation of the ER stress decreased cardiac injury in aged hearts by improving mitochondrial function before ischemia. To challenge the role of CPN1 as an effector of the ER stress, aged mice were treated with MDL-28170 (MDL, an inhibitor of calpain 1). MDL treatment improved mitochondrial function in aged SSM and IFM. MDL-treated 24-mo-old hearts sustained less cardiac injury following ischemia-reperfusion. These results support that age-induced ER stress augments cardiac injury during ischemia-reperfusion by impairing mitochondrial function through activation of CPN1.
Collapse
Affiliation(s)
- Qun Chen
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jeremy Thompson
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Ying Hu
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Division of Cardiology, Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia
- McGuire Department of Veterans Affairs Medical Center, Richmond, Virginia
| |
Collapse
|
26
|
Wang T, Zhou J, Zhang X, Wu Y, Jin K, Wang Y, Xu R, Yang G, Li W, Jiao L. X-box Binding Protein 1: An Adaptor in the Pathogenesis of Atherosclerosis. Aging Dis 2022; 14:350-369. [PMID: 37008067 PMCID: PMC10017146 DOI: 10.14336/ad.2022.0824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis (AS), the formation of fibrofatty lesions in the vessel wall, is the primary cause of heart disease and stroke and is closely associated with aging. Disrupted metabolic homeostasis is a primary feature of AS and leads to endoplasmic reticulum (ER) stress, which is an abnormal accumulation of unfolded proteins. By orchestrating signaling cascades of the unfolded protein response (UPR), ER stress functions as a double-edged sword in AS, where adaptive UPR triggers synthetic metabolic processes to restore homeostasis, whereas the maladaptive response programs the cell to the apoptotic pathway. However, little is known regarding their precise coordination. Herein, an advanced understanding of the role of UPR in the pathological process of AS is reviewed. In particular, we focused on a critical mediator of the UPR, X-box binding protein 1 (XBP1), and its important role in balancing adaptive and maladaptive responses. The XBP1 mRNA is processed from the unspliced isoform (XBP1u) to the spliced isoform of XBP1 (XBP1s). Compared with XBP1u, XBP1s predominantly functions downstream of inositol-requiring enzyme-1α (IRE1α) and transcript genes involved in protein quality control, inflammation, lipid metabolism, carbohydrate metabolism, and calcification, which are critical for the pathogenesis of AS. Thus, the IRE1α/XBP1 axis is a promising pharmaceutical candidate against AS.
Collapse
Affiliation(s)
- Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Jia Zhou
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.
- Correspondence should be addressed to: Dr. Ge Yang, Chinese Academy of Sciences, Beijing, China. , Dr. Wenjing Li, Chinese Academy of Sciences, Beijing, China. ; Dr. Liqun Jiao, Xuanwu Hospital, Capital Medical University, Beijing, China. .
| | - Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.
- Correspondence should be addressed to: Dr. Ge Yang, Chinese Academy of Sciences, Beijing, China. , Dr. Wenjing Li, Chinese Academy of Sciences, Beijing, China. ; Dr. Liqun Jiao, Xuanwu Hospital, Capital Medical University, Beijing, China. .
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Correspondence should be addressed to: Dr. Ge Yang, Chinese Academy of Sciences, Beijing, China. , Dr. Wenjing Li, Chinese Academy of Sciences, Beijing, China. ; Dr. Liqun Jiao, Xuanwu Hospital, Capital Medical University, Beijing, China. .
| |
Collapse
|
27
|
Ajoolabady A, Wang S, Kroemer G, Klionsky DJ, Uversky VN, Sowers JR, Aslkhodapasandhokmabad H, Bi Y, Ge J, Ren J. ER Stress in Cardiometabolic Diseases: From Molecular Mechanisms to Therapeutics. Endocr Rev 2021; 42:839-871. [PMID: 33693711 DOI: 10.1210/endrev/bnab006] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Indexed: 02/08/2023]
Abstract
The endoplasmic reticulum (ER) hosts linear polypeptides and fosters natural folding of proteins through ER-residing chaperones and enzymes. Failure of the ER to align and compose proper protein architecture leads to accumulation of misfolded/unfolded proteins in the ER lumen, which disturbs ER homeostasis to provoke ER stress. Presence of ER stress initiates the cytoprotective unfolded protein response (UPR) to restore ER homeostasis or instigates a rather maladaptive UPR to promote cell death. Although a wide array of cellular processes such as persistent autophagy, dysregulated mitophagy, and secretion of proinflammatory cytokines may contribute to the onset and progression of cardiometabolic diseases, it is well perceived that ER stress also evokes the onset and development of cardiometabolic diseases, particularly cardiovascular diseases (CVDs), diabetes mellitus, obesity, and chronic kidney disease (CKD). Meanwhile, these pathological conditions further aggravate ER stress, creating a rather vicious cycle. Here in this review, we aimed at summarizing and updating the available information on ER stress in CVDs, diabetes mellitus, obesity, and CKD, hoping to offer novel insights for the management of these cardiometabolic comorbidities through regulation of ER stress.
Collapse
Affiliation(s)
- Amir Ajoolabady
- University of Wyoming College of Health Sciences, Laramie, Wyoming 82071, USA
| | - Shuyi Wang
- University of Wyoming College of Health Sciences, Laramie, Wyoming 82071, USA
- School of Medicine Shanghai University, Shanghai 200444, China
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida 33612, USA
| | - James R Sowers
- Dalton and Diabetes and Cardiovascular Center, University of Missouri Columbia, Columbia, Missouri 65212, USA
| | | | - Yaguang Bi
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Jun Ren
- University of Wyoming College of Health Sciences, Laramie, Wyoming 82071, USA
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
28
|
The Unfolded Protein Response as a Guardian of the Secretory Pathway. Cells 2021; 10:cells10112965. [PMID: 34831188 PMCID: PMC8616143 DOI: 10.3390/cells10112965] [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: 09/08/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is the major site of membrane biogenesis in most eukaryotic cells. As the entry point to the secretory pathway, it handles more than 10,000 different secretory and membrane proteins. The insertion of proteins into the membrane, their folding, and ER exit are affected by the lipid composition of the ER membrane and its collective membrane stiffness. The ER is also a hotspot of lipid biosynthesis including sterols, glycerophospholipids, ceramides and neural storage lipids. The unfolded protein response (UPR) bears an evolutionary conserved, dual sensitivity to both protein-folding imbalances in the ER lumen and aberrant compositions of the ER membrane, referred to as lipid bilayer stress (LBS). Through transcriptional and non-transcriptional mechanisms, the UPR upregulates the protein folding capacity of the ER and balances the production of proteins and lipids to maintain a functional secretory pathway. In this review, we discuss how UPR transducers sense unfolded proteins and LBS with a particular focus on their role as guardians of the secretory pathway.
Collapse
|
29
|
Adsi H, Levkovich SA, Haimov E, Kreiser T, Meli M, Engel H, Simhaev L, Karidi-Heller S, Colombo G, Gazit E, Laor Bar-Yosef D. Chemical Chaperones Modulate the Formation of Metabolite Assemblies. Int J Mol Sci 2021; 22:9172. [PMID: 34502079 PMCID: PMC8431448 DOI: 10.3390/ijms22179172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
The formation of amyloid-like structures by metabolites is associated with several inborn errors of metabolism (IEMs). These structures display most of the biological, chemical and physical properties of protein amyloids. However, the molecular interactions underlying the assembly remain elusive, and so far, no modulating therapeutic agents are available for clinical use. Chemical chaperones are known to inhibit protein and peptide amyloid formation and stabilize misfolded enzymes. Here, we provide an in-depth characterization of the inhibitory effect of osmolytes and hydrophobic chemical chaperones on metabolite assemblies, thus extending their functional repertoire. We applied a combined in vivo-in vitro-in silico approach and show their ability to inhibit metabolite amyloid-induced toxicity and reduce cellular amyloid content in yeast. We further used various biophysical techniques demonstrating direct inhibition of adenine self-assembly and alteration of fibril morphology by chemical chaperones. Using a scaffold-based approach, we analyzed the physiochemical properties of various dimethyl sulfoxide derivatives and their role in inhibiting metabolite self-assembly. Lastly, we employed whole-atom molecular dynamics simulations to elucidate the role of hydrogen bonds in osmolyte inhibition. Our results imply a dual mode of action of chemical chaperones as IEMs therapeutics, that could be implemented in the rational design of novel lead-like molecules.
Collapse
Affiliation(s)
- Hanaa Adsi
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (H.A.); (S.A.L.); (T.K.)
| | - Shon A. Levkovich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (H.A.); (S.A.L.); (T.K.)
| | - Elvira Haimov
- BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (E.H.); (H.E.); (L.S.)
| | - Topaz Kreiser
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (H.A.); (S.A.L.); (T.K.)
| | | | - Hamutal Engel
- BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (E.H.); (H.E.); (L.S.)
| | - Luba Simhaev
- BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (E.H.); (H.E.); (L.S.)
| | - Shai Karidi-Heller
- The Future Scientists Center–Alpha Program at Tel Aviv Youth University, Tel Aviv 6997801, Israel;
| | - Giorgio Colombo
- SCITEC-CNR, via Mario Bianco 9, 20131 Milano, Italy; (M.M.); (G.C.)
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Ehud Gazit
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (H.A.); (S.A.L.); (T.K.)
- BLAVATNIK CENTER for Drug Discovery, Tel Aviv University, Tel Aviv 6997801, Israel; (E.H.); (H.E.); (L.S.)
- Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dana Laor Bar-Yosef
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (H.A.); (S.A.L.); (T.K.)
| |
Collapse
|
30
|
Hartwick Bjorkman S, Oliveira Pereira R. The Interplay Between Mitochondrial Reactive Oxygen Species, Endoplasmic Reticulum Stress, and Nrf2 Signaling in Cardiometabolic Health. Antioxid Redox Signal 2021; 35:252-269. [PMID: 33599550 PMCID: PMC8262388 DOI: 10.1089/ars.2020.8220] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Mitochondria-derived reactive oxygen species (mtROS) are by-products of normal physiology that may disrupt cellular redox homeostasis on a regular basis. Nonetheless, failure to resolve sustained mitochondrial stress to mitigate high levels of mtROS might contribute to the etiology of numerous pathological conditions, such as obesity, insulin resistance, and cardiovascular disease (CVD). Recent Advances: Notably, recent studies have demonstrated that moderate mitochondrial stress might result in the induction of different stress response pathways that ultimately improve the organism's ability to deal with subsequent stress, a process termed mitohormesis. mtROS have been shown to play a key role in regulating this adaptation. Critical Issue: mtROS regulate the convergence of different signaling pathways that, when disturbed, might impair cardiometabolic health. Conversely, mtROS seem to be required to mediate activation of prosurvival pathways, contributing to improved cardiometabolic fitness. In the present review, we will primarily focus on the role of mtROS in the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant pathway and examine the role of endoplasmic reticulum (ER) stress in coordinating the convergence of ER stress and oxidative stress signaling through activation of Nrf2 and activating transcription factor 4 (ATF4). Future Directions: The mechanisms underlying cardiometabolic protection in response to mitochondrial stress have only started to be investigated. Integrated understanding of how mtROS and ER stress cooperatively promote activation of prosurvival pathways might shed mechanistic insight into the role of mitohormesis in mediating cardiometabolic protection and might inform future therapeutic avenues for the treatment of metabolic diseases contributing to CVD. Antioxid. Redox Signal. 35, 252-269.
Collapse
Affiliation(s)
- Sarah Hartwick Bjorkman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Renata Oliveira Pereira
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
31
|
Guzmán Mendoza NA, Homma K, Osada H, Toda E, Ban N, Nagai N, Negishi K, Tsubota K, Ozawa Y. Neuroprotective Effect of 4-Phenylbutyric Acid against Photo-Stress in the Retina. Antioxidants (Basel) 2021; 10:1147. [PMID: 34356380 PMCID: PMC8301054 DOI: 10.3390/antiox10071147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Exposure to excessive visible light causes retinal degeneration and may influence the progression of retinal blinding diseases. However, there are currently no applied treatments. Here, we focused on endoplasmic reticulum (ER) stress, which can cause cellular degeneration and apoptosis in response to stress. We analyzed functional, histological, and molecular changes in the light-exposed retina and the effects of administering an ER-stress inhibitor, 4-phenylbutyric acid (4-PBA), in mice. We found that light-induced visual function impairment related to photoreceptor cell loss and outer segment degeneration were substantially suppressed by 4-PBA administration, following attenuated photoreceptor apoptosis. Induction of retinal ER stress soon after light exposure, represented by upregulation of the immunoglobulin heavy chain binding protein (BiP) and C/EBP-Homologous Protein (CHOP), were suppressed by 4-PBA. Concurrently, light-induced oxidative stress markers, Nuclear factor erythroid 2-related factor 2 (Nrf2) and Heme Oxygenase 1 (HO-1), and mitochondrial apoptotic markers, B-cell lymphoma 2 apoptosis regulator (Bcl-2)-associated death promoter (Bad), and Bcl-2-associated X protein (Bax), were suppressed by 4-PBA administration. Increased expression of glial fibrillary acidic protein denoted retinal neuroinflammation, and inflammatory cytokines were induced after light exposure; however, 4-PBA acted as an anti-inflammatory. Suppression of ER stress by 4-PBA may be a new therapeutic approach to suppress the progression of retinal neurodegeneration and protect visual function against photo-stress.
Collapse
Affiliation(s)
- Naymel Alejandra Guzmán Mendoza
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.A.G.M.); (K.H.); (H.O.); (E.T.); (N.B.); (N.N.)
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
| | - Kohei Homma
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.A.G.M.); (K.H.); (H.O.); (E.T.); (N.B.); (N.N.)
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
| | - Hideto Osada
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.A.G.M.); (K.H.); (H.O.); (E.T.); (N.B.); (N.N.)
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
| | - Eriko Toda
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.A.G.M.); (K.H.); (H.O.); (E.T.); (N.B.); (N.N.)
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
| | - Norimitsu Ban
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.A.G.M.); (K.H.); (H.O.); (E.T.); (N.B.); (N.N.)
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
| | - Norihiro Nagai
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.A.G.M.); (K.H.); (H.O.); (E.T.); (N.B.); (N.N.)
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
- Department of Ophthalmology, St. Luke’s International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo 104-8560, Japan
- Laboratory of Retinal Cell Biology, St. Luke’s International University, 9-1 Akashi-cho, Chuo-ku, Tokyo 104-8560, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
| | - Kazuo Tsubota
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
| | - Yoko Ozawa
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.A.G.M.); (K.H.); (H.O.); (E.T.); (N.B.); (N.N.)
- Department of Ophthalmology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (K.T.)
- Department of Ophthalmology, St. Luke’s International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo 104-8560, Japan
- Laboratory of Retinal Cell Biology, St. Luke’s International University, 9-1 Akashi-cho, Chuo-ku, Tokyo 104-8560, Japan
| |
Collapse
|
32
|
Fibrosis, the Bad Actor in Cardiorenal Syndromes: Mechanisms Involved. Cells 2021; 10:cells10071824. [PMID: 34359993 PMCID: PMC8307805 DOI: 10.3390/cells10071824] [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] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiorenal syndrome is a term that defines the complex bidirectional nature of the interaction between cardiac and renal disease. It is well established that patients with kidney disease have higher incidence of cardiovascular comorbidities and that renal dysfunction is a significant threat to the prognosis of patients with cardiac disease. Fibrosis is a common characteristic of organ injury progression that has been proposed not only as a marker but also as an important driver of the pathophysiology of cardiorenal syndromes. Due to the relevance of fibrosis, its study might give insight into the mechanisms and targets that could potentially be modulated to prevent fibrosis development. The aim of this review was to summarize some of the pathophysiological pathways involved in the fibrotic damage seen in cardiorenal syndromes, such as inflammation, oxidative stress and endoplasmic reticulum stress, which are known to be triggers and mediators of fibrosis.
Collapse
|
33
|
Baba B, Caliskan M, Boyuk G, Hacisevki A. Chemical Chaperone PBA Attenuates ER Stress and Upregulates SOCS3 Expression as a Regulator of Leptin Signaling. BIOCHEMISTRY (MOSCOW) 2021; 86:480-488. [PMID: 33941068 DOI: 10.1134/s0006297921040088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Endoplasmic reticulum (ER) is very sensitive to the nutritional and energy states of the cells. Disruption of ER homeostasis leads to the accumulation of unfolded/misfolded proteins in the ER lumen, which is defined as ER stress. ER stress triggers the unfolded protein response (UPR). It is suggested that chronic ER stress is associated with obesity and leptin resistance. We investigated the role of ER stress and the effect of the ER stress inhibitor phenylbutyric acid (PBA) of ER stress, in obesity, as well as their impact on leptin signaling. This study involved twenty-four lean and twenty-four leptin-deficient (ob/ob) mice divided into PBA- and vehicle-treated groups. Pancreatic islets were isolated, incubated with leptin for 48 h, and assayed for the expression of CHOP and XBP1s (UPR signaling indicators) and SOCS3 (regulator of leptin signaling) by RT-qPCR. The expression levels of XBP1s and CHOP were markedly increased in the ob/ob controls compared to other groups with and without leptin treatment. No significant differences in the XBP1s and CHOP expression levels were found between the PBA-treated ob/ob and lean mice. SOCS3 expression was significantly upregulated in the PBA-treated ob/ob mice compared to the ob/ob controls after leptin treatment; but no significant difference in the SOCS3 expression was found between the PBA-treated ob/ob and lean mice with and without leptin treatment. Our findings suggested that ER stress plays an important role in the pathology of obesity, while PBA reduces ER stress and may potentially ameliorate leptin signaling.
Collapse
Affiliation(s)
- Burcu Baba
- Department of Medical Biochemistry, Faculty of Medicine, Yuksek Ihtisas University, Ankara, 06520, Turkey.
| | - Mursel Caliskan
- Department of Genetic, Gaziosmanpaşa Hospital, Yeni Yuzyil University, Istanbul, 34245, Turkey.
| | - Gulbahar Boyuk
- Department of Physiology, Faculty of Medicine, Ankara Medipol University, Ankara, 06050, Turkey.
| | - Aysun Hacisevki
- Department of Biochemistry, Faculty of Pharmacy, Gazi University, Ankara, 06100, Turkey.
| |
Collapse
|
34
|
Mechanisms linking endoplasmic reticulum (ER) stress and microRNAs to adipose tissue dysfunction in obesity. Crit Rev Biochem Mol Biol 2021; 56:455-481. [PMID: 34182855 DOI: 10.1080/10409238.2021.1925219] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over accumulation of lipids in adipose tissue disrupts metabolic homeostasis by affecting cellular processes. Endoplasmic reticulum (ER) stress is one such process affected by obesity. Biochemical and physiological alterations in adipose tissue due to obesity interfere with adipose ER functions causing ER stress. This is in line with increased irregularities in other cellular processes such as inflammation and autophagy, affecting overall metabolic integrity within adipocytes. Additionally, microRNAs (miRNAs), which can post-transcriptionally regulate genes, are differentially modulated in obesity. A better understanding and identification of such miRNAs could be used as novel therapeutic targets to fight against diseases. In this review, we discuss ways in which ER stress participates as a common molecular process in the pathogenesis of obesity-associated metabolic disorders. Moreover, our review discusses detailed underlying mechanisms through which ER stress and miRNAs contribute to metabolic alteration in adipose tissue in obesity. Hence, identifying mechanistic involvement of miRNAs-ER stress cross-talk in regulating adipose function during obesity could be used as a potential therapeutic approach to combat chronic diseases, including obesity.
Collapse
|
35
|
4-Phenylbutyrate (PBA) treatment reduces hyperglycemia and islet amyloid in a mouse model of type 2 diabetes and obesity. Sci Rep 2021; 11:11878. [PMID: 34088954 PMCID: PMC8178353 DOI: 10.1038/s41598-021-91311-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Amyloid deposits in pancreatic islets, mainly formed by human islet amyloid polypeptide (hIAPP) aggregation, have been associated with loss of β-cell mass and function, and are a pathological hallmark of type 2 diabetes (T2D). Treatment with chaperones has been associated with a decrease in endoplasmic reticulum stress leading to improved glucose metabolism. The aim of this work was to investigate whether the chemical chaperone 4-phenylbutyrate (PBA) prevents glucose metabolism abnormalities and amyloid deposition in obese agouti viable yellow (Avy) mice that overexpress hIAPP in β cells (Avy hIAPP mice), which exhibit overt diabetes. Oral PBA treatment started at 8 weeks of age, when Avy hIAPP mice already presented fasting hyperglycemia, glucose intolerance, and impaired insulin secretion. PBA treatment strongly reduced the severe hyperglycemia observed in obese Avy hIAPP mice in fasting and fed conditions throughout the study. This effect was paralleled by a decrease in hyperinsulinemia. Importantly, PBA treatment reduced the prevalence and the severity of islet amyloid deposition in Avy hIAPP mice. Collectively, these results show that PBA treatment elicits a marked reduction of hyperglycemia and reduces amyloid deposits in obese and diabetic mice, highlighting the potential of chaperones for T2D treatment.
Collapse
|
36
|
Gadallah SH, Ghanem HM, Abdel-Ghaffar A, Metwaly FG, Hanafy LK, Ahmed EK. 4-Phenylbutyric acid and rapamycin improved diabetic status in high fat diet/streptozotocin-induced type 2 diabetes through activation of autophagy. Arch Physiol Biochem 2021; 127:235-244. [PMID: 31215250 DOI: 10.1080/13813455.2019.1628069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 05/31/2019] [Indexed: 10/26/2022]
Abstract
An accumulating body of evidence supports the role of autophagy in the pathophysiology of T2DM. Also, abnormal endoplasmic reticulum (ER) stress response that has been implicated as a cause of insulin resistance (IR) could also be affected by the autophagic status in β-cells. The present study was designed to investigate whether autophagy is regulated in T2DM as well as to investigate the modulatory effect of the ER stress inhibitor 4-phenylbutyric acid (4-PBA) and the autophagy inducer rapamycin (Rapa) on the autophagic and diabetic status using type 2 diabetic animal model with IR. Treatment of diabetic rats with either 4-PBA or Rapa improved significantly the states of hyperglycaemia and dyslipidaemia, increased the antioxidant capacity, reduced the levels of lipid peroxidation and ER stress and increased the autophagic flux. The obtained improvements were attributed mainly to the induction of autophagy with subsequent regulation of ER stress-oxidative activation and prevention of β-cell apoptosis.
Collapse
Affiliation(s)
- Shaimaa H Gadallah
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Hala M Ghanem
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Amany Abdel-Ghaffar
- Department of Biochemistry and Pharmacology, Research Institute of Ophthalmology, Giza, Egypt
| | - Fatma G Metwaly
- Department of Histology, Research Institute of Ophthalmology, Giza, Egypt
| | - Laila K Hanafy
- Department of Histology, Research Institute of Ophthalmology, Giza, Egypt
| | - Emad K Ahmed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
37
|
Calcium as a reliable marker for the quantitative assessment of endoplasmic reticulum stress in live cells. J Biol Chem 2021; 296:100779. [PMID: 34000299 PMCID: PMC8191341 DOI: 10.1016/j.jbc.2021.100779] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/01/2021] [Accepted: 05/11/2021] [Indexed: 11/20/2022] Open
Abstract
Calcium (Ca2+) is an essential mineral of endoplasmic reticulum (ER) luminal biochemistry because of the Ca2+ dependence of ER-resident chaperones charged with folding de novo proteins that transit this cellular compartment. ER Ca2+ depletion reduces the ability of chaperones to properly fold the proteins entering the ER, thus leading to an accumulation of misfolded proteins and the onset of a state known as ER stress. However, not all conditions that cause ER stress do so in a manner dependent on ER Ca2+ depletion. Agents such as tunicamycin inhibit the glycosylation of de novo polypeptides, a key step in the maturation process of newly synthesized proteins. Despite this established effect of tunicamycin, our understanding of how such conditions modulate ER Ca2+ levels is still limited. In the present study, we report that a variety of ER stress–inducing agents that have not been known to directly alter ER Ca2+ homeostasis can also cause a marked reduction in ER Ca2+ levels. Consistent with these observations, protecting against ER stress using small chemical chaperones, such as 4-phenylbutyrate and tauroursodeoxycholic acid, also attenuated ER Ca2+ depletion caused by these agents. We also describe a novel high-throughput and low-cost assay for the rapid quantification of ER stress using ER Ca2+ levels as a surrogate marker. This report builds on our understanding of ER Ca2+ levels in the context of ER stress and also provides the scientific community with a new, reliable tool to study this important cellular process in vitro.
Collapse
|
38
|
Tokugawa M, Inoue Y, Ishiuchi K, Kujirai C, Matsuno M, Ri M, Itoh Y, Miyajima C, Morishita D, Ohoka N, Iida S, Mizukami H, Makino T, Hayashi H. Periplocin and cardiac glycosides suppress the unfolded protein response. Sci Rep 2021; 11:9528. [PMID: 33947921 PMCID: PMC8097017 DOI: 10.1038/s41598-021-89074-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
The unfolded protein response (UPR) controls protein homeostasis through transcriptional and translational regulation. However, dysregulated UPR signaling has been associated with the pathogenesis of many human diseases. Therefore, the compounds modulating UPR may provide molecular insights for these pathologies in the context of UPR. Here, we screened small-molecule compounds that suppress UPR, using a library of Myanmar wild plant extracts. The screening system to track X-box binding protein 1 (XBP1) splicing activity revealed that the ethanol extract of the Periploca calophylla stem inhibited the inositol-requiring enzyme 1 (IRE1)-XBP1 pathway. We isolated and identified periplocin as a potent inhibitor of the IRE1-XBP1 axis. Periplocin also suppressed other UPR axes, protein kinase R-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6). Examining the structure–activity relationship of periplocin revealed that cardiac glycosides also inhibited UPR. Moreover, periplocin suppressed the constitutive activation of XBP1 and exerted cytotoxic effects in the human multiple myeloma cell lines, AMO1 and RPMI8226. These results reveal a novel suppressive effect of periplocin or the other cardiac glycosides on UPR regulation, suggesting that these compounds will contribute to our understanding of the pathological or physiological importance of UPR.
Collapse
Affiliation(s)
- Muneshige Tokugawa
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Yasumichi Inoue
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan. .,Department of Innovative Therapeutic Sciences, Cooperative Major in Nanopharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| | - Kan'ichiro Ishiuchi
- Department of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Chisane Kujirai
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Michiyo Matsuno
- The Kochi Prefectural Makino Botanical Garden, Kochi, 781-8125, Japan
| | - Masaki Ri
- Department of Hematology and Oncology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, 467-8601, Japan
| | - Yuka Itoh
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Chiharu Miyajima
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.,Department of Innovative Therapeutic Sciences, Cooperative Major in Nanopharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Daisuke Morishita
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.,Chordia Therapeutics Inc., Kanagawa, 251-0012, Japan
| | - Nobumichi Ohoka
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, Kawasaki, 210-9501, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, 467-8601, Japan
| | - Hajime Mizukami
- The Kochi Prefectural Makino Botanical Garden, Kochi, 781-8125, Japan
| | - Toshiaki Makino
- Department of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan. .,Department of Innovative Therapeutic Sciences, Cooperative Major in Nanopharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| |
Collapse
|
39
|
Garibaldi N, Contento BM, Babini G, Morini J, Siciliani S, Biggiogera M, Raspanti M, Marini JC, Rossi A, Forlino A, Besio R. Targeting cellular stress in vitro improves osteoblast homeostasis, matrix collagen content and mineralization in two murine models of osteogenesis imperfecta. Matrix Biol 2021; 98:1-20. [PMID: 33798677 PMCID: PMC11162743 DOI: 10.1016/j.matbio.2021.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022]
Abstract
Most cases of dominantly inherited osteogenesis imperfecta (OI) are caused by glycine substitutions in the triple helical domain of type I collagen α chains, which delay collagen folding, and cause the synthesis of collagen triple helical molecules with abnormal structure and post-translational modification. A variable extent of mutant collagen ER retention and other secondary mutation effects perturb osteoblast homeostasis and impair bone matrix quality. Amelioration of OI osteoblast homeostasis could be beneficial both to osteoblast anabolic activity and to the content of the extracellular matrix they deposit. Therefore, the effect of the chemical chaperone 4-phenylbutyrate (4-PBA) on cell homeostasis, collagen trafficking, matrix production and mineralization was investigated in primary osteoblasts from two murine models of moderate OI, Col1a1+/G349C and Col1a2+/G610C. At the cellular level, 4-PBA prevented intracellular accumulation of collagen and increased protein secretion, reducing aggregates within the mutant cells and normalizing ER morphology. At the extracellular level, increased collagen incorporation into matrix, associated with more mature collagen fibrils, was observed in osteoblasts from both models. 4-PBA also promoted OI osteoblast mineral deposition by increasing alkaline phosphatase expression and activity. Targeting osteoblast stress with 4-PBA improved both cellular and matrix abnormalities in culture, supporting further in vivo studies of its effect on bone tissue composition, strength and mineralization as a potential treatment for classical OI.
Collapse
Affiliation(s)
- Nadia Garibaldi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy; Istituto Universitario di Studi Superiori - IUSS, Pavia, Italy.
| | - Barbara M Contento
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | | | - Jacopo Morini
- Department of Physics, University of Pavia, Pavia, Italy.
| | - Stella Siciliani
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy.
| | - Marco Biggiogera
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy.
| | - Mario Raspanti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Joan C Marini
- Bone and Extracellular Matrix Branch, NICHD, National Institute of Health, Bethesda, MD 20892, USA.
| | - Antonio Rossi
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| |
Collapse
|
40
|
Xu X, Li Q, Li L, Zeng M, Zhou X, Cheng Z. Endoplasmic reticulum stress/XBP1 promotes airway mucin secretion under the influence of neutrophil elastase. Int J Mol Med 2021; 47:81. [PMID: 33760106 PMCID: PMC7979262 DOI: 10.3892/ijmm.2021.4914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 02/17/2021] [Indexed: 12/27/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is an important reaction of airway epithelial cells in response to various stimuli, and may also be involved in the mucin secretion process. In the present study, the effect of ER stress on neutrophil elastase (NE)-induced mucin (MUC)5AC production in human airway epithelial cells was explored. 16HBE14o-airway epithelial cells were cultured and pre-treated with the reactive oxygen species (ROS) inhibitor, N-acetylcysteine (NAC), or the ER stress chemical inhibitor, 4-phenylbutyric acid (4-PBA), or the cells were transfected with inositol-requiring kinase 1α (IRE1α) small interfering RNA (siRNA) or X-box-binding protein 1 (XBP1) siRNA, respectively, and subsequently incubated with NE. The results obtained revealed that NE increased ROS production in the 16HBE14o-cells, with marked increases in the levels of ER stress-associated proteins, such as glucose-regulated protein 78 (GRP78), activating transcription factor 6 (ATF6), phosphorylated protein kinase R-like endoplasmic reticulum kinase (pPERK) and phosphorylated (p)IRE1α. The protein and mRNA levels of spliced XBP1 were also increased, and the level of MUC5AC protein was notably increased. The ROS scavenger NAC and ER stress inhibitor 4-PBA were found to reduce ER stress-associated protein expression and MUC5AC production and secretion. Further analyses revealed that MUC5AC secretion was also attenuated by IRE1α and XBP1 siRNAs, accompanied by a decreased mRNA expression of spliced XBP1. Taken together, these results demonstrate that NE induces ER stress by promoting ROS production in 16HBE14o-airway epithelial cells, leading to increases in MUC5AC protein production and secretion via the IRE1α and XBP1 signaling pathways.
Collapse
Affiliation(s)
- Xiaoyan Xu
- Department of Respiratory Medicine, The Affiliated Hospital of Qingdao University, Shinan, Qingdao 266071, P.R. China
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Liang Li
- Department of Respiratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Man Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Zhaozhong Cheng
- Department of Respiratory Medicine, The Affiliated Hospital of Qingdao University, Shinan, Qingdao 266071, P.R. China
| |
Collapse
|
41
|
Chronic metformin treatment decreases cardiac injury during ischemia-reperfusion by attenuating endoplasmic reticulum stress with improved mitochondrial function. Aging (Albany NY) 2021; 13:7828-7845. [PMID: 33746115 PMCID: PMC8034968 DOI: 10.18632/aging.202858] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/11/2021] [Indexed: 11/25/2022]
Abstract
Aging impairs mitochondrial function that leads to greater cardiac injury during ischemia and reperfusion. Cardiac endoplasm reticulum (ER) stress increases with age and contributes to mitochondrial dysfunction. Metformin is an anti-diabetic drug that protects cardiac mitochondria during acute ER stress. We hypothesized that metformin treatment would improve preexisting mitochondrial dysfunction in aged hearts by attenuating ER stress, followed by a decrease in cardiac injury during subsequent ischemia and reperfusion. Male young (3 mo.) and aged mice (24 mo.) received metformin (300 mg/kg/day) dissolved in drinking water with sucrose (0.2 g/100 ml) as sweetener for two weeks versus sucrose vehicle alone. Cytosol, subsarcolemmal (SSM), and interfibrillar mitochondria (IFM) were isolated. In separate groups, cardioprotection was evaluated using ex vivo isolated heart perfusion with 25 min. global ischemia and 60 min. reperfusion. Infarct size was measured. The contents of CHOP and cleaved ATF6 were decreased in metformin-treated 24 mo. mice compared to vehicle, supporting a decrease in ER stress. Metformin treatment improved OXPHOS in IFM in 24 mo. using a complex I substrate. Metformin treatment decreased infarct size following ischemia-reperfusion. Thus, metformin feeding decreased cardiac injury in aged mice during ischemia-reperfusion by improving pre-ischemic mitochondrial function via inhibition of ER stress.
Collapse
|
42
|
Lee JM, Park S, Lee D, Ginting RP, Lee MR, Lee MW, Han J. Reduction in endoplasmic reticulum stress activates beige adipocytes differentiation and alleviates high fat diet-induced metabolic phenotypes. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166099. [PMID: 33556486 DOI: 10.1016/j.bbadis.2021.166099] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
Endoplasmic reticulum (ER) stress is closely associated with various metabolic diseases, such as obesity and diabetes. Development of beige/brite adipocytes increases thermogenesis and helps to reduce obesity. Although the relationship between ER stress and white adipocytes has been studied considerably, the possible role of ER stress and the unfolded protein response (UPR) induction in beige adipocytes differentiation remain to be investigated. In this study we investigated how ER stress affected beige adipocytes differentiation both in vitro and in vivo. Phosphorylation of eIF2α was transiently decreased in the early phase (day 2), whereas it was induced at the late phase with concomitant induction of C/EBP homologous protein (CHOP) during beige adipocytes differentiation. Forced expression of CHOP inhibited the expression of beige adipocytes markers, including Ucp1, Cox8b, Cidea, Prdm16, and Pgc-1α, following the induction of beige adipocytes differentiation. When ER stress was reduced by the chemical chaperone tauroursodeoxycholic acid (TUDCA), the expression of the beige adipocytes marker uncoupling protein 1 (UCP1) was significantly enhanced in inguinal white adipose tissue (iWAT) and high fat diet (HFD)-induced abnormal metabolic phenotype was improved. In summary, we found that ER stress and the UPR induction were closely involved in beige adipogenesis. These results suggest that modulating ER stress could be a potential therapeutic intervention against metabolic dysfunctions via activation of iWAT browning.
Collapse
Affiliation(s)
- Ji-Min Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Soyoung Park
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Republic of Korea
| | - Duckgue Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea
| | - Rehna Paula Ginting
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Republic of Korea
| | - Man Ryul Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea; Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Republic of Korea.
| | - Min-Woo Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea; Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Republic of Korea.
| | - Jaeseok Han
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, 31151, Republic of Korea; Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si 31151, Republic of Korea.
| |
Collapse
|
43
|
Zubarioglu T, Dede E, Cigdem H, Kiykim E, Cansever MS, Aktuglu-Zeybek C. Impact of sodium phenylbutyrate treatment in acute management of maple syrup urine disease attacks: a single-center experience. J Pediatr Endocrinol Metab 2021; 34:121-126. [PMID: 33180043 DOI: 10.1515/jpem-2020-0356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Accurate management of metabolic decompensation in maple syrup urine disease (MSUD) has a crucial role, as acute attacks can cause neurological sequels and can be life threatening. Here, we aimed to evaluate effect of sodium phenylbutyrate (NaPBA) in acute management of MSUD attacks. METHODS Episodes with an initial plasma leucine (Leu) level above 750 µmoL/L and that require hospitalization due to clinical findings of Leu neurotoxicity and/or feeding difficulties were included to the study. Patients who had no molecular diagnosis and a regular follow-up were excluded. Clinical findings, laboratory results and therapy responses were reviewed, retrospectively. RESULTS Ten patients who experienced 19 distinct episodes of MSUD attacks were enrolled. Initial median Leu level was 901.67 (range 756-1989.11) and 33.9 µmoL/L (range 7.91-347.3 µmoL/L) at the end of therapy. None of our patients underwent extracorporeal toxin removal during the course of attack. In patients with serial plasma quantitative amino acid sampling, mean Leu reduction rate was calculated to be 529.68 ± 250.08 µmoL/L/day at the 24th h of treatment and 318.72 ± 191.52 µmoL/L/day at the 48th h of treatment. CONCLUSIONS This study is the first original study that investigates the effect of NaPBA in management of acute attacks of MSUD patients from Turkey. We suggest that NaPBA treatment in MSUD attacks can ameliorate clinical and biochemical findings. This therapeutic option should be considered especially in smaller centers without the toxin removal chance and for patients who were not appropriate for extracorporeal toxin removal like hemodynamic instability.
Collapse
Affiliation(s)
- Tanyel Zubarioglu
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatrics Division of Nutrition and Metabolism, Istanbul, Turkey
| | - Elif Dede
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatrics, Istanbul, Turkey
| | - Humeyra Cigdem
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatrics Division of Nutrition and Metabolism, Istanbul, Turkey
| | - Ertugrul Kiykim
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatrics Division of Nutrition and Metabolism, Istanbul, Turkey
| | - Mehmet S Cansever
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Central Laboratory, Istanbul, Turkey
| | - Cigdem Aktuglu-Zeybek
- Istanbul University-Cerrahpasa, Cerrahpasa Medical Faculty, Department of Pediatrics Division of Nutrition and Metabolism, Istanbul, Turkey
| |
Collapse
|
44
|
Gong L, Liu G, Zhu H, Li C, Li P, Liu C, Tang H, Wu K, Wu J, Liu D, Tang X. IL-32 induces epithelial-mesenchymal transition by triggering endoplasmic reticulum stress in A549 cells. BMC Pulm Med 2020; 20:278. [PMID: 33097029 PMCID: PMC7585222 DOI: 10.1186/s12890-020-01319-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/18/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is a key process in the onset and development of idiopathic pulmonary fibrosis (IPF) with unclear mechanisms. Our previous studies found that bleomycin and tunicamycin could induce ER stress and consequently trigger EMT accompanying with IL-32 overexpression. This study was aimed to investigate the effects of IL-32 on EMT and ER stress to elucidate the pathogenesis of IPF. METHODS Human lung adenocarcinoma A549 cells were treated with recombinant human (rh)IL-32, IL-32 siRNA and EMT inducer tunicamycin, or 4-phenylbutyric acid (4-PBA), respectively. Then the cell morphology was observed and the expression of ER-related markers and EMT-related markers were detected by RT-qPCR or western blotting. RESULTS Stimulation of A549 cells with rhIL-32 led to a morphological change from a pebble-like shape to an elongated shape in a portion of the cells, accompanied by down regulated expression of the epithelial cell marker E-cadherin and up regulated expression of the mesenchymal cell markers N-cadherin, Vimentin, and Zeb-1. However, these rhIL-32 induced changes were inhibited by the ER stress inhibitor 4-PBA. Suppression of IL-32 expression with siRNA inhibited TM-induced EMT. Further stimulation of the A549 cells with rhIL-32 demonstrated an increase in the expression of GRP78, although this increase was also inhibited by 4-PBA. CONCLUSIONS These results suggest that IL-32 induces EMT in A549 cells by triggering ER stress, and IL-32 may be a novel marker for IPF.
Collapse
Affiliation(s)
- Ling Gong
- The First Clinical Medical College, Jinan University, 601 W. Huangpu Avenue, Guangzhou, 510630, China
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Gang Liu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Honglan Zhu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Caihong Li
- Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Pengmei Li
- Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Changlu Liu
- Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hongbo Tang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Kaifeng Wu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
- Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Jie Wu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
- Scientific Research Center, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China
| | - Daishun Liu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, 563000, Guizhou, China.
- Department of Respiratory, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), No.98 Fenghuang Road, Zunyi, 563002, Guizhou, China.
| | - Xiaoping Tang
- The First Clinical Medical College, Jinan University, 601 W. Huangpu Avenue, Guangzhou, 510630, China.
| |
Collapse
|
45
|
Thompson J, Maceyka M, Chen Q. Targeting ER stress and calpain activation to reverse age-dependent mitochondrial damage in the heart. Mech Ageing Dev 2020; 192:111380. [PMID: 33045249 DOI: 10.1016/j.mad.2020.111380] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/17/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Severity of cardiovascular disease increases markedly in elderly patients. In addition, many therapeutic strategies that decrease cardiac injury in adult patients are invalid in elderly patients. Thus, it is a challenge to protect the aged heart in the context of underlying chronic or acute cardiac diseases including ischemia-reperfusion injury. The cause(s) of this age-related increased damage remain unknown. Aging impairs the function of the mitochondrial electron transport chain (ETC), leading to decreased energy production and increased oxidative stress due to generation of reactive oxygen species (ROS). Additionally, ROS-induced oxidative stress can increase cardiac injury during ischemia-reperfusion by potentiating mitochondrial permeability transition pore (MPTP) opening. Aging leads to increased endoplasmic reticulum (ER) stress, which contributes to mitochondrial dysfunction, including reduced function of the ETC. The activation of both cytosolic and mitochondrial calcium-activated proteases termed calpains leads to mitochondrial dysfunction and decreased ETC function. Intriguingly, mitochondrial ROS generation also induces ER stress, highlighting the dynamic interaction between mitochondria and ER. Here, we discuss the role of ER stress in sensitizing and potentiating mitochondrial dysfunction in response to ischemia-reperfusion, and the promising potential therapeutic benefit of inhibition of ER stress and / or calpains to attenuate cardiac injury in elderly patients.
Collapse
Affiliation(s)
- Jeremy Thompson
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, United States
| | - Michael Maceyka
- Department of Biochemistry & Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, United States
| | - Qun Chen
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298, United States.
| |
Collapse
|
46
|
Chen Q, Samidurai A, Thompson J, Hu Y, Das A, Willard B, Lesnefsky EJ. Endoplasmic reticulum stress-mediated mitochondrial dysfunction in aged hearts. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165899. [PMID: 32698045 DOI: 10.1016/j.bbadis.2020.165899] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/04/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022]
Abstract
Aging impairs the mitochondrial electron transport chain (ETC), especially in interfibrillar mitochondria (IFM). Mitochondria are in close contact with the endoplasmic reticulum (ER). Induction of ER stress leads to ETC injury in adult heart mitochondria. We asked if ER stress contributes to the mitochondrial dysfunction during aging. Subsarcolemmal mitochondria (SSM) and IFM were isolated from 3, 18, and 24 mo. C57Bl/6 mouse hearts. ER stress progressively increased with age, especially in 24 mo. mice that manifest mitochondrial dysfunction. OXPHOS was decreased in 24 mo. IFM oxidizing complex I and complex IV substrates. Proteomic analysis showed that the content of multiple complex I subunits was decreased in IFM from 24 mo. hearts, but remained unchanged in in 18 mo. IFM without a decrease in OXPHOS. Feeding 24 mo. old mice with 4-phenylbutyrate (4-PBA) for two weeks attenuated the ER stress and improved mitochondrial function. These results indicate that ER stress contributes to the mitochondrial dysfunction in aged hearts. Attenuation of ER stress is a potential approach to improve mitochondrial function in aged hearts.
Collapse
Affiliation(s)
- Qun Chen
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Arun Samidurai
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Jeremy Thompson
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Ying Hu
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Anindita Das
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Belinda Willard
- Proteomics Core, Cleveland Clinic, Cleveland, OH 44106, United States of America
| | - Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, United States of America; Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298, United States of America; McGuire Department of Veterans Affairs Medical Center, Richmond, VA 23249, United States of America.
| |
Collapse
|
47
|
Truong NTT, Lydic TA, Bazil JN, Suryadevara A, Olson LK. Regulation of lipid metabolism in pancreatic beta cells by interferon gamma: A link to anti-viral function. Cytokine 2020; 133:155147. [PMID: 32492632 DOI: 10.1016/j.cyto.2020.155147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/01/2020] [Accepted: 05/25/2020] [Indexed: 12/22/2022]
Abstract
Interferons (IFN) have been shown to alter lipid metabolism in immune and some non-hematopoietic cells and this affects host cell response to pathogens. In type 1 diabetes, IFNγ acts as a proinflammatory cytokine that, along with other cytokines, is released during pancreatic beta cell autoinflammation and contributes to immune response and beta cell dysfunction. The hypothesis tested herein is that IFN modifies beta cell lipid metabolism and this is associated with enhanced anti-viral response and beta cell stress. Treatment of INS-1 cells with IFNγ for 6 to 24 h led to a dynamic change in TAG and lipid droplet (LD) levels, with a decrease at 6 h and an increase at 24 h. The later accumulation of TAG was associated with increased de novo lipogenesis (DNL), and impaired mitochondrial fatty acid oxidation (FAO). Gene expression results suggested that IFNγ regulates lipolytic, lipogenic, LD and FAO genes in a temporal manner. The changes in lipid gene expression are dependent on the classical Janus kinase (JAK) pathway. Pretreatment with IFNγ robustly enhanced anti-viral gene expression induced by the viral mimetic polyinosinic: polycytidylic acid (PIC), and this potentiating effect of IFNγ was markedly attenuated by inhibitors of DNL. The IFNγ-induced accumulation of lipid, however, was insufficient to cause endoplasmic reticulum (ER) stress. These studies demonstrated a non-canonical effect of IFNγ in regulation of pancreatic beta cell lipid metabolism that is intimately linked with host cell defense and might alter cellular function early in the progression to type 1 diabetes.
Collapse
Affiliation(s)
- Nguyen T T Truong
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Todd A Lydic
- Department of Physiology, Michigan State University, East Lansing, MI, United States; Mass Spectrometry Core (MMD-CMSC), Michigan State University, East Lansing, MI, United States
| | - Jason N Bazil
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Abhijeet Suryadevara
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - L Karl Olson
- Department of Physiology, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
48
|
Menikdiwela KR, Ramalingam L, Abbas MM, Bensmail H, Scoggin S, Kalupahana NS, Palat A, Gunaratne P, Moustaid-Moussa N. Role of microRNA 690 in Mediating Angiotensin II Effects on Inflammation and Endoplasmic Reticulum Stress. Cells 2020; 9:cells9061327. [PMID: 32466437 PMCID: PMC7348980 DOI: 10.3390/cells9061327] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 12/27/2022] Open
Abstract
Overactivation of the renin–angiotensin system (RAS) during obesity disrupts adipocyte metabolic homeostasis and induces endoplasmic reticulum (ER) stress and inflammation; however, underlying mechanisms are not well known. We propose that overexpression of angiotensinogen (Agt), the precursor protein of RAS in adipose tissue or treatment of adipocytes with Angiotensin II (Ang II), RAS bioactive hormone, alters specific microRNAs (miRNA), that target ER stress and inflammation leading to adipocyte dysfunction. Epididymal white adipose tissue (WAT) from B6 wild type (Wt) and transgenic male mice overexpressing Agt (Agt-Tg) in adipose tissue and adipocytes treated with Ang II were used. Small RNA sequencing and microarray in WAT identified differentially expressed miRNAs and genes, out of which miR-690 and mitogen-activated protein kinase kinase 3 (MAP2K3) were validated as significantly up- and down-regulated, respectively, in Agt-Tg, and in Ang II-treated adipocytes compared to respective controls. Additionally, the direct regulatory role of miR-690 on MAP2K3 was confirmed using mimic, inhibitors and dual-luciferase reporter assay. Downstream protein targets of MAP2K3 which include p38, NF-κB, IL-6 and CHOP were all reduced. These results indicate a critical post-transcriptional role for miR-690 in inflammation and ER stress. In conclusion, miR-690 plays a protective function and could be a useful target to reduce obesity.
Collapse
Affiliation(s)
- Kalhara R. Menikdiwela
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
| | - Latha Ramalingam
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
| | - Mostafa M. Abbas
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha 34110, Qatar; (M.M.A.); (H.B.)
- Department of Imaging Science and Innovation, Geisinger Health System, Danville, PA 17822, USA
| | - Halima Bensmail
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha 34110, Qatar; (M.M.A.); (H.B.)
| | - Shane Scoggin
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
| | - Nishan S. Kalupahana
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
- Department of Physiology, University of Peradeniya, Peradeniya 20400, Sri Lanka
| | - Asha Palat
- Biology and Biochemistry, University of Houston, Houston, TX 77204, USA; (A.P.); (P.G.)
| | - Preethi Gunaratne
- Biology and Biochemistry, University of Houston, Houston, TX 77204, USA; (A.P.); (P.G.)
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA; (K.R.M.); (L.R.); (S.S.); (N.S.K.)
- Correspondence: ; Tel.: +806-834-7946
| |
Collapse
|
49
|
Chlorantraniliprole induces adipogenesis in 3T3-L1 adipocytes via the AMPKα pathway but not the ER stress pathway. Food Chem 2020; 311:125953. [DOI: 10.1016/j.foodchem.2019.125953] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/19/2019] [Accepted: 11/24/2019] [Indexed: 01/22/2023]
|
50
|
Tan L, Harper L, McNulty MA, Carlson CS, Yammani RR. High-fat diet induces endoplasmic reticulum stress to promote chondrocyte apoptosis in mouse knee joints. FASEB J 2020; 34:5818-5826. [PMID: 32124494 DOI: 10.1096/fj.201902746r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/31/2022]
Abstract
Mice fed a high-fat diet (HFD) become obese and develop osteoarthritis (OA)-like lesions, including chondrocyte apoptosis, in the knee joints. However, the mechanism by which HFD/obesity induces chondrocyte apoptosis is not clearly understood. In the present study, male mice were fed a low-fat diet (LFD, 10% kcal), HFD (45% kcal), or a HFD administered with 0.5 g/kg bodyweight of 4-phenyl butyric acid (PBA, a small chaperone known to ease endoplasmic reticulum [ER] stress), via the drinking water. At the end of the 18-week study, stifle (knee) joints from all animals were collected, fixed, paraffin embedded, and sectioned. Immunostaining of joints from the HFD group showed increased expression of ER stress and apoptotic markers and increased expression of nuclear protein 1 and tribbles related protein-3 compared to the LFD group. Mice on HFD also showed higher percentage of chondrocyte death, lower chondrocyte numbers per cartilage area, and thickening of subchondral bone. Administration of PBA alleviated all of the HFD-induced symptoms. Our study demonstrated that HFD induces ER stress to promote chondrocyte death and subchondral bone thickening, which could be relieved by alleviating ER stress via PBA administration, suggesting that ER stress could play an important role in obesity-linked OA and could be targeted for OA therapeutics.
Collapse
Affiliation(s)
- Li Tan
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lindsey Harper
- Veterinary Clinical Sciences Department, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Margaret A McNulty
- Department of Anatomy, Cell Biology, & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cathy S Carlson
- Veterinary Clinical Sciences Department, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Raghunatha R Yammani
- Section of Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|