1
|
Lin M, Mo Y, Li CM, Liu YZ, Feng XP. GRP78 as a potential therapeutic target in cancer treatment: an updated review of its role in chemoradiotherapy resistance of cancer cells. Med Oncol 2025; 42:49. [PMID: 39827214 DOI: 10.1007/s12032-024-02586-0] [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: 11/11/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
GRP78 (Glucose-related protein 78, BiP/HSPA5) is commonly overexpressed in cancer cells. Acting as an activator of endoplasmic reticulum stress, GRP78 is involved in the resistance of cancer cells to injury. Current evidence suggests that GRP78 plays a significant role in the radiotherapy resistance and chemotherapy resistance of cancers, which is accomplished through a variety of complex pathways. These include the promotion of tumor stemness, inhibition of apoptosis, regulation of autophagy, maintenance of tumor microenvironment homeostasis, protection of dormant cells, evasion of senescence, counteraction of autoantibodies against GRP78, facilitation of DNA damage repair, suppression of ferroptosis, and modulation of metabolic reprogramming in tumor cells. Importantly, chemoradiotherapy resistance in cancers are the main reasons for treatment failure in patients, severely affecting their survival. Investigating the mechanisms of GRP78 in tumor therapeutic resistance is essential. In this article, we review the mechanisms by which GRP78 mediates cell survival and chemoradiotherapy resistance in cancers and provide an overview of clinical trials targeting GRP78 therapy.
Collapse
Affiliation(s)
- Min Lin
- Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yan Mo
- Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Cheng-Min Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ying-Zhe Liu
- Xiangya International Medical Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Xue-Ping Feng
- Department of Oncology and Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
2
|
Liu Q, Wang L, An L, Liu Y, Qu H, Huang S, Zhao L, Yin Y, Ma Q. Vitamin E Mitigates Apoptosis in Ovarian Granulosa Cells by Inhibiting Zearalenone-Induced Activation of the PERK/eIF-2α/ATF4/Chop Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28390-28399. [PMID: 39610174 DOI: 10.1021/acs.jafc.4c07623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
A study aimed to investigate the signaling pathway of zearalenone (ZEA) leading to the apoptosis of ovarian granulosa cells (GCs) and explore the potential of vitamin E (VE) in alleviating ZEA-induced apoptosis of GCs. We constructed an apoptosis model for GCs based on exposure to the environmental toxin ZEA. Transcriptome analysis revealed that ZEA induced endoplasmic reticulum stress by activating the ATF4-Chop pathway. The addition of inhibitors targeting the estrogen receptor (ER) demonstrated that ZEA activates the ATF4-Chop pathway through ER-beta. As a strong antioxidant, VE is thought to mitigate ZEA-induced toxicity. Interestingly, molecular docking analysis at the PERK active site of the endoplasmic reticulum stress revealed a high binding capacity of VE. VE supplementation reduced apoptosis in GCs and decreased the expression of p-eIF-2α, ATF4, and Chop. Mouse tests also demonstrated that VE supplementation effectively mitigated ovarian dysfunction induced by ZEA, as evidenced by increased body weight gain, reduced oxidative stress, and decreased cell death. In summary, the present study demonstrates that ZEA activates the PERK-eIF-2α-ATF4-Chop pathway through ERβ, leading to endoplasmic reticulum stress and apoptosis of GCs. Conversely, VE inhibits the PERK/eIF-2α/ATF4/Chop signaling pathways, mitigating endoplasmic reticulum stress and improving ZEA-induced reproductive toxicity.
Collapse
Affiliation(s)
- Qingxiu Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Leli Wang
- Centre of Healthy Animal Husbandry, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Lei An
- State Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yafei Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Honglei Qu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lihong Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yulong Yin
- Centre of Healthy Animal Husbandry, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
3
|
Wang SF, Ho YC, Chou CY, Chang YL, Lee HC, Tseng LM. Integrated stress response-upregulated mitochondrial SLC1A5var enhances glucose dependency in human breast cancer cells in vitro. Int J Biochem Cell Biol 2024; 177:106688. [PMID: 39505107 DOI: 10.1016/j.biocel.2024.106688] [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: 06/19/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Breast cancer is the most commonly diagnosed cancer among women. The growth of triple-negative breast cancer (TNBC) cells is glucose-dependent. The integrated stress response (ISR) is a cellular stress response to glucose depletion. The ISR-solute carrier family 7 member 11 pathway is activated during glucose depletion and contributes to glucose dependence by decreasing intracellular glutamate levels. Solute carrier family 1 member 5 (SLC1A5) and the mitochondrial solute carrier family 1 member 5 variant (SLC1A5var) are glutamine transporters that play essential roles in the reprogramming of cancer metabolism. However, whether ISR can regulate mitochondrial SLC1A5var expression and further affect glucose dependence remains unclear. Glucose depletion-, oligomycin-, and salubrinal-activated activating transcription factor-4 (ATF4) induced SLC1A5var expression. ATF4 is critical for SLC1A5var regulation, as it binds to specific regulatory elements in its promoter. SLC1A5var knockdown decreases glucose depletion-induced cell death, whereas SLC1A5var overexpression increases glucose depletion-induced cell death in TNBC cells. SLC1A5var knockdown reduced cancer cell proliferation, colony formation, and migration, whereas SLC1A5var overexpression increased cell proliferation and migration. Moreover, the knockdown of SLC1A5var reduces the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) while increasing the maximal OCR and ECAR under glucose depletion. These results suggest that activated ISR-induced increased expression of SLC1A5var may regulate mitochondrial oxidative phosphorylation and glycolytic metabolic characteristics to enhance glucose depletion-induced cell death. In conclusion, SLC1A5var plays a vital role in metabolic reprogramming and may be a potential target for breast cancer treatment.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Rd., Beitou Dist., Taipei 112, Taiwan; School of Pharmacy, Taipei Medical University, No. 250, Wuxing St., Xinyi Dist., Taipei 110, Taiwan; Department and Institute of Pharmacology, College of Medicine, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan; Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan.
| | - Yu-Chieh Ho
- Department and Institute of Pharmacology, College of Medicine, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan.
| | - Chian-Ying Chou
- Department of Pharmacy, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Rd., Beitou Dist., Taipei 112, Taiwan; Department of Pharmacy, College of Pharmaceutical Sciences, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan.
| | - Yuh-Lih Chang
- Department of Pharmacy, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Rd., Beitou Dist., Taipei 112, Taiwan; Department of Pharmacy, College of Pharmaceutical Sciences, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan.
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, College of Medicine, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan; Department of Pharmacy, College of Pharmaceutical Sciences, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Rd., Beitou Dist., Taipei 112, Taiwan.
| | - Ling-Ming Tseng
- Division of General Surgery, Department of Surgery, Comprehensive Breast Health Center, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Rd., Beitou Dist., Taipei 112, Taiwan; Department of Surgery, College of Medicine, National Yang-Ming Chiao-Tung University, No. 155, Section 2, Li-Nong St., Beitou Dist., Taipei 112, Taiwan.
| |
Collapse
|
4
|
Na MJ, Lee WY, Park HJ. Difenoconazole Induced Damage of Bovine Mammary Epithelial Cells via ER Stress and Inflammatory Response. Cells 2024; 13:1715. [PMID: 39451231 PMCID: PMC11506304 DOI: 10.3390/cells13201715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Difenoconazole (DIF) is a fungicide used to control various fungi. It is absorbed on the surface of different plants and contributes significantly to increased crop production. However, DIF is reported to exhibit toxicity to fungi and to aquatic plants, fish, and mammals, including humans, causing adverse effects. However, research on the impact of DIF on the mammary epithelial cells of herbivorous bovines is limited. DIF-induced damage and accumulation in the mammary glands can have direct and indirect effects on humans. Therefore, we investigated the effects and mechanisms of DIF toxicity in MAC-T cells. The current study revealed that DIF reduces cell viability and proliferation while triggering apoptotic cell death through the upregulation of pro-apoptotic proteins, including cleaved caspase 3 and Bcl-2-associated X protein (BAX), and the downregulation of leukemia type 2 (BCL-2). DIF also induced endoplasmic reticulum (ER) stress by increasing the expression of genes or proteins of Bip/GRP78, protein disulfide isomerase (PDI), activating transcription factor 4 (ATF4), C/EBP homologous protein (CHOP), and endoplasmic reticulum oxidoreductase 1 Alpha (ERO1-Lα). We demonstrated that DIF induces mitochondria-mediated apoptosis in MAC-T cells by activating ER stress pathways. This cellular damage resulted in a significant increase in the expression of inflammatory response genes and proteins, including cyclooxygenase 2 (COX2), transforming growth factor beta 3 (TGFB3), CCAAT enhancer binding protein delta (CEBPD), and iNOS, in DIF-treated groups. In addition, spheroid formation by MAC-T cells was suppressed by DIF treatment. Our findings suggest that DIF exposure in dairy cows may harm mammary gland function and health and may indirectly affect human consumption of milk.
Collapse
Affiliation(s)
- Myoung-Jun Na
- Department of Animal Biotechnology, College of Life Science, Sangji University, Wonju-si 26339, Republic of Korea
| | - Won-Young Lee
- Department of Livestock, Korea National University of Agriculture and Fisheries, Jeonju-si 54874, Republic of Korea;
| | - Hyun-Jung Park
- Department of Animal Biotechnology, College of Life Science, Sangji University, Wonju-si 26339, Republic of Korea
| |
Collapse
|
5
|
Doll L, Welte K, Skokowa J, Bajoghli B. A JAGN1-associated severe congenital neutropenia zebrafish model revealed an altered G-CSFR signaling and UPR activation. Blood Adv 2024; 8:4050-4065. [PMID: 38739706 PMCID: PMC11342096 DOI: 10.1182/bloodadvances.2023011656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
ABSTRACT A variety of autosomal recessive mutations in the JAGN1 gene cause severe congenital neutropenia (CN). However, the underlying pathomechanism remains poorly understood, mainly because of the limited availability of primary hematopoietic stem cells from JAGN1-CN patients and the absence of animal models. In this study, we aimed to address these limitations by establishing a zebrafish model of JAGN1-CN. We found 2 paralogs of the human JAGN1 gene, namely jagn1a and jagn1b, which play distinct roles during zebrafish hematopoiesis. Using various approaches such as morpholino-based knockdown, CRISPR/Cas9-based gene editing, and misexpression of a jagn1b harboring a specific human mutation, we successfully developed neutropenia while leaving other hematopoietic lineages unaffected. Further analysis of our model revealed significant upregulation of apoptosis and genes involved in the unfolded protein response (UPR). However, neither UPR nor apoptosis is the primary mechanism that leads to neutropenia in zebrafish. Instead, Jagn1b has a critical role in granulocyte colony-stimulating factor receptor signaling and steady-state granulopoiesis, shedding light on the pathogenesis of neutropenia associated with JAGN1 mutations. The establishment of a zebrafish model for JAGN1-CN represents a significant advancement in understanding the specific pathologic pathways underlying the disease. This model provides a valuable in vivo tool for further investigation and exploration of potential therapeutic strategies.
Collapse
Affiliation(s)
- Larissa Doll
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Karl Welte
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Children’s Hospital, University Hospital Tuebingen, Tuebingen, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Gene and RNA Therapy Center, Tuebingen University, Tuebingen, Germany
| | - Baubak Bajoghli
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Austrian BioImaging/CMI, Vienna, Austria
| |
Collapse
|
6
|
Wang W, Liu P, Ma J, Li J, Leng L. Establishment of a CD8+ T cells-related prognostic risk model for acral melanoma based on single-cell and bulk RNA sequencing. Skin Res Technol 2024; 30:e13900. [PMID: 39093712 PMCID: PMC11296306 DOI: 10.1111/srt.13900] [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: 06/29/2024] [Accepted: 07/20/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND CD8+ T cells have been recognized as crucial factors in the prognosis of melanoma. However, there is currently a lack of gene markers that accurately describe their characteristics and functions in acral melanoma (AM), which hinders the development of personalized medicine. METHODS Firstly, we explored the composition differences of immune cells in AM using single-cell RNA sequencing (scRNA-seq) data and comprehensively characterized the immune microenvironment of AM in terms of composition, developmental differentiation, function, and cell communication. Subsequently, we constructed and validated a prognostic risk scoring model based on differentially expressed genes (DEGs) of CD8+ T cells using the TCGA-SKCM cohort through Lasso-Cox method. Lastly, immunofluorescence staining was performed to validate the expression of four genes (ISG20, CCL4, LPAR6, DDIT3) in AM and healthy skin tissues as included in the prognostic model. RESULTS The scRNA-seq data revealed that memory CD8+ T cells accounted for the highest proportion in the immune microenvironment of AM, reaching 70.5%. Cell-cell communication analysis showed extensive communication relationships among effector CD8+ T cells. Subsequently, we constructed a prognostic scoring model based on DEGs derived from CD8+ T cell sources. Four CD8+ T cell-related genes were included in the construction and validation of the prognostic model. Additionally, immunofluorescence results demonstrated that ISG20 and CCL4 were downregulated, while LPAR6 and DDIT3 were upregulated in AM tissues compared to normal skin tissues. CONCLUSION Identifying biomarkers based on the expression levels of CD8+ T cell-related genes may be an effective approach for establishing prognostic models in AM patients. The independently prognostic risk evaluation model we constructed provides new insights and theoretical support for immunotherapy in AM.
Collapse
Affiliation(s)
- Wenwen Wang
- Department of DermatologyPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Stem Cell and Regenerative Medicine LabDepartment of Medical Science Research CenterState Key Laboratory for ComplexSevere, and Rare DiseasesCenter for Translational MedicinePeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pu Liu
- Chongqing Key Laboratory on Big Data for Bio IntelligenceChongqing University of Posts and TelecommunicationsChongqingChina
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijingChina
| | - Jie Ma
- State Key Laboratory of Medical ProteomicsBeijing Proteome Research CenterNational Center for Protein Sciences (Beijing)Beijing Institute of LifeomicsBeijingChina
| | - Jun Li
- Department of DermatologyPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ling Leng
- Stem Cell and Regenerative Medicine LabDepartment of Medical Science Research CenterState Key Laboratory for ComplexSevere, and Rare DiseasesCenter for Translational MedicinePeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
7
|
Nair KA, Liu B. Navigating the landscape of the unfolded protein response in CD8 + T cells. Front Immunol 2024; 15:1427859. [PMID: 39026685 PMCID: PMC11254671 DOI: 10.3389/fimmu.2024.1427859] [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: 05/05/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Endoplasmic reticulum stress occurs due to large amounts of misfolded proteins, hypoxia, nutrient deprivation, and more. The unfolded protein is a complex intracellular signaling network designed to operate under this stress. Composed of three individual arms, inositol-requiring enzyme 1, protein kinase RNA-like ER kinase, and activating transcription factor-6, the unfolded protein response looks to resolve stress and return to proteostasis. The CD8+ T cell is a critical cell type for the adaptive immune system. The unfolded protein response has been shown to have a wide-ranging spectrum of effects on CD8+ T cells. CD8+ T cells undergo cellular stress during activation and due to environmental insults. However, the magnitude of the effects this response has on CD8+ T cells is still understudied. Thus, studying these pathways is important to unraveling the inner machinations of these powerful cells. In this review, we will highlight the recent literature in this field, summarize the three pathways of the unfolded protein response, and discuss their roles in CD8+ T cell biology and functionality.
Collapse
Affiliation(s)
- Keith Alan Nair
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
8
|
Magg V, Manetto A, Kopp K, Wu CC, Naghizadeh M, Lindner D, Eke L, Welsch J, Kallenberger SM, Schott J, Haucke V, Locker N, Stoecklin G, Ruggieri A. Turnover of PPP1R15A mRNA encoding GADD34 controls responsiveness and adaptation to cellular stress. Cell Rep 2024; 43:114069. [PMID: 38602876 DOI: 10.1016/j.celrep.2024.114069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 01/25/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
The integrated stress response (ISR) is a key cellular signaling pathway activated by environmental alterations that represses protein synthesis to restore homeostasis. To prevent sustained damage, the ISR is counteracted by the upregulation of growth arrest and DNA damage-inducible 34 (GADD34), a stress-induced regulatory subunit of protein phosphatase 1 that mediates translation reactivation and stress recovery. Here, we uncover a novel ISR regulatory mechanism that post-transcriptionally controls the stability of PPP1R15A mRNA encoding GADD34. We establish that the 3' untranslated region of PPP1R15A mRNA contains an active AU-rich element (ARE) recognized by proteins of the ZFP36 family, promoting its rapid decay under normal conditions and stabilization for efficient expression of GADD34 in response to stress. We identify the tight temporal control of PPP1R15A mRNA turnover as a component of the transient ISR memory, which sets the threshold for cellular responsiveness and mediates adaptation to repeated stress conditions.
Collapse
Affiliation(s)
- Vera Magg
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Alessandro Manetto
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Katja Kopp
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Chia Ching Wu
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Mohsen Naghizadeh
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany
| | - Doris Lindner
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany
| | - Lucy Eke
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Julia Welsch
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Stefan M Kallenberger
- Digital Health Center, Berlin Institute of Health (BIH) and Charité, 10178 Berlin, Germany; Medical Oncology, National Center for Tumor Diseases, Heidelberg University, 69120 Heidelberg, Germany
| | - Johanna Schott
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany; Freie Universität Berlin, Faculty of Biology, Chemistry, and Pharmacy, 14195 Berlin, Germany
| | - Nicolas Locker
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK; The Pirbright Institute, GU24 0NF Pirbright, UK
| | - Georg Stoecklin
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany; Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany.
| | - Alessia Ruggieri
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany.
| |
Collapse
|
9
|
García-López D, Zaragoza-Ojeda M, Eguía-Aguilar P, Arenas-Huertero F. Endoplasmic Reticulum Stress in Gliomas: Exploiting a Dual-Effect Dysfunction through Chemical Pharmaceutical Compounds and Natural Derivatives for Therapeutical Uses. Int J Mol Sci 2024; 25:4078. [PMID: 38612890 PMCID: PMC11012637 DOI: 10.3390/ijms25074078] [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: 10/31/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 04/14/2024] Open
Abstract
The endoplasmic reticulum maintains proteostasis, which can be disrupted by oxidative stress, nutrient deprivation, hypoxia, lack of ATP, and toxicity caused by xenobiotic compounds, all of which can result in the accumulation of misfolded proteins. These stressors activate the unfolded protein response (UPR), which aims to restore proteostasis and avoid cell death. However, endoplasmic response-associated degradation (ERAD) is sometimes triggered to degrade the misfolded and unassembled proteins instead. If stress persists, cells activate three sensors: PERK, IRE-1, and ATF6. Glioma cells can use these sensors to remain unresponsive to chemotherapeutic treatments. In such cases, the activation of ATF4 via PERK and some proteins via IRE-1 can promote several types of cell death. The search for new antitumor compounds that can successfully and directly induce an endoplasmic reticulum stress response ranges from ligands to oxygen-dependent metabolic pathways in the cell capable of activating cell death pathways. Herein, we discuss the importance of the ER stress mechanism in glioma and likely therapeutic targets within the UPR pathway, as well as chemicals, pharmaceutical compounds, and natural derivatives of potential use against gliomas.
Collapse
Affiliation(s)
- Daniel García-López
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
- Facultad de Ciencia y Tecnología, Universidad Simón Bolívar, Mexico City 03920, Mexico
| | - Montserrat Zaragoza-Ojeda
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
| | - Pilar Eguía-Aguilar
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
- Departamento de Patología Clínica y Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | - Francisco Arenas-Huertero
- Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico; (D.G.-L.); (M.Z.-O.); (P.E.-A.)
- Centro de Investigación en Biomedicina y Bioseguridad, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| |
Collapse
|
10
|
Tang H, Kang R, Liu J, Tang D. ATF4 in cellular stress, ferroptosis, and cancer. Arch Toxicol 2024; 98:1025-1041. [PMID: 38383612 DOI: 10.1007/s00204-024-03681-x] [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: 08/23/2023] [Accepted: 01/15/2024] [Indexed: 02/23/2024]
Abstract
Activating transcription factor 4 (ATF4), a member of the ATF/cAMP response element-binding (CREB) family, plays a critical role as a stress-induced transcription factor. It orchestrates cellular responses, particularly in the management of endoplasmic reticulum stress, amino acid deprivation, and oxidative challenges. ATF4's primary function lies in regulating gene expression to ensure cell survival during stressful conditions. However, when considering its involvement in ferroptosis, characterized by severe lipid peroxidation and pronounced endoplasmic reticulum stress, the ATF4 pathway can either inhibit or promote ferroptosis. This intricate relationship underscores the complexity of cellular responses to varying stress levels. Understanding the connections between ATF4, ferroptosis, and endoplasmic reticulum stress holds promise for innovative cancer therapies, especially in addressing apoptosis-resistant cells. In this review, we provide an overview of ATF4, including its structure, modifications, and functions, and delve into its dual role in both ferroptosis and cancer.
Collapse
Affiliation(s)
- Hu Tang
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
11
|
Kim R, Kin T, Beck WT. Impact of Complex Apoptotic Signaling Pathways on Cancer Cell Sensitivity to Therapy. Cancers (Basel) 2024; 16:984. [PMID: 38473345 DOI: 10.3390/cancers16050984] [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: 01/26/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Anticancer drugs induce apoptotic and non-apoptotic cell death in various cancer types. The signaling pathways for anticancer drug-induced apoptotic cell death have been shown to differ between drug-sensitive and drug-resistant cells. In atypical multidrug-resistant leukemia cells, the c-Jun/activator protein 1 (AP-1)/p53 signaling pathway leading to apoptotic death is altered. Cancer cells treated with anticancer drugs undergo c-Jun/AP-1-mediated apoptotic death and are involved in c-Jun N-terminal kinase activation and growth arrest- and DNA damage-inducible gene 153 (Gadd153)/CCAAT/enhancer-binding protein homologous protein pathway induction, regardless of the p53 genotype. Gadd153 induction is associated with mitochondrial membrane permeabilization after anticancer drug treatment and involves a coupled endoplasmic reticulum stress response. The induction of apoptosis by anticancer drugs is mediated by the intrinsic pathway (cytochrome c, Cyt c) and subsequent activation of the caspase cascade via proapoptotic genes (e.g., Bax and Bcl-xS) and their interactions. Anticancer drug-induced apoptosis involves caspase-dependent and caspase-independent pathways and occurs via intrinsic and extrinsic pathways. The targeting of antiapoptotic genes such as Bcl-2 enhances anticancer drug efficacy. The modulation of apoptotic signaling by Bcl-xS transduction increases the sensitivity of multidrug resistance-related protein-overexpressing epidermoid carcinoma cells to anticancer drugs. The significance of autophagy in cancer therapy remains to be elucidated. In this review, we summarize current knowledge of cancer cell death-related signaling pathways and their alterations during anticancer drug treatment and discuss potential strategies to enhance treatment efficacy.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima 730-0051, Japan
| | - Takanori Kin
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - William T Beck
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Ahlstedt BA, Ganji R, Mukkavalli S, Paulo JA, Gygi SP, Raman M. UBXN1 maintains ER proteostasis and represses UPR activation by modulating translation. EMBO Rep 2024; 25:672-703. [PMID: 38177917 PMCID: PMC10897191 DOI: 10.1038/s44319-023-00027-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
ER protein homeostasis (proteostasis) is essential for proper folding and maturation of proteins in the secretory pathway. Loss of ER proteostasis can lead to the accumulation of misfolded or aberrant proteins in the ER and triggers the unfolded protein response (UPR). In this study, we find that the p97 adaptor UBXN1 is an important negative regulator of the UPR. Loss of UBXN1 sensitizes cells to ER stress and activates the UPR. This leads to widespread upregulation of the ER stress transcriptional program. Using comparative, quantitative proteomics we show that deletion of UBXN1 results in a significant enrichment of proteins involved in ER-quality control processes including those involved in protein folding and import. Notably, we find that loss of UBXN1 does not perturb p97-dependent ER-associated degradation (ERAD). Our studies indicate that loss of UBXN1 increases translation in both resting and ER-stressed cells. Surprisingly, this process is independent of p97 function. Taken together, our studies have identified a new role for UBXN1 in repressing translation and maintaining ER proteostasis in a p97 independent manner.
Collapse
Affiliation(s)
- Brittany A Ahlstedt
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
- ALPCA diagnostics, Salem, NH, USA
| | - Rakesh Ganji
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Sirisha Mukkavalli
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
- Dana Farber Cancer Research Institute, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology Harvard Medical School, Boston, MA, USA
| | - Steve P Gygi
- Department of Cell Biology Harvard Medical School, Boston, MA, USA
| | - Malavika Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
13
|
Kim LC, Lesner NP, Simon MC. Cancer Metabolism under Limiting Oxygen Conditions. Cold Spring Harb Perspect Med 2024; 14:a041542. [PMID: 37848248 PMCID: PMC10835619 DOI: 10.1101/cshperspect.a041542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Molecular oxygen (O2) is essential for cellular bioenergetics and numerous biochemical reactions necessary for life. Solid tumors outgrow the native blood supply and diffusion limits of O2, and therefore must engage hypoxia response pathways that evolved to withstand acute periods of low O2 Hypoxia activates coordinated gene expression programs, primarily through hypoxia inducible factors (HIFs), to support survival. Many of these changes involve metabolic rewiring such as increasing glycolysis to support ATP generation while suppressing mitochondrial metabolism. Since low O2 is often coupled with nutrient stress in the tumor microenvironment, other responses to hypoxia include activation of nutrient uptake pathways, metabolite scavenging, and regulation of stress and growth signaling cascades. Continued development of models that better recapitulate tumors and their microenvironments will lead to greater understanding of oxygen-dependent metabolic reprogramming and lead to more effective cancer therapies.
Collapse
Affiliation(s)
- Laura C Kim
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
14
|
Zhang SX, Wang JJ, Starr CR, Lee EJ, Park KS, Zhylkibayev A, Medina A, Lin JH, Gorbatyuk M. The endoplasmic reticulum: Homeostasis and crosstalk in retinal health and disease. Prog Retin Eye Res 2024; 98:101231. [PMID: 38092262 PMCID: PMC11056313 DOI: 10.1016/j.preteyeres.2023.101231] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The endoplasmic reticulum (ER) is the largest intracellular organelle carrying out a broad range of important cellular functions including protein biosynthesis, folding, and trafficking, lipid and sterol biosynthesis, carbohydrate metabolism, and calcium storage and gated release. In addition, the ER makes close contact with multiple intracellular organelles such as mitochondria and the plasma membrane to actively regulate the biogenesis, remodeling, and function of these organelles. Therefore, maintaining a homeostatic and functional ER is critical for the survival and function of cells. This vital process is implemented through well-orchestrated signaling pathways of the unfolded protein response (UPR). The UPR is activated when misfolded or unfolded proteins accumulate in the ER, a condition known as ER stress, and functions to restore ER homeostasis thus promoting cell survival. However, prolonged activation or dysregulation of the UPR can lead to cell death and other detrimental events such as inflammation and oxidative stress; these processes are implicated in the pathogenesis of many human diseases including retinal disorders. In this review manuscript, we discuss the unique features of the ER and ER stress signaling in the retina and retinal neurons and describe recent advances in the research to uncover the role of ER stress signaling in neurodegenerative retinal diseases including age-related macular degeneration, inherited retinal degeneration, achromatopsia and cone diseases, and diabetic retinopathy. In some chapters, we highlight the complex interactions between the ER and other intracellular organelles focusing on mitochondria and illustrate how ER stress signaling regulates common cellular stress pathways such as autophagy. We also touch upon the integrated stress response in retinal degeneration and diabetic retinopathy. Finally, we provide an update on the current development of pharmacological agents targeting the UPR response and discuss some unresolved questions and knowledge gaps to be addressed by future research.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States.
| | - Josh J Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Christopher R Starr
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eun-Jin Lee
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Karen Sophia Park
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Assylbek Zhylkibayev
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jonathan H Lin
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
15
|
Hicks D, Giresh K, Wrischnik LA, Weiser DC. The PPP1R15 Family of eIF2-alpha Phosphatase Targeting Subunits (GADD34 and CReP). Int J Mol Sci 2023; 24:17321. [PMID: 38139150 PMCID: PMC10743859 DOI: 10.3390/ijms242417321] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The vertebrate PPP1R15 family consists of the proteins GADD34 (growth arrest and DNA damage-inducible protein 34, the product of the PPP1R15A gene) and CReP (constitutive repressor of eIF2α phosphorylation, the product of the PPP1R15B gene), both of which function as targeting/regulatory subunits for protein phosphatase 1 (PP1) by regulating subcellular localization, modulating substrate specificity and assembling complexes with target proteins. The primary cellular function of these proteins is to facilitate the dephosphorylation of eukaryotic initiation factor 2-alpha (eIF2α) by PP1 during cell stress. In this review, we will provide a comprehensive overview of the cellular function, biochemistry and pharmacology of GADD34 and CReP, starting with a brief introduction of eIF2α phosphorylation via the integrated protein response (ISR). We discuss the roles GADD34 and CReP play as feedback inhibitors of the unfolded protein response (UPR) and highlight the critical function they serve as inhibitors of the PERK-dependent branch, which is particularly important since it can mediate cell survival or cell death, depending on how long the stressful stimuli lasts, and GADD34 and CReP play key roles in fine-tuning this cellular decision. We briefly discuss the roles of GADD34 and CReP homologs in model systems and then focus on what we have learned about their function from knockout mice and human patients, followed by a brief review of several diseases in which GADD34 and CReP have been implicated, including cancer, diabetes and especially neurodegenerative disease. Because of the potential importance of GADD34 and CReP in aspects of human health and disease, we will discuss several pharmacological inhibitors of GADD34 and/or CReP that show promise as treatments and the controversies as to their mechanism of action. This review will finish with a discussion of the biochemical properties of GADD34 and CReP, their regulation and the additional interacting partners that may provide insight into the roles these proteins may play in other cellular pathways. We will conclude with a brief outline of critical areas for future study.
Collapse
Affiliation(s)
- Danielle Hicks
- Department of Science, Mathematics and Engineering, Modesto Junior College, Modesto, CA 95350, USA
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Krithika Giresh
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Lisa A. Wrischnik
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Douglas C. Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| |
Collapse
|
16
|
Pan Y, Cai W, Cheng A, Wang M, Huang J, Chen S, Yang Q, Wu Y, Sun D, Mao S, Zhu D, Liu M, Zhao X, Zhang S, Gao Q, Ou X, Tian B, Yin Z, Jia R. Duck Tembusu virus NS3 protein induces apoptosis by activating the PERK/PKR pathway and mitochondrial pathway. J Virol 2023; 97:e0149723. [PMID: 37877719 PMCID: PMC10688375 DOI: 10.1128/jvi.01497-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
IMPORTANCE Duck Tembusu virus (DTMUV) is an emerging pathogenic flavivirus that replicates well in mosquito, bird, and mammalian cells. An in vivo study revealed that BALB/c mice and Kunming mice were susceptible to DTMUV after intracerebral inoculation. Moreover, there are no reports about DTMUV-related human disease, but antibodies against DTMUV and viral RNA were detected in the serum samples of duck industry workers. This information implies that DTMUV has expanded its host range and poses a threat to mammalian health. Thus, understanding the pathogenic mechanism of DTMUV is crucial for identifying potential antiviral targets. In this study, we discovered that NS3 can induce the mitochondria-mediated apoptotic pathway through the PERK/PKR pathway; it can also interact with voltage-dependent anion channel 2 to induce apoptosis. Our findings provide a theoretical basis for understanding the pathogenic mechanism of DTMUV infection and identifying potential antiviral targets and may also serve as a reference for exploring the pathogenesis of other flaviviruses.
Collapse
Affiliation(s)
- Yuhong Pan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Wenjun Cai
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Juan Huang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Di Sun
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Sai Mao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Qun Gao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Xumin Ou
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Bin Tian
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Vived C, Lee-Papastavros A, Aparecida da Silva Pereira J, Yi P, MacDonald TL. β Cell Stress and Endocrine Function During T1D: What Is Next to Discover? Endocrinology 2023; 165:bqad162. [PMID: 37947352 DOI: 10.1210/endocr/bqad162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/27/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Canonically, type 1 diabetes (T1D) is a disease characterized by autoreactive T cells as perpetrators of endocrine dysfunction and β cell death in the spiral toward loss of β cell mass, hyperglycemia, and insulin dependence. β Cells have mostly been considered as bystanders in a flurry of autoimmune processes. More recently, our framework for understanding and investigating T1D has evolved. It appears increasingly likely that intracellular β cell stress is an important component of T1D etiology/pathology that perpetuates autoimmunity during the progression to T1D. Here we discuss the emerging and complex role of β cell stress in initiating, provoking, and catalyzing T1D. We outline the bridges between hyperglycemia, endoplasmic reticulum stress, oxidative stress, and autoimmunity from the viewpoint of intrinsic β cell (dys)function, and we extend this discussion to the potential role for a therapeutic β cell stress-metabolism axis in T1D. Lastly, we mention research angles that may be pursued to improve β cell endocrine function during T1D. Biology gleaned from studying T1D will certainly overlap to innovate therapeutic strategies for T2D, and also enhance the pursuit of creating optimized stem cell-derived β cells as endocrine therapy.
Collapse
Affiliation(s)
- Celia Vived
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jéssica Aparecida da Silva Pereira
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Peng Yi
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Tara L MacDonald
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
18
|
Tak J, Kim SG. Effects of toxicants on endoplasmic reticulum stress and hepatic cell fate determination. Toxicol Res 2023; 39:533-547. [PMID: 37779594 PMCID: PMC10541383 DOI: 10.1007/s43188-023-00201-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 10/03/2023] Open
Abstract
Toxicant-induced injury is a significant global health issue. However, the mechanisms through which toxicants such as carbon tetrachloride, acetaminophen, dimethylformamide, cocaine, and morphine induce the death of multiple cell types and contribute to liver toxicity are highly complex. This phenomenon involves intricate signaling pathways in association with oxidative stress, inflammation, and activation of death receptors, which are closely linked to endoplasmic reticulum (ER) stress. ER stress initially triggers the unfolded protein response, which either promotes cell survival or causes cell death at later times, depending on the severity and duration of the stress. Thus, comprehending the molecular basis governing cell fate determination in the context of ER stress may provide key insights into the prevention and treatment of toxicant-induced injury. This review summarizes our current understanding of agents that trigger different forms of ER stress-mediated cell death, necroptosis, ferroptosis, pyroptosis, and apoptosis, and covers the underlying molecular basis of toxicant-induced ER stress, as well as potential target molecules.
Collapse
Affiliation(s)
- Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Kyeonggi-do 10326 Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Kyeonggi-do 10326 Republic of Korea
| |
Collapse
|
19
|
Hanquier Z, Misra J, Baxter R, Maiers JL. Stress and Liver Fibrogenesis: Understanding the Role and Regulation of Stress Response Pathways in Hepatic Stellate Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1363-1376. [PMID: 37422148 PMCID: PMC10548279 DOI: 10.1016/j.ajpath.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 07/10/2023]
Abstract
Stress response pathways are crucial for cells to adapt to physiological and pathologic conditions. Increased transcription and translation in response to stimuli place a strain on the cell, necessitating increased amino acid supply, protein production and folding, and disposal of misfolded proteins. Stress response pathways, such as the unfolded protein response (UPR) and the integrated stress response (ISR), allow cells to adapt to stress and restore homeostasis; however, their role and regulation in pathologic conditions, such as hepatic fibrogenesis, are unclear. Liver injury promotes fibrogenesis through activation of hepatic stellate cells (HSCs), which produce and secrete fibrogenic proteins to promote tissue repair. This process is exacerbated in chronic liver disease, leading to fibrosis and, if unchecked, cirrhosis. Fibrogenic HSCs exhibit activation of both the UPR and ISR, due in part to increased transcriptional and translational demands, and these stress responses play important roles in fibrogenesis. Targeting these pathways to limit fibrogenesis or promote HSC apoptosis is a potential antifibrotic strategy, but it is limited by our lack of mechanistic understanding of how the UPR and ISR regulate HSC activation and fibrogenesis. This article explores the role of the UPR and ISR in the progression of fibrogenesis, and highlights areas that require further investigation to better understand how the UPR and ISR can be targeted to limit hepatic fibrosis progression.
Collapse
Affiliation(s)
- Zachary Hanquier
- Department of Molecular and Medical Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jagannath Misra
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Reese Baxter
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jessica L Maiers
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
20
|
Wek RC, Anthony TG, Staschke KA. Surviving and Adapting to Stress: Translational Control and the Integrated Stress Response. Antioxid Redox Signal 2023; 39:351-373. [PMID: 36943285 PMCID: PMC10443206 DOI: 10.1089/ars.2022.0123] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/23/2023]
Abstract
Significance: Organisms adapt to changing environments by engaging cellular stress response pathways that serve to restore proteostasis and enhance survival. A primary adaptive mechanism is the integrated stress response (ISR), which features phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eIF2). Four eIF2α kinases respond to different stresses, enabling cells to rapidly control translation to optimize management of resources and reprogram gene expression for stress adaptation. Phosphorylation of eIF2 blocks its guanine nucleotide exchange factor, eIF2B, thus lowering the levels of eIF2 bound to GTP that is required to deliver initiator transfer RNA (tRNA) to ribosomes. While bulk messenger RNA (mRNA) translation can be sharply lowered by heightened phosphorylation of eIF2α, there are other gene transcripts whose translation is unchanged or preferentially translated. Among the preferentially translated genes is ATF4, which directs transcription of adaptive genes in the ISR. Recent Advances and Critical Issues: This review focuses on how eIF2α kinases function as first responders of stress, the mechanisms by which eIF2α phosphorylation and other stress signals regulate the exchange activity of eIF2B, and the processes by which the ISR triggers differential mRNA translation. To illustrate the synergy between stress pathways, we describe the mechanisms and functional significance of communication between the ISR and another key regulator of translation, mammalian/mechanistic target of rapamycin complex 1 (mTORC1), during acute and chronic amino acid insufficiency. Finally, we discuss the pathological conditions that stem from aberrant regulation of the ISR, as well as therapeutic strategies targeting the ISR to alleviate disease. Future Directions: Important topics for future ISR research are strategies for modulating this stress pathway in disease conditions and drug development, molecular processes for differential translation and the coordinate regulation of GCN2 and other stress pathways during physiological and pathological conditions. Antioxid. Redox Signal. 39, 351-373.
Collapse
Affiliation(s)
- Ronald C. Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| | - Tracy G. Anthony
- Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey, USA
| | - Kirk A. Staschke
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA
| |
Collapse
|
21
|
Zhao N, Li N, Wang T. PERK prevents rhodopsin degradation during retinitis pigmentosa by inhibiting IRE1-induced autophagy. J Cell Biol 2023; 222:e202208147. [PMID: 37022709 PMCID: PMC10082367 DOI: 10.1083/jcb.202208147] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/04/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023] Open
Abstract
Chronic endoplasmic reticulum (ER) stress is the underlying cause of many degenerative diseases, including autosomal dominant retinitis pigmentosa (adRP). In adRP, mutant rhodopsins accumulate and cause ER stress. This destabilizes wild-type rhodopsin and triggers photoreceptor cell degeneration. To reveal the mechanisms by which these mutant rhodopsins exert their dominant-negative effects, we established an in vivo fluorescence reporter system to monitor mutant and wild-type rhodopsin in Drosophila. By performing a genome-wide genetic screen, we found that PERK signaling plays a key role in maintaining rhodopsin homeostasis by attenuating IRE1 activities. Degradation of wild-type rhodopsin is mediated by selective autophagy of ER, which is induced by uncontrolled IRE1/XBP1 signaling and insufficient proteasome activities. Moreover, upregulation of PERK signaling prevents autophagy and suppresses retinal degeneration in the adRP model. These findings establish a pathological role for autophagy in this neurodegenerative condition and indicate that promoting PERK activity could be used to treat ER stress-related neuropathies, including adRP.
Collapse
Affiliation(s)
- Ning Zhao
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ning Li
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Tao Wang
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- College of Biological Sciences, China Agricultural University, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
- National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
22
|
Ong G, Logue SE. Unfolding the Interactions between Endoplasmic Reticulum Stress and Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12050981. [PMID: 37237847 DOI: 10.3390/antiox12050981] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress is caused by an imbalance in cellular redox state due to the accumulation of reactive oxygen species (ROS). While homeostatic levels of ROS are important for cell physiology and signaling, excess ROS can induce a variety of negative effects ranging from damage to biological macromolecules to cell death. Additionally, oxidative stress can disrupt the function of redox-sensitive organelles including the mitochondria and endoplasmic reticulum (ER). In the case of the ER, the accumulation of misfolded proteins can arise due to oxidative stress, leading to the onset of ER stress. To combat ER stress, cells initiate a highly conserved stress response called the unfolded protein response (UPR). While UPR signaling, within the context of resolving ER stress, is well characterised, how UPR mediators respond to and influence oxidative stress is less defined. In this review, we evaluate the interplay between oxidative stress, ER stress and UPR signaling networks. Specifically, we assess how UPR signaling mediators can influence antioxidant responses.
Collapse
Affiliation(s)
- Gideon Ong
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Susan E Logue
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
23
|
Wang Y, Li J, Zhang Z, Wang R, Bo H, Zhang Y. Exercise Improves the Coordination of the Mitochondrial Unfolded Protein Response and Mitophagy in Aging Skeletal Muscle. Life (Basel) 2023; 13:life13041006. [PMID: 37109535 PMCID: PMC10142204 DOI: 10.3390/life13041006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) and mitophagy are two mitochondrial quality control (MQC) systems that work at the molecular and organelle levels, respectively, to maintain mitochondrial homeostasis. Under stress conditions, these two processes are simultaneously activated and compensate for each other when one process is insufficient, indicating mechanistic coordination between the UPRmt and mitophagy that is likely controlled by common upstream signals. This review focuses on the molecular signals regulating this coordination and presents evidence showing that this coordination mechanism is impaired during aging and promoted by exercise. Furthermore, the bidirectional regulation of reactive oxygen species (ROS) and AMPK in modulating this mechanism is discussed. The hierarchical surveillance network of MQC can be targeted by exercise-derived ROS to attenuate aging, which offers a molecular basis for potential therapeutic interventions for sarcopenia.
Collapse
Affiliation(s)
- Yan Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
- School of Physical Education, Guangdong Institute of Petrochemical Technology, Maoming 525000, China
| | - Jialin Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| | - Ziyi Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| | - Runzi Wang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| | - Hai Bo
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
- Department of Military Training Medicines, Logistics University of Chinese People's Armed Police Force, Tianjin 300162, China
| | - Yong Zhang
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, School of Exercise and Health, Tianjin University of Sport, Tianjin 301617, China
| |
Collapse
|
24
|
Choi BE, Shin S, Evans S, Singh BB, Bandyopadhyay BC. Ablation of TRPC3 disrupts Ca 2+ signaling in salivary ductal cells and promotes sialolithiasis. Sci Rep 2023; 13:5772. [PMID: 37031239 PMCID: PMC10082769 DOI: 10.1038/s41598-023-32602-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/30/2023] [Indexed: 04/10/2023] Open
Abstract
Clinical studies and structural analyses of salivary stones strongly suggest a linkage between higher saliva calcium (Ca2+) and salivary stone formation, sialolithiasis; however, the process and the mechanism leading to Ca2+ overload during sialolithiasis is not well understood. Here, we show that TRPC3 null (-/-) mice presented with a reduction in Ca2+ entry and current in ductal cells with higher saliva [Ca2+] suggesting diminished transepithelial Ca2+ flux across the salivary ductal cells, leaving more Ca2+ in ductal fluid. Significantly, we found that TRPC3 was expressed in mice and human salivary ductal cells, while intraductal stones were detected in both mice (TRPC3-/-) and patient (sialolithiasis) salivary glands. To identify the mechanism, we found that TRPC3 was crucial in preventing the expression of calcification genes (BMP2/6, Runx2) in ductal cells which may be due to higher extracellular Ca2+ in SMG tissues. Similarly, inflammatory (IL6, NLRP3), fibrotic (FN1, TGFβ1) and apoptotic (Bax1/Bcl2) markers were also elevated, suggesting that the loss of TRPC3 induces genetic changes that leads to salivary gland cell death and induction of inflammatory response. Overall, ablation of TRPC3-/- leads to higher saliva [Ca2+], along with elevated detrimental gene expressions, altogether contributing to salivary gland stone formation.
Collapse
Affiliation(s)
- Bok-Eum Choi
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
| | - Samuel Shin
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC, 20064, USA
| | - Sade Evans
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA
| | - Brij B Singh
- Department of Periodontics, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, 151 Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC, 20422, USA.
- Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC, 20064, USA.
| |
Collapse
|
25
|
Osman A, Lindén M, Österlund T, Vannas C, Andersson L, Escobar M, Ståhlberg A, Åman P. Identification of genomic binding sites and direct target genes for the transcription factor DDIT3/CHOP. Exp Cell Res 2023; 422:113418. [PMID: 36402425 DOI: 10.1016/j.yexcr.2022.113418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
DDIT3 is a tightly regulated basic leucine zipper (bZIP) transcription factor and key regulator in cellular stress responses. It is involved in a variety of pathological conditions and may cause cell cycle block and apoptosis. It is also implicated in differentiation of some specialized cell types and as an oncogene in several types of cancer. DDIT3 was originally believed to act as a dominant-negative inhibitor by forming heterodimers with other bZIP transcription factors, preventing their DNA binding and transactivating functions. DDIT3 has, however, been reported to bind DNA and regulate target genes. Here, we employed ChIP sequencing combined with microarray-based expression analysis to identify direct binding motifs and target genes of DDIT3. The results reveal DDIT3 binding to motifs similar to other bZIP transcription factors, known to form heterodimers with DDIT3. Binding to a class III satellite DNA repeat sequence was also detected. DDIT3 acted as a DNA-binding transcription factor and bound mainly to the promotor region of regulated genes. ChIP sequencing analysis of histone H3K27 methylation and acetylation showed a strong overlap between H3K27-acetylated marks and DDIT3 binding. These results support a role for DDIT3 as a transcriptional regulator of H3K27ac-marked genes in transcriptionally active chromatin.
Collapse
Affiliation(s)
- Ayman Osman
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Malin Lindén
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tobias Österlund
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden
| | - Christoffer Vannas
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lisa Andersson
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mandy Escobar
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden
| | - Pierre Åman
- Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
26
|
Bartoszewska S, Collawn JF, Bartoszewski R. The Role of the Hypoxia-Related Unfolded Protein Response (UPR) in the Tumor Microenvironment. Cancers (Basel) 2022; 14:4870. [PMID: 36230792 PMCID: PMC9562011 DOI: 10.3390/cancers14194870] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/19/2022] Open
Abstract
Despite our understanding of the unfolded protein response (UPR) pathways, the crosstalk between the UPR and the complex signaling networks that different cancers utilize for cell survival remains to be, in most cases, a difficult research barrier. A major problem is the constant variability of different cancer types and the different stages of cancer as well as the complexity of the tumor microenvironments (TME). This complexity often leads to apparently contradictory results. Furthermore, the majority of the studies that have been conducted have utilized two-dimensional in vitro cultures of cancer cells that were exposed to continuous hypoxia, and this approach may not mimic the dynamic and cyclic conditions that are found in solid tumors. Here, we discuss the role of intermittent hypoxia, one of inducers of the UPR in the cellular component of TME, and the way in which intermittent hypoxia induces high levels of reactive oxygen species, the activation of the UPR, and the way in which cancer cells modulate the UPR to aid in their survival. Although the past decade has resulted in defining the complex, novel non-coding RNA-based regulatory networks that modulate the means by which hypoxia influences the UPR, we are now just to beginning to understand some of the connections between hypoxia, the UPR, and the TME.
Collapse
Affiliation(s)
- Sylwia Bartoszewska
- Department of Inorganic Chemistry, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - James F. Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383 Wroclaw, Poland
| |
Collapse
|
27
|
Restoring TRAILR2/DR5-Mediated Activation of Apoptosis upon Endoplasmic Reticulum Stress as a Therapeutic Strategy in Cancer. Int J Mol Sci 2022; 23:ijms23168987. [PMID: 36012252 PMCID: PMC9409255 DOI: 10.3390/ijms23168987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
The uncontrolled proliferation of malignant cells in growing tumors results in the generation of different stressors in the tumor microenvironment, such as nutrient shortage, hypoxia and acidosis, among others, that disrupt endoplasmic reticulum (ER) homeostasis and may lead to ER stress. As a response to ER stress, both normal and tumor cells launch a set of signaling pathways known as the unfolded protein response (UPR) to restore ER proteostasis and maintain cell viability and function. However, under sustained ER stress, an apoptotic cell death process can be induced and this has been the subject of different review articles, although the role of the TRAIL-R2/DR5-activated extrinsic pathway of apoptosis has not yet been thoroughly summarized. In this Review, we provide an updated overview of the molecular mechanisms regulating cell fate decisions in tumor cells undergoing ER stress and discuss the role of the tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor 2 (TRAIL-R2/DR5) in the final outcome of UPR signaling. Particularly, we focus on the mechanisms controlling cellular FLICE-like inhibitory protein (FLIP) levels in tumor cells undergoing ER stress, which may represent a potential target for therapeutic intervention in cancer.
Collapse
|
28
|
Carreras-Sureda A, Kroemer G, Cardenas JC, Hetz C. Balancing energy and protein homeostasis at ER-mitochondria contact sites. Sci Signal 2022; 15:eabm7524. [DOI: 10.1126/scisignal.abm7524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The endoplasmic reticulum (ER) is the largest organelle of the cell and participates in multiple essential functions, including the production of secretory proteins, lipid synthesis, and calcium storage. Sustaining proteostasis requires an intimate coupling with energy production. Mitochondrial respiration evolved to be functionally connected to ER physiology through a physical interface between both organelles known as mitochondria-associated membranes. This quasi-synaptic structure acts as a signaling hub that tunes the function of both organelles in a bidirectional manner and controls proteostasis, cell death pathways, and mitochondrial bioenergetics. Here, we discuss the main signaling mechanisms governing interorganellar communication and their putative role in diseases including cancer and neurodegeneration.
Collapse
Affiliation(s)
- Amado Carreras-Sureda
- Department of Cell Physiology and Metabolism, University of Geneva, 1, rue Michel-Servet, 1211 Geneva, Switzerland
| | - 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, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Julio Cesar Cardenas
- Center for Integrative Biology, Mayor University, 7510041 Santiago, Chile
- Center for Geroscience, Brain Health, and Metabolism, 70086 Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Claudio Hetz
- Center for Geroscience, Brain Health, and Metabolism, 70086 Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, 70086 Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, 70086 Santiago, Chile
| |
Collapse
|
29
|
Riess C, del Moral K, Fiebig A, Kaps P, Linke C, Hinz B, Rupprecht A, Frank M, Fiedler T, Koczan D, Troschke-Meurer S, Lode HN, Engel N, Freitag T, Classen CF, Maletzki C. Implementation of a combined CDK inhibition and arginine-deprivation approach to target arginine-auxotrophic glioblastoma multiforme cells. Cell Death Dis 2022; 13:555. [PMID: 35717443 PMCID: PMC9206658 DOI: 10.1038/s41419-022-05006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/18/2022] [Accepted: 06/08/2022] [Indexed: 01/21/2023]
Abstract
Constitutive activation of cyclin-dependent kinases (CDKs) or arginine auxotrophy are hallmarks of Glioblastoma multiforme (GBM). The latter metabolic defect renders tumor cells vulnerable to arginine-depleting substances, such as arginine deiminase from Streptococcus pyogenes (SpyADI). Previously, we confirmed the susceptibility of patient-derived GBM cells towards SpyADI as well as CDK inhibitors (CDKis). To improve therapeutic effects, we here applied a combined approach based on SpyADI and CDKis (dinaciclib, abemaciclib). Three arginine-auxotrophic patient-derived GBM lines with different molecular characteristics were cultured in 2D and 3D and effects of this combined SpyADI/CDKi approach were analyzed in-depth. All CDKi/SpyADI combinations yielded synergistic antitumoral effects, especially when given sequentially (SEQ), i.e., CDKi in first-line and most pronounced in the 3D models. SEQ application demonstrated impaired cell proliferation, invasiveness, and viability. Mitochondrial impairment was demonstrated by increasing mitochondrial membrane potential and decreasing oxygen consumption rate and extracellular acidification rate after SpyADI/abemaciclib monotherapy or its combination regimens. The combined treatment even induced autophagy in target cells (abemaciclib/SpyADI > dinaciclib/SpyADI). By contrast, the unfolded protein response and p53/p21 induced senescence played a minor role. Transmission electron microscopy confirmed damaged mitochondria and endoplasmic reticulum together with increased vacuolization under CDKi mono- and combination therapy. SEQ-abemaciclib/SpyADI treatment suppressed the DSB repair system via NHEJ and HR, whereas SEQ-dinaciclib/SpyADI treatment increased γ-H2AX accumulation and induced Rad51/Ku80. The latter combination also activated the stress sensor GADD45 and β-catenin antagonist AXIN2 and induced expression changes of genes involved in cellular/cytoskeletal integrity. This study highlights the strong antitumoral potential of a combined arginine deprivation and CDK inhibition approach via complex effects on mitochondrial dysfunction, invasiveness as well as DNA-damage response. This provides a good starting point for further in vitro and in vivo proof-of-concept studies to move forward with this strategy.
Collapse
Affiliation(s)
- Christin Riess
- grid.413108.f0000 0000 9737 0454University Children’s Hospital, Rostock University Medical Center, Ernst-Heydemann-Straße 8, 18057 Rostock, Germany ,grid.413108.f0000 0000 9737 0454Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Straße 6, 18057 Rostock, Germany
| | - Katharina del Moral
- grid.413108.f0000 0000 9737 0454University Children’s Hospital, Rostock University Medical Center, Ernst-Heydemann-Straße 8, 18057 Rostock, Germany
| | - Adina Fiebig
- grid.413108.f0000 0000 9737 0454Institute for Medical Microbiology, Virology, and Hygiene, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Philipp Kaps
- grid.413108.f0000 0000 9737 0454University Children’s Hospital, Rostock University Medical Center, Ernst-Heydemann-Straße 8, 18057 Rostock, Germany ,grid.413108.f0000 0000 9737 0454Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Straße 6, 18057 Rostock, Germany ,grid.413108.f0000 0000 9737 0454Institute for Medical Microbiology, Virology, and Hygiene, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Charlotte Linke
- grid.413108.f0000 0000 9737 0454University Children’s Hospital, Rostock University Medical Center, Ernst-Heydemann-Straße 8, 18057 Rostock, Germany
| | - Burkhard Hinz
- grid.413108.f0000 0000 9737 0454Institute for Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Anne Rupprecht
- grid.413108.f0000 0000 9737 0454Institute for Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Marcus Frank
- grid.413108.f0000 0000 9737 0454Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Rostock, Germany ,grid.10493.3f0000000121858338Department of Life, Light & Matter, University of Rostock, Rostock, Germany
| | - Tomas Fiedler
- grid.413108.f0000 0000 9737 0454Institute for Medical Microbiology, Virology, and Hygiene, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany
| | - Dirk Koczan
- grid.10493.3f0000000121858338Institute for Immunology, University of Rostock, 18055 Rostock, Germany
| | - Sascha Troschke-Meurer
- grid.5603.0Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany
| | - Holger N. Lode
- grid.5603.0Department of Pediatric Oncology and Hematology, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse, 17475 Greifswald, Germany
| | - Nadja Engel
- grid.413108.f0000 0000 9737 0454Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, Rostock University Medical Center, Rostock, Germany
| | - Thomas Freitag
- grid.413108.f0000 0000 9737 0454Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Straße 6, 18057 Rostock, Germany
| | - Carl Friedrich Classen
- grid.413108.f0000 0000 9737 0454University Children’s Hospital, Rostock University Medical Center, Ernst-Heydemann-Straße 8, 18057 Rostock, Germany
| | - Claudia Maletzki
- grid.413108.f0000 0000 9737 0454Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Straße 6, 18057 Rostock, Germany
| |
Collapse
|
30
|
Dyskerin Downregulation Can Induce ER Stress and Promote Autophagy via AKT-mTOR Signaling Deregulation. Biomedicines 2022; 10:biomedicines10051092. [PMID: 35625829 PMCID: PMC9138296 DOI: 10.3390/biomedicines10051092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/05/2023] Open
Abstract
Dyskerin is an evolutionarily conserved nucleolar protein implicated in a wide range of fundamental biological roles, including telomere maintenance and ribosome biogenesis. Germline mutations of DKC1, the human gene encoding dyskerin, cause the hereditary disorders known as X-linked dyskeratosis congenita (X-DC). Moreover, dyskerin is upregulated in several cancers. Due to the pleiotropic functions of dyskerin, the X-DC clinical features overlap with those of both telomeropathies and ribosomopathies. In this paper, we evaluate the telomerase-independent effects of dyskerin depletion on cellular physiology by using inducible DCK1 knockdown. This system allows the downregulation of DKC1 expression within a short timeframe. We report that, in these cellular systems, dyskerin depletion induces the accumulation of unfolded/misfolded proteins in the endoplasmic reticulum, which in turn induces the activation of the PERK branch of the unfolded protein response. We also demonstrate that the PERK-eIF2a-ATF4-CHOP signaling pathway, activated by dyskerin downregulation, triggers a functional autophagic flux through the inhibition of the PI3K/AKT/mTOR pathway. By revealing a novel unpredicted connection between the loss of dyskerin, autophagy and UPR, our results establish a firm link between the lowering of dyskerin levels and the activation of the ER stress response, that plays a key role in the pathogenesis of several diseases.
Collapse
|
31
|
Su S, Zhang D, Liu J, Zhao H, Tang X, Che H, Wang Q, Ren W, Zhen D. Folate ameliorates homocysteine-induced osteoblast dysfunction by reducing endoplasmic reticulum stress-activated PERK/ATF-4/CHOP pathway in MC3T3-E1 cells. J Bone Miner Metab 2022; 40:422-433. [PMID: 35190897 DOI: 10.1007/s00774-022-01313-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 01/14/2022] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Homocysteine (Hcy) is considered a newly identified risk factor for osteoporosis. Nevertheless, the underlying mechanism of folate (FA), a key factor in the metabolism of Hcy, in protection against osteoblast dysfunction remains unclear. The purpose of this study was to investigate the mechanism by which FA attenuates Hcy-induced osteoblast damage. MATERIALS AND METHODS The Hcy-induced MC3T3-E1 cells were treated with different concentrations of FA. Cell morphology, cell density, cell proliferation ability, alkaline phosphatase (ALP) activity and mineralization capacity were observed and determined; the gene expression of B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (BAX) and ERS-associated factors, including glucose-regulated protein 78 (GRP-78), activating transcription factor 4 (ATF-4) and growth arrest and DNA damage inducible gene 153 (CHOP/GADD153), were assessed by RT-PCR; and protein levels of GRP-78 and ATF-4 were analyzed by western blotting. RESULTS Hcy suppressed the proliferation, differentiation and mineralization ability of MC3T3-E1 cells in a concentration-dependent manner and activated the ERS signaling pathway. After intervention with different concentrations of FA, the cell viability and density, ALP activity, number of mineralized nodules, calcium content and Bcl-2 gene expression were all significantly increased, whereas the gene expression of GRP-78, CHOP/GADD153, ATF-4 and Bax was markedly downregulated, and protein levels of GRP-78 and ATF-4 were also markedly decreased. CONCLUSION The adverse effects of Hcy on osteoblast differentiation are dose dependent. FA not only protects against osteoblasts apoptosis but also has a direct osteogenic effect on Hcy-induced osteoblasts, which could be partially mediated by inhibition of the PERK-activated ERS pathway.
Collapse
Affiliation(s)
- Shan Su
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Di Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Jinjin Liu
- Department of Endocrinology, The First Hospital of Lanzhou University, 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Haiyan Zhao
- Department of Paediatrics, Gansu Province People's Hospital, Lanzhou, 730000, Gansu Province, China
| | - Xulei Tang
- Department of Endocrinology, The First Hospital of Lanzhou University, 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China
| | - Hongxia Che
- Department of Endocrinology, The Third People's Hospital, Lanzhou, 730000, Gansu Province, China
| | - Qiangmei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Wanna Ren
- Department of Opthalmology, The Second Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Donghu Zhen
- Department of Endocrinology, The First Hospital of Lanzhou University, 1 Donggang West Road, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
32
|
Ebert SM, Rasmussen BB, Judge AR, Judge SM, Larsson L, Wek RC, Anthony TG, Marcotte GR, Miller MJ, Yorek MA, Vella A, Volpi E, Stern JI, Strub MD, Ryan Z, Talley JJ, Adams CM. Biology of Activating Transcription Factor 4 (ATF4) and Its Role in Skeletal Muscle Atrophy. J Nutr 2022; 152:926-938. [PMID: 34958390 PMCID: PMC8970988 DOI: 10.1093/jn/nxab440] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
Activating transcription factor 4 (ATF4) is a multifunctional transcription regulatory protein in the basic leucine zipper superfamily. ATF4 can be expressed in most if not all mammalian cell types, and it can participate in a variety of cellular responses to specific environmental stresses, intracellular derangements, or growth factors. Because ATF4 is involved in a wide range of biological processes, its roles in human health and disease are not yet fully understood. Much of our current knowledge about ATF4 comes from investigations in cultured cell models, where ATF4 was originally characterized and where further investigations continue to provide new insights. ATF4 is also an increasingly prominent topic of in vivo investigations in fully differentiated mammalian cell types, where our current understanding of ATF4 is less complete. Here, we review some important high-level concepts and questions concerning the basic biology of ATF4. We then discuss current knowledge and emerging questions about the in vivo role of ATF4 in one fully differentiated cell type, mammalian skeletal muscle fibers.
Collapse
Affiliation(s)
- Scott M Ebert
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Emmyon, Inc., Rochester, MN, USA
| | - Blake B Rasmussen
- Emmyon, Inc., Rochester, MN, USA
- Department of Nutrition, Metabolism and Rehabilitation Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Andrew R Judge
- Emmyon, Inc., Rochester, MN, USA
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Sarah M Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska, Stockholm, Sweden
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - George R Marcotte
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Matthew J Miller
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Mark A Yorek
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
- Department of Internal Medicine, Iowa City VA Medical Center, Iowa City, IA, USA
| | - Adrian Vella
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Emmyon, Inc., Rochester, MN, USA
| | - Elena Volpi
- Department of Nutrition, Metabolism and Rehabilitation Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Jennifer I Stern
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Matthew D Strub
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Zachary Ryan
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | | | - Christopher M Adams
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
- Emmyon, Inc., Rochester, MN, USA
- Department of Internal Medicine, Iowa City VA Medical Center, Iowa City, IA, USA
| |
Collapse
|
33
|
Klein P, Kallenberger SM, Roth H, Roth K, Ly-Hartig TBN, Magg V, Aleš J, Talemi SR, Qiang Y, Wolf S, Oleksiuk O, Kurilov R, Di Ventura B, Bartenschlager R, Eils R, Rohr K, Hamprecht FA, Höfer T, Fackler OT, Stoecklin G, Ruggieri A. Temporal control of the integrated stress response by a stochastic molecular switch. SCIENCE ADVANCES 2022; 8:eabk2022. [PMID: 35319985 PMCID: PMC8942376 DOI: 10.1126/sciadv.abk2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Stress granules (SGs) are formed in the cytosol as an acute response to environmental cues and activation of the integrated stress response (ISR), a central signaling pathway controlling protein synthesis. Using chronic virus infection as stress model, we previously uncovered a unique temporal control of the ISR resulting in recurrent phases of SG assembly and disassembly. Here, we elucidate the molecular network generating this fluctuating stress response by integrating quantitative experiments with mathematical modeling and find that the ISR operates as a stochastic switch. Key elements controlling this switch are the cooperative activation of the stress-sensing kinase PKR, the ultrasensitive response of SG formation to the phosphorylation of the translation initiation factor eIF2α, and negative feedback via GADD34, a stress-induced subunit of protein phosphatase 1. We identify GADD34 messenger RNA levels as the molecular memory of the ISR that plays a central role in cell adaptation to acute and chronic stress.
Collapse
Affiliation(s)
- Philipp Klein
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Stefan M. Kallenberger
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Digital Health Center, Berlin Institute of Health (BIH) and Charité, Berlin, Germany
- Medical Oncology, National Center for Tumor Diseases, Heidelberg University, Heidelberg, Germany
| | - Hanna Roth
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Karsten Roth
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Thi Bach Nga Ly-Hartig
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Vera Magg
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Janez Aleš
- HCI/IWR, Heidelberg University, Heidelberg, Germany
| | - Soheil Rastgou Talemi
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yu Qiang
- Biomedical Computer Vision Group, BioQuant, IPMB, Heidelberg University, Heidelberg, Germany
| | - Steffen Wolf
- HCI/IWR, Heidelberg University, Heidelberg, Germany
| | - Olga Oleksiuk
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Roma Kurilov
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Di Ventura
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland Eils
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Digital Health Center, Berlin Institute of Health (BIH) and Charité, Berlin, Germany
| | - Karl Rohr
- Biomedical Computer Vision Group, BioQuant, IPMB, Heidelberg University, Heidelberg, Germany
| | | | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver T. Fackler
- Department of Infectious Diseases, Integrative Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
| | - Georg Stoecklin
- Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Alessia Ruggieri
- Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Diseases Research, Heidelberg University, Heidelberg, Germany
- Corresponding author.
| |
Collapse
|
34
|
Sidhom E, O’Brien JT, Butcher AJ, Smith HL, Mallucci GR, Underwood BR. Targeting the Unfolded Protein Response as a Disease-Modifying Pathway in Dementia. Int J Mol Sci 2022; 23:2021. [PMID: 35216136 PMCID: PMC8877151 DOI: 10.3390/ijms23042021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 01/27/2023] Open
Abstract
Dementia is a global medical and societal challenge; it has devastating personal, social and economic costs, which will increase rapidly as the world's population ages. Despite this, there are no disease-modifying treatments for dementia; current therapy modestly improves symptoms but does not change the outcome. Therefore, new treatments are urgently needed-particularly any that can slow down the disease's progression. Many of the neurodegenerative diseases that lead to dementia are characterised by common pathological responses to abnormal protein production and misfolding in brain cells, raising the possibility of the broad application of therapeutics that target these common processes. The unfolded protein response (UPR) is one such mechanism. The UPR is a highly conserved cellular stress response to abnormal protein folding and is widely dysregulated in neurodegenerative diseases. In this review, we describe the basic machinery of the UPR, as well as the evidence for its overactivation and pathogenicity in dementia, and for the marked neuroprotective effects of its therapeutic manipulation in murine models of these disorders. We discuss drugs identified as potential UPR-modifying therapeutic agents-in particular the licensed antidepressant trazodone-and we review epidemiological and trial data from their use in human populations. Finally, we explore future directions for investigating the potential benefit of using trazodone or similar UPR-modulating compounds for disease modification in patients with dementia.
Collapse
Affiliation(s)
- Emad Sidhom
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK; (E.S.); (A.J.B.); (H.L.S.); (G.R.M.)
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Windsor Research Unit, Fulbourn Hospital, Cambridge CB21 5EF, UK
- Gnodde Goldman Sachs Translational Neuroscience Unit, Windsor Research Unit, University of Cambridge, Cambridge CB2 1TN, UK
| | - John T. O’Brien
- Department of Psychiatry, University of Cambridge, Herchel Smith Building, Forvie Site, Cambridge CB2 0SZ, UK;
| | - Adrian J. Butcher
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK; (E.S.); (A.J.B.); (H.L.S.); (G.R.M.)
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Heather L. Smith
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK; (E.S.); (A.J.B.); (H.L.S.); (G.R.M.)
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Giovanna R. Mallucci
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK; (E.S.); (A.J.B.); (H.L.S.); (G.R.M.)
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Benjamin R. Underwood
- Cambridgeshire and Peterborough NHS Foundation Trust, Windsor Research Unit, Fulbourn Hospital, Cambridge CB21 5EF, UK
- Gnodde Goldman Sachs Translational Neuroscience Unit, Windsor Research Unit, University of Cambridge, Cambridge CB2 1TN, UK
- Department of Psychiatry, University of Cambridge, Herchel Smith Building, Forvie Site, Cambridge CB2 0SZ, UK;
| |
Collapse
|
35
|
Ma Q, Long S, Gan Z, Tettamanti G, Li K, Tian L. Transcriptional and Post-Transcriptional Regulation of Autophagy. Cells 2022; 11:cells11030441. [PMID: 35159248 PMCID: PMC8833990 DOI: 10.3390/cells11030441] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/03/2022] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a widely conserved process in eukaryotes that is involved in a series of physiological and pathological events, including development, immunity, neurodegenerative disease, and tumorigenesis. It is regulated by nutrient deprivation, energy stress, and other unfavorable conditions through multiple pathways. In general, autophagy is synergistically governed at the RNA and protein levels. The upstream transcription factors trigger or inhibit the expression of autophagy- or lysosome-related genes to facilitate or reduce autophagy. Moreover, a significant number of non-coding RNAs (microRNA, circRNA, and lncRNA) are reported to participate in autophagy regulation. Finally, post-transcriptional modifications, such as RNA methylation, play a key role in controlling autophagy occurrence. In this review, we summarize the progress on autophagy research regarding transcriptional regulation, which will provide the foundations and directions for future studies on this self-eating process.
Collapse
Affiliation(s)
- Qiuqin Ma
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Q.M.); (Z.G.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shihui Long
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China;
| | - Zhending Gan
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Q.M.); (Z.G.)
| | - Gianluca Tettamanti
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
- BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Napoli Federico II, 80138 Napoli, Italy
| | - Kang Li
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China;
- Correspondence: (K.L.); (L.T.)
| | - Ling Tian
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (Q.M.); (Z.G.)
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
- Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (K.L.); (L.T.)
| |
Collapse
|
36
|
Nakagawa K, Islam S, Ueda M, Nakagawa T. Endoplasmic reticulum stress contributes to the decline in doublecortin expression in the immature neurons of mice with long-term obesity. Sci Rep 2022; 12:1022. [PMID: 35046482 PMCID: PMC8770636 DOI: 10.1038/s41598-022-05012-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 01/03/2022] [Indexed: 12/25/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) plays an important role in hippocampus-dependent function. The number of doublecortin (Dcx)-positive immature neurons in the dentate gyrus decreases over time, especially in the early stages of Alzheimer’s disease (AD), and is further reduced in later stages of AD. Obesity in midlife is associated with dementia later in life; however, the underlying mechanisms by which obesity results in the development of dementia later in life remain unknown. Here, we show that endoplasmic reticulum (ER) stress was activated in the hippocampus and processes of Dcx-expressing immature neurons were shortened, coexpressing CHOP in APP23 AD model mice with high-fat diet-induced long-term obesity and in aged Leprdb/db (db/db) mice. Moreover, in cells differentiating from hippocampal neurospheres, Dcx mRNA was rapidly degraded via a microRNA (miRNA) pathway after thapsigargin treatment in vitro. These results indicate that loss of Dcx mRNA induced by ER stress during AHN may cause memory impairment in obese individuals later in life.
Collapse
Affiliation(s)
- Kiyomi Nakagawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Saiful Islam
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram Laboratories, Chattogram, 4220, Bangladesh
| | - Masashi Ueda
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.,Department of Mental Retardation and Birth Defect Research, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Toshiyuki Nakagawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan.
| |
Collapse
|
37
|
Reduced DNAJC3 Expression Affects Protein Translocation across the ER Membrane and Attenuates the Down-Modulating Effect of the Translocation Inhibitor Cyclotriazadisulfonamide. Int J Mol Sci 2022; 23:ijms23020584. [PMID: 35054769 PMCID: PMC8775681 DOI: 10.3390/ijms23020584] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 12/20/2022] Open
Abstract
One of the reported substrates for the endoplasmic reticulum (ER) translocation inhibitor cyclotriazadisulfonamide (CADA) is DNAJC3, a chaperone of the unfolded protein response during ER stress. In this study, we investigated the impact of altered DNAJC3 protein levels on the inhibitory activity of CADA. By comparing WT DNAJC3 with a CADA-resistant DNAJC3 mutant, we observed the enhanced sensitivity of human CD4, PTK7 and ERLEC1 for CADA when DNAJC3 was expressed at high levels. Combined treatment of CADA with a proteasome inhibitor resulted in synergistic inhibition of protein translocation and in the rescue of a small preprotein fraction, which presumably corresponds to the CADA affected protein fraction that is stalled at the Sec61 translocon. We demonstrate that DNAJC3 enhances the protein translation of a reporter protein that is expressed downstream of the CADA-stalled substrate, suggesting that DNAJC3 promotes the clearance of the clogged translocon. We propose a model in which a reduced DNAJC3 level by CADA slows down the clearance of CADA-stalled substrates. This results in higher residual translocation into the ER lumen due to the longer dwelling time of the temporarily stalled substrates in the translocon. Thus, by directly reducing DNAJC3 protein levels, CADA attenuates its net down-modulating effect on its substrates.
Collapse
|
38
|
Korneeva NL. Integrated Stress Response in Neuronal Pathology and in Health. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:S111-S127. [PMID: 35501991 DOI: 10.1134/s0006297922140103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 06/14/2023]
Abstract
Neurodegeneration involves progressive pathological loss of a specific population of neurons, glial activation, and dysfunction of myelinating oligodendrocytes leading to cognitive impairment and altered movement, breathing, and senses. Neuronal degeneration is a hallmark of aging, stroke, drug abuse, toxic chemical exposure, viral infection, chronic inflammation, and a variety of neurological diseases. Accumulation of intra- and extracellular protein aggregates is a common characteristic of cell pathologies. Excessive production of reactive oxygen species and nitric oxide, induction of endoplasmic reticulum stress, and accumulation of misfolded protein aggregates have been shown to trigger a defensive mechanism called integrated stress response (ISR). Activation of ISR is important for synaptic plasticity in learning and memory formation. However, sustaining of ISR may lead to the development of neuronal pathologies and altered patterns in behavior and perception.
Collapse
Affiliation(s)
- Nadejda L Korneeva
- Louisiana State University Health Science Center, Shreveport, LA 71103, USA.
| |
Collapse
|
39
|
Passeri G, Northcote-Smith J, Suntharalingam K. Delivery of an immunogenic cell death-inducing copper complex to cancer stem cells using polymeric nanoparticles. RSC Adv 2022; 12:5290-5299. [PMID: 35425564 PMCID: PMC8981415 DOI: 10.1039/d1ra08788f] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/28/2022] [Indexed: 01/04/2023] Open
Abstract
The major cause for cancer related deaths worldwide is tumour relapse and metastasis, both of which have been heavily linked to the existence of cancer stem cells (CSCs). CSCs are able to escape current treatment regimens, reform tumours, and promote their spread to secondary sites. Recently, our research group reported the first metal-based agent 1 (a copper(ii) compound ligated by a bidentate 4,7-diphenyl-1,10-phenanthroline and a tridentate Schiff base ligand) to potently kill CSCs via cytotoxic and immunogenic mechanisms. Here we show that encapsulation of 1 by polymeric nanoparticles at the appropriate feed (10%, 1 NP10) enhances CSC uptake and improves potency towards bulk cancer cells and CSCs (grown in monolayer and three-dimensional cultures). The nanoparticle formulation triggers a similar cellular response to the payload, which bodes well for further translation. Specifically, the nanoparticle formulation elevates intracellular reactive oxygen species levels, induces ER stress, and evokes damage-associated molecular patterns consistent with immunogenic cell death. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver immunogenic metal complexes into CSCs. In this study we deliver an immunogenic cell death-inducing copper(ii) complex, comprising of 4,7-diphenyl-1,10-phenanthroline and a Schiff base ligand, to breast cancer stem cells.![]()
Collapse
Affiliation(s)
- Ginevra Passeri
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK
| | | | | |
Collapse
|
40
|
Bahamondes Lorca VA, Bastidas Mayorga BD, Tong L, Wu S. UVB-induced eIF2α phosphorylation in keratinocytes depends on decreased ATF4, GADD34 and CReP expression levels. Life Sci 2021; 286:120044. [PMID: 34637792 DOI: 10.1016/j.lfs.2021.120044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 02/05/2023]
Abstract
AIM To elucidate the mechanism behind the sustained high levels of phosphorylated eIF2α in HaCaT cells post-UVB. MAIN METHODS In this study, expression levels of the machinery involved in the dephosphorylation of eIF2α (GADD34, CReP and PP1), as well as the PERK-eIF2α-ATF4-CHOP, IRE1α/XBP1s and ATF6α signaling cascades, were analyzed by western blot and fluorescence microscope. KEY FINDINGS Our data showed that UVB induces the phosphorylation of eIF2α, which induces the translation of ATF4 and consequently the expression of CHOP and GADD34. Nevertheless, UVB also suppresses the translation of ATF4 and GADD34 in HaCaT cells via a p-eIF2α independent mechanism. Therefore, the lack of ATF4, GADD34 and CReP is responsible for the sustained phosphorylation of eIF2α. Finally, our data also showed that UVB selectively modifies PERK and downregulates ATF6α expression but does not induce activation of the IRE1α/XBP1s pathway in HaCaT cells. SIGNIFICANCE Novel mechanism to explain the prolonged phosphorylation of eIF2α post-UVB irradiation.
Collapse
Affiliation(s)
- Verónica A Bahamondes Lorca
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA; Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Bernardo D Bastidas Mayorga
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA
| | - Lingying Tong
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.
| | - Shiyong Wu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
41
|
Mutation in FBXO32 causes dilated cardiomyopathy through up-regulation of ER-stress mediated apoptosis. Commun Biol 2021; 4:884. [PMID: 34272480 PMCID: PMC8285540 DOI: 10.1038/s42003-021-02391-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum (ER) stress induction of cell death is implicated in cardiovascular diseases. Sustained activation of ER-stress induces the unfolded protein response (UPR) pathways, which in turn activate three major effector proteins. We previously reported a missense homozygous mutation in FBXO32 (MAFbx, Atrogin-1) causing advanced heart failure by impairing autophagy. In the present study, we performed transcriptional profiling and biochemical assays, which unexpectedly revealed a reduced activation of UPR effectors in patient mutant hearts, while a strong up-regulation of the CHOP transcription factor and of its target genes are observed. Expression of mutant FBXO32 in cells is sufficient to induce CHOP-associated apoptosis, to increase the ATF2 transcription factor and to impair ATF2 ubiquitination. ATF2 protein interacts with FBXO32 in the human heart and its expression is especially high in FBXO32 mutant hearts. These findings provide a new underlying mechanism for FBXO32-mediated cardiomyopathy, implicating abnormal activation of CHOP. These results suggest alternative non-canonical pathways of CHOP activation that could be considered to develop new therapeutic targets for the treatment of FBXO32-associated DCM. Al-Yacoub et al. investigate the consequences of FBXO32 mutation on dilated cardiomyopathy. ER stress, abnormal CHOP activation and CHOP-induced apoptosis with no UPR effector activation are found to underlie the FBXO32 mutation induced cardiomyopathy, suggesting an alternative pathway that can be considered to develop new therapeutic targets for its treatment.
Collapse
|
42
|
Zhu L, Huang X, Li Z, Cao G, Zhu X, She S, Huang T, Lu G. Evaluation of hepatotoxicity induced by 2-ethylhexyldiphenyl phosphate based on transcriptomics and its potential metabolism pathway in human hepatocytes. JOURNAL OF HAZARDOUS MATERIALS 2021; 413:125281. [PMID: 33582465 DOI: 10.1016/j.jhazmat.2021.125281] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/18/2021] [Accepted: 01/28/2021] [Indexed: 06/12/2023]
Abstract
Increasing use of organophosphorus flame retardants (OPFRs) has aroused great concern to their uncertain environment risk, especially to human health risk. In our study, hepatotoxicity screening of six aryl-OPFRs, potential hepatotoxicity mechanism of 2-ethylhexyldiphenyl phosphate (EHDPP) using RNA-sequencing and its metabolites were investigated in human hepatocytes (L02). The toxicity results demonstrated that EHDPP should be prioritized for further research with the highest toxicity. Further RNA-seq results through GO and KEGG enrichment analysis indicated that exposure to 10 mg/L of EHDPP significantly affected energy homeostasis, endoplasmic reticulum (ER) stress, apoptosis, cell cycle, and inflammation response in cells. The top 12 hub genes were validated by RT-qPCR and conformed to be mainly related to glycolysis and ER stress, followed by cell cycle and inflammation response. Western blot, apoptosis detection, glycolysis stress test, and cell cycle analysis were further performed to verify the above main pathways. Additionally, it was found in the metabolism experiment that detoxification of EHDPP by phase I and phase II metabolism in cells wasn't significant until 48 h with a metabolic rate of 6.12%. EHDPP was stable and still dominated the induction of toxicity. Overall, this study provided valuable information regarding the toxicity and potential metabolism pathway of EHDPP.
Collapse
Affiliation(s)
- Lingfei Zhu
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Xiaohan Huang
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Zhenhua Li
- The First Affiliated Hospital, Biomedical Translational Research Institute and School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Gang Cao
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Xuanjin Zhu
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Shaohua She
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Tenghao Huang
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China
| | - Gang Lu
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
43
|
Misra J, Holmes MJ, T Mirek E, Langevin M, Kim HG, Carlson KR, Watford M, Dong XC, Anthony TG, Wek RC. Discordant regulation of eIF2 kinase GCN2 and mTORC1 during nutrient stress. Nucleic Acids Res 2021; 49:5726-5742. [PMID: 34023907 PMCID: PMC8191763 DOI: 10.1093/nar/gkab362] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 11/13/2022] Open
Abstract
Appropriate regulation of the Integrated stress response (ISR) and mTORC1 signaling are central for cell adaptation to starvation for amino acids. Halofuginone (HF) is a potent inhibitor of aminoacylation of tRNAPro with broad biomedical applications. Here, we show that in addition to translational control directed by activation of the ISR by general control nonderepressible 2 (GCN2), HF increased free amino acids and directed translation of genes involved in protein biogenesis via sustained mTORC1 signaling. Deletion of GCN2 reduced cell survival to HF whereas pharmacological inhibition of mTORC1 afforded protection. HF treatment of mice synchronously activated the GCN2-mediated ISR and mTORC1 in liver whereas Gcn2-null mice allowed greater mTORC1 activation to HF, resulting in liver steatosis and cell death. We conclude that HF causes an amino acid imbalance that uniquely activates both GCN2 and mTORC1. Loss of GCN2 during HF creates a disconnect between metabolic state and need, triggering proteostasis collapse.
Collapse
Affiliation(s)
- Jagannath Misra
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Michael J Holmes
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Emily T Mirek
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901 USA
| | - Michael Langevin
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901 USA
| | - Hyeong-Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Kenneth R Carlson
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Malcolm Watford
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901 USA
| | - X Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA.,Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Tracy G Anthony
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ 08901 USA
| | - Ronald C Wek
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
44
|
Activation of the Integrated Stress Response and ER Stress Protect from Fluorizoline-Induced Apoptosis in HEK293T and U2OS Cell Lines. Int J Mol Sci 2021; 22:ijms22116117. [PMID: 34204139 PMCID: PMC8201103 DOI: 10.3390/ijms22116117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
The prohibitin (PHB)-binding compound fluorizoline as well as PHB-downregulation activate the integrated stress response (ISR) in HEK293T and U2OS human cell lines. This activation is denoted by phosphorylation of eIF2α and increases in ATF4, ATF3, and CHOP protein levels. The blockage of the activation of the ISR by overexpression of GRP78, as well as an increase in IRE1 activity, indicate the presence of ER stress after fluorizoline treatment. The inhibition of the ER stress response in HEK293T and U2OS led to increased sensitivity to fluorizoline-induced apoptosis, indicating a pro-survival role of this pathway after fluorizoline treatment in these cell lines. Fluorizoline induced an increase in calcium concentration in the cytosol and the mitochondria. Finally, two different calcium chelators reduced fluorizoline-induced apoptosis in U2OS cells. Thus, we have found that fluorizoline causes increased ER stress and activation of the integrated stress response, which in HEK293T and U2OS cells are protective against fluorizoline-induced apoptosis.
Collapse
|
45
|
Demmings MD, Tennyson EC, Petroff GN, Tarnowski-Garner HE, Cregan SP. Activating transcription factor-4 promotes neuronal death induced by Parkinson's disease neurotoxins and α-synuclein aggregates. Cell Death Differ 2021; 28:1627-1643. [PMID: 33277577 PMCID: PMC8167173 DOI: 10.1038/s41418-020-00688-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/26/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra resulting in severe and progressive motor impairments. However, the mechanisms underlying this neuronal loss remain largely unknown. Oxidative stress and ER stress have been implicated in PD and these factors are known to activate the integrated stress response (ISR). Activating transcription factor 4 (ATF4), a key mediator of the ISR, and has been reported to induce the expression of genes involved in cellular homeostasis. However, during prolonged activation ATF4 can also induce the expression of pro-death target genes. Therefore, in the present study, we investigated the role of ATF4 in neuronal cell death in models of PD. We demonstrate that PD neurotoxins (MPP+ and 6-OHDA) and α-synuclein aggregation induced by pre-formed human alpha-synuclein fibrils (PFFs) cause sustained upregulation of ATF4 expression in mouse cortical and mesencephalic dopaminergic neurons. Furthermore, we demonstrate that PD neurotoxins induce the expression of the pro-apoptotic factors Chop, Trb3, and Puma in dopaminergic neurons in an ATF4-dependent manner. Importantly, we have determined that PD neurotoxin and α-synuclein PFF induced neuronal death is attenuated in ATF4-deficient dopaminergic neurons. Furthermore, ectopic expression of ATF4 but not transcriptionally defective ATF4ΔRK restores sensitivity of ATF4-deficient neurons to PD neurotoxins. Finally, we demonstrate that the eIF2α kinase inhibitor C16 suppresses MPP+ and 6-OHDA induced ATF4 activation and protects against PD neurotoxin induced dopaminergic neuronal death. Taken together these results indicate that ATF4 promotes dopaminergic cell death induced by PD neurotoxins and pathogenic α-synuclein aggregates and highlight the ISR factor ATF4 as a potential therapeutic target in PD.
Collapse
Affiliation(s)
- Matthew D Demmings
- Neuroscience Program, University of Western Ontario, London, ON, Canada
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- University of Western Ontario, London, ON, Canada
| | - Elizabeth C Tennyson
- Neuroscience Program, University of Western Ontario, London, ON, Canada
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- University of Western Ontario, London, ON, Canada
| | - Gillian N Petroff
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- University of Western Ontario, London, ON, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Heather E Tarnowski-Garner
- Robarts Research Institute, University of Western Ontario, London, ON, Canada
- University of Western Ontario, London, ON, Canada
| | - Sean P Cregan
- Neuroscience Program, University of Western Ontario, London, ON, Canada.
- Robarts Research Institute, University of Western Ontario, London, ON, Canada.
- University of Western Ontario, London, ON, Canada.
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
46
|
Obert DP, Wolpert AK, Grimm NL, Korff S. ER stress preconditioning ameliorates liver damage after hemorrhagic shock and reperfusion. Exp Ther Med 2021; 21:248. [PMID: 33603856 PMCID: PMC7851603 DOI: 10.3892/etm.2021.9679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022] Open
Abstract
The mismatch of oxygen supply and demand during hemorrhagic shock disturbs endoplasmic reticulum (ER) homeostasis. The resulting accumulation of unfolded proteins in the ER lumen, which is a condition that is defined as ER stress, triggers the unfolded protein response (UPR). Since the UPR influences the extent of organ damage following hemorrhagic shock/reperfusion (HS/R) and mediates the protective effects of stress preconditioning before ischemia-reperfusion injury, the current study investigated the mechanisms of ER stress preconditioning and its impact on post-hemorrhagic liver damage. Male C56BL/6-mice were injected intraperitoneally with the ER stress inductor tunicamycin (TM) or its drug vehicle 48 h prior to being subjected to a 90 min pressure-controlled hemorrhagic shock (30±5 mmHg). A period of 14 h after hemorrhagic shock induction, mice were sacrificed. Hepatocellular damage was quantified by analyzing hepatic transaminases and hematoxylin-eosin stained liver tissue sections. Additionally, the topographic expression patterns of the ER stress marker binding immunoglobulin protein (BiP), UPR signaling pathways, and the autophagy marker Beclin1 were evaluated. TM injection significantly increased BiP expression and modified the topographic expression patterns of the UPR signaling proteins. In addition, immunohistochemical analysis of Beclin1 revealed an increased pericentral staining intensity following TM pretreatment. The histologic analysis of hepatocellular damage demonstrated a significant reduction in cell death areas in HS/R+TM (P=0.024). ER stress preconditioning influences the UPR and alleviates post-hemorrhagic liver damage. The beneficial effects were, at least partially, mediated by the upregulation of BiP and autophagy induction. These results underscore the importance of the UPR in the context of HS/R and may help identify novel therapeutic targets.
Collapse
Affiliation(s)
- David Peter Obert
- Department of Anesthesiology and Intensive Care, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Department of Trauma Surgery, University of Heidelberg, 69118 Heidelberg, Germany
| | - Alexander Karl Wolpert
- Department of Trauma Surgery, University of Heidelberg, 69118 Heidelberg, Germany
- Department of Trauma Surgery, Paracelsus Medical University, 90471 Nuremberg, Germany
| | - Nathan Lewis Grimm
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27708, USA
| | - Sebastian Korff
- Department of Trauma Surgery, University of Heidelberg, 69118 Heidelberg, Germany
- Department of Orthopaedic Surgery, Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
47
|
Pang HF, Li XX, Zhao YH, Kang JK, Li JY, Tian W, Wang CM, Hou HX, Li DR. Confirming whether novel rhein derivative 4a induces paraptosis-like cell death by endoplasmic reticulum stress in ovarian cancer cells. Eur J Pharmacol 2020; 886:173526. [PMID: 32890460 DOI: 10.1016/j.ejphar.2020.173526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022]
Abstract
Ovarian cancer is the leading cause of death among gynecologic cancer patients. Although platinum-based chemotherapy as a frontline treatment for ovarian cancer has been widely used in clinical settings, its clinical efficacy is not satisfactory due to the resistance of ovarian cancer cells to apoptosis. Therefore, it is of great significance to induce non-apoptotic programed cell death patterns, such as paraptosis, in ovarian cancer. In this study, we aimed to explore the potential anticancer mechanisms of novel rhein derivative 4a, which was modified with rhein as a lead compound. The results showed that a wide range of vacuoles from the endoplasmic reticulum and mitochondria appeared in ovarian SKOV3, SKOV3-PM4, and A2780 cells treated with derivative 4a, and the cell death caused by derivative 4a is a type of non-apoptotic and non-autophagic death, which is caused by expansion and damage of the endoplasmic reticulum or mitochondria, showing the characteristics of para-apoptotic death. Furthermore, derivative 4a stimulated the unfolded protein reaction of ovarian cancer cells by upregulating the expression of Bip78 and activating the PERK-eIF2α-ATF4 pathways. Notably, rhein derivative 4a-induced cell death was positively correlated with activation of p38, ERK, and JNK, and negatively correlated with Alix, a known protein that inhibits paraptosis. In addition, derivative 4a treatment also induced G2/M phase arrest in ovarian cancer cells. Taken together, our study reveals that derivative 4a induces paraptosis, and this finding can serve as a basis in developing a new strategy for the treatment of antiapoptotic ovarian cancer.
Collapse
Affiliation(s)
- Hui-Feng Pang
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021 China
| | - Xin-Xiao Li
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021 China
| | - Yu-Hua Zhao
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021 China
| | - Jian-Kang Kang
- Life Sciences Institute, Guangxi Medical University, Nanning 530021, China
| | - Jun-Ying Li
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Wei Tian
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Chun-Miao Wang
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
| | - Hua-Xin Hou
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| | - Dan-Rong Li
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021 China.
| |
Collapse
|
48
|
Boyle ST, Poltavets V, Kular J, Pyne NT, Sandow JJ, Lewis AC, Murphy KJ, Kolesnikoff N, Moretti PAB, Tea MN, Tergaonkar V, Timpson P, Pitson SM, Webb AI, Whitfield RJ, Lopez AF, Kochetkova M, Samuel MS. ROCK-mediated selective activation of PERK signalling causes fibroblast reprogramming and tumour progression through a CRELD2-dependent mechanism. Nat Cell Biol 2020; 22:882-895. [PMID: 32451439 DOI: 10.1038/s41556-020-0523-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/17/2020] [Indexed: 01/05/2023]
Abstract
It is well accepted that cancers co-opt the microenvironment for their growth. However, the molecular mechanisms that underlie cancer-microenvironment interactions are still poorly defined. Here, we show that Rho-associated kinase (ROCK) in the mammary tumour epithelium selectively actuates protein-kinase-R-like endoplasmic reticulum kinase (PERK), causing the recruitment and persistent education of tumour-promoting cancer-associated fibroblasts (CAFs), which are part of the cancer microenvironment. An analysis of tumours from patients and mice reveals that cysteine-rich with EGF-like domains 2 (CRELD2) is the paracrine factor that underlies PERK-mediated CAF education downstream of ROCK. We find that CRELD2 is regulated by PERK-regulated ATF4, and depleting CRELD2 suppressed tumour progression, demonstrating that the paracrine ROCK-PERK-ATF4-CRELD2 axis promotes the progression of breast cancer, with implications for cancer therapy.
Collapse
Affiliation(s)
- Sarah Theresa Boyle
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Valentina Poltavets
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Jasreen Kular
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Natasha Theresa Pyne
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Jarrod John Sandow
- Division of Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Alexander Charles Lewis
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia.,Translational Haematology Program, Peter McCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kendelle Joan Murphy
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, University of NSW, Sydney, New South Wales, Australia
| | - Natasha Kolesnikoff
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | | | - Melinda Nay Tea
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - Vinay Tergaonkar
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia.,Institute of Molecular and Cell Biology, A*STAR and Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Paul Timpson
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, University of NSW, Sydney, New South Wales, Australia
| | - Stuart Maxwell Pitson
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Andrew Ian Webb
- Division of Systems Biology and Personalised Medicine, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Robert John Whitfield
- Breast, Endocrine and Surgical Oncology Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Angel Francisco Lopez
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Marina Kochetkova
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia.
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia. .,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
49
|
Dash S, Aydin Y, Wu T. Integrated stress response in hepatitis C promotes Nrf2-related chaperone-mediated autophagy: A novel mechanism for host-microbe survival and HCC development in liver cirrhosis. Semin Cell Dev Biol 2020; 101:20-35. [PMID: 31386899 PMCID: PMC7007355 DOI: 10.1016/j.semcdb.2019.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/26/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023]
Abstract
The molecular mechanism(s) how liver damage during the chronic hepatitis C virus (HCV) infection evolve into cirrhosis and hepatocellular carcinoma (HCC) is unclear. HCV infects hepatocyte, the major cell types in the liver. During infection, large amounts of viral proteins and RNA replication intermediates accumulate in the endoplasmic reticulum (ER) of the infected hepatocyte, which creates a substantial amount of stress response. Infected hepatocyte activates a different type of stress adaptive mechanisms such as unfolded protein response (UPR), antioxidant response (AR), and the integrated stress response (ISR) to promote virus-host cell survival. The hepatic stress is also amplified by another layer of innate and inflammatory response associated with cellular sensing of virus infection through the production of interferon (IFN) and inflammatory cytokines. The interplay between various types of cellular stress signal leads to different forms of cell death such as apoptosis, necrosis, and autophagy depending on the intensity of the stress and nature of the adaptive cellular response. How do the adaptive cellular responses decode such death programs that promote host-microbe survival leading to the establishment of chronic liver disease? In this review, we discuss how the adaptive cellular response through the Nrf2 pathway that promotes virus and cell survival. Furthermore, we provide a glimpse of novel stress-induced Nrf2 mediated compensatory autophagy mechanisms in virus-cell survival that degrade tumor suppressor gene and activation of oncogenic signaling during HCV infection. Based on these facts, we hypothesize that the balance between hepatic stress, inflammation and different types of cell death determines liver disease progression outcomes. We propose that a more nuanced understanding of virus-host interactions under excessive cellular stress may provide an answer to the fundamental questions why some individuals with chronic HCV infection remain at risk of developing cirrhosis, cancer and some do not.
Collapse
Affiliation(s)
- Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA.
| | - Yucel Aydin
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA
| |
Collapse
|
50
|
Ebert SM, Bullard SA, Basisty N, Marcotte GR, Skopec ZP, Dierdorff JM, Al-Zougbi A, Tomcheck KC, DeLau AD, Rathmacher JA, Bodine SC, Schilling B, Adams CM. Activating transcription factor 4 (ATF4) promotes skeletal muscle atrophy by forming a heterodimer with the transcriptional regulator C/EBPβ. J Biol Chem 2020; 295:2787-2803. [PMID: 31953319 PMCID: PMC7049960 DOI: 10.1074/jbc.ra119.012095] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/10/2020] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle atrophy is a highly-prevalent and debilitating condition that remains poorly understood at the molecular level. Previous work found that aging, fasting, and immobilization promote skeletal muscle atrophy via expression of activating transcription factor 4 (ATF4) in skeletal muscle fibers. However, the direct biochemical mechanism by which ATF4 promotes muscle atrophy is unknown. ATF4 is a member of the basic leucine zipper transcription factor (bZIP) superfamily. Because bZIP transcription factors are obligate dimers, and because ATF4 is unable to form highly-stable homodimers, we hypothesized that ATF4 may promote muscle atrophy by forming a heterodimer with another bZIP family member. To test this hypothesis, we biochemically isolated skeletal muscle proteins that associate with the dimerization- and DNA-binding domain of ATF4 (the bZIP domain) in mouse skeletal muscle fibers in vivo Interestingly, we found that ATF4 forms at least five distinct heterodimeric bZIP transcription factors in skeletal muscle fibers. Furthermore, one of these heterodimers, composed of ATF4 and CCAAT enhancer-binding protein β (C/EBPβ), mediates muscle atrophy. Within skeletal muscle fibers, the ATF4-C/EBPβ heterodimer interacts with a previously unrecognized and evolutionarily conserved ATF-C/EBP composite site in exon 4 of the Gadd45a gene. This three-way interaction between ATF4, C/EBPβ, and the ATF-C/EBP composite site activates the Gadd45a gene, which encodes a critical mediator of muscle atrophy. Together, these results identify a biochemical mechanism by which ATF4 induces skeletal muscle atrophy, providing molecular-level insights into the etiology of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Scott M Ebert
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246; Emmyon, Inc., Coralville, Iowa 52241
| | - Steven A Bullard
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Nathan Basisty
- Buck Institute for Research on Aging, Novato, California 94945
| | - George R Marcotte
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Zachary P Skopec
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Jason M Dierdorff
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Asma Al-Zougbi
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Kristin C Tomcheck
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Austin D DeLau
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Jacob A Rathmacher
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Sue C Bodine
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Emmyon, Inc., Coralville, Iowa 52241
| | | | - Christopher M Adams
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa 52242; Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246; Emmyon, Inc., Coralville, Iowa 52241.
| |
Collapse
|