101
|
Mann MJ, Melendez-Suchi C, Sukhoplyasova M, Flory AR, Carson Irvine M, Iyer AR, Vorndran H, Guerriero CJ, Brodsky JL, Hendershot LM, Buck TM. Loss of Grp170 results in catastrophic disruption of endoplasmic reticulum functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563191. [PMID: 37905119 PMCID: PMC10614942 DOI: 10.1101/2023.10.19.563191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
GRP170, a product of the Hyou1 gene, is required for mouse embryonic development, and its ablation in kidney nephrons leads to renal failure. Unlike most chaperones, GRP170 is the lone member of its chaperone family in the ER lumen. However, the cellular requirement for GRP170, which both binds non-native proteins and acts as nucleotide exchange factor for BiP, is poorly understood. Here, we report on the isolation of embryonic fibroblasts from mice in which LoxP sites were engineered in the Hyou1 loci ( Hyou1 LoxP/LoxP ). A doxycycline-regulated Cre recombinase was also stably introduced into these cells. Induction of Cre resulted in excision of Hyou1 and depletion of Grp170 protein, culminating in apoptotic cell death. As Grp170 levels fell we observed increased steady-state binding of BiP to a client, slowed degradation of a misfolded BiP substrate, and BiP accumulation in NP40-insoluble fractions. Consistent with disrupted BiP functions, we observed reactivation of BiP storage pools and induction of the unfolded protein response (UPR) in futile attempts to provide compensatory increases in ER chaperones and folding enzymes. Together, these results provide insights into the cellular consequences of controlled Grp170 loss and insights into mutations in the Hyou1 locus and human disease.
Collapse
|
102
|
Webber CJ, Murphy CN, Rondón-Ortiz AN, van der Spek SJF, Kelly EX, Lampl NM, Chiesa G, Khalil AS, Emili A, Wolozin B. Human herpesvirus 8 ORF57 protein is able to reduce TDP-43 pathology: network analysis identifies interacting pathways. Hum Mol Genet 2023; 32:2966-2980. [PMID: 37522762 PMCID: PMC10549787 DOI: 10.1093/hmg/ddad122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023] Open
Abstract
Aggregation of TAR DNA-binding protein 43 kDa (TDP-43) is thought to drive the pathophysiology of amyotrophic lateral sclerosis and some frontotemporal dementias. TDP-43 is normally a nuclear protein that in neurons translocates to the cytoplasm and can form insoluble aggregates upon activation of the integrated stress response (ISR). Viruses evolved to control the ISR. In the case of Herpesvirus 8, the protein ORF57 acts to bind protein kinase R, inhibit phosphorylation of eIF2α and reduce activation of the ISR. We hypothesized that ORF57 might also possess the ability to inhibit aggregation of TDP-43. ORF57 was expressed in the neuronal SH-SY5Y line and its effects on TDP-43 aggregation characterized. We report that ORF57 inhibits TDP-43 aggregation by 55% and elicits a 2.45-fold increase in the rate of dispersion of existing TDP-43 granules. These changes were associated with a 50% decrease in cell death. Proteomic studies were carried out to identify the protein interaction network of ORF57. We observed that ORF57 directly binds to TDP-43 as well as interacts with many components of the ISR, including elements of the proteostasis machinery known to reduce TDP-43 aggregation. We propose that viral proteins designed to inhibit a chronic ISR can be engineered to remove aggregated proteins and dampen a chronic ISR.
Collapse
Affiliation(s)
- Chelsea J Webber
- Departments of Pharmacology, Physiology and Biophysics, Boston University, Boston, MA 02215, USA
| | - Caroline N Murphy
- Departments of Pharmacology, Physiology and Biophysics, Boston University, Boston, MA 02215, USA
| | - Alejandro N Rondón-Ortiz
- Departments of Pharmacology, Physiology and Biophysics, Boston University, Boston, MA 02215, USA
- Center for Network Systems Biology, Boston University, Boston, MA 02215, USA
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Sophie J F van der Spek
- Departments of Pharmacology, Physiology and Biophysics, Boston University, Boston, MA 02215, USA
| | - Elena X Kelly
- Departments of Pharmacology, Physiology and Biophysics, Boston University, Boston, MA 02215, USA
| | - Noah M Lampl
- Center for Network Systems Biology, Boston University, Boston, MA 02215, USA
| | - Giulio Chiesa
- Biological Design Center, Boston University, Boston, MA 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Ahmad S Khalil
- Biological Design Center, Boston University, Boston, MA 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University, Boston, MA 02215, USA
- Department of Biochemistry, Boston University, Boston, MA 02115, USA
- Department of Biochemistry, Oregon Health Sciences University, Portland, OR 97239, USA
| | - Benjamin Wolozin
- Departments of Pharmacology, Physiology and Biophysics, Boston University, Boston, MA 02215, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02115, USA
- Center for Neurophotonics, Boston University, Boston, MA 02115, USA
- Department of Neurology, Boston University, Boston, MA 02115, USA
| |
Collapse
|
103
|
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
|
104
|
Silva Barcelos EC, Rompietti C, Adamo FM, Dorillo E, De Falco F, Del Papa B, Baldoni S, Nogarotto M, Esposito A, Capoccia S, Geraci C, Sorcini D, Stella A, Arcaleni R, Tini V, Imbroisi Valle Errera F, Rosati E, Sportoletti P. NOTCH1-mutated chronic lymphocytic leukemia displays high endoplasmic reticulum stress response with druggable potential. Front Oncol 2023; 13:1218989. [PMID: 37817771 PMCID: PMC10561002 DOI: 10.3389/fonc.2023.1218989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction Constitutive activation of NOTCH1-wild-type (NT1-WT) signaling is associated with poor outcomes in chronic lymphocytic leukemia (CLL), and NOTCH1 mutation (c.7541_7542delCT), which potentiates NOTCH1 signaling, worsens the prognosis. However, the specific mechanisms of NOTCH1 deregulation are still poorly understood. Accumulative evidence mentioned endoplasmic reticulum (ER) stress/unfolded protein response (UPR) as a key targetable pathway in CLL. In this study, we investigated the impact of NOTCH1 deregulation on CLL cell response to ER stress induction, with the aim of identifying new therapeutic opportunities for CLL. Methods We performed a bioinformatics analysis of NOTCH1-mutated (NT1-M) and NT1-WT CLL to identify differentially expressed genes (DEGs) using the rank product test. Quantitative real-time polymerase chain reaction (qPCR), Western blotting, cytosolic Ca2+, and annexin V/propidium iodide (PI) assay were used to detect curcumin ER stress induction effects. A median-effect equation was used for drug combination tests. The experimental mouse model Eμ-TCL1 was used to evaluate the impact of ER stress exacerbation by curcumin treatment on the progression of leukemic cells and NOTCH1 signaling. Results and discussion Bioinformatics analysis revealed gene enrichment of the components of the ER stress/UPR pathway in NT1-M compared to those in NT1-WT CLL. Ectopic expression of NOTCH1 mutation upregulated the levels of ER stress response markers in the PGA1 CLL cell line. Primary NT1-M CLL was more sensitive to curcumin as documented by a significant perturbation in Ca2+ homeostasis and higher expression of ER stress/UPR markers compared to NT1-WT cells. It was also accompanied by a significantly higher apoptotic response mediated by C/EBP homologous protein (CHOP) expression, caspase 4 cleavage, and downregulation of NOTCH1 signaling in NT1-M CLL cells. Curcumin potentiated the apoptotic effect of venetoclax in NT1-M CLL cells. In Eμ-TCL1 leukemic mice, the administration of curcumin activated ER stress in splenic B cells ex vivo and significantly reduced the percentage of CD19+/CD5+ cells infiltrating the spleen, liver, and bone marrow (BM). These cellular effects were associated with reduced NOTCH1 activity in leukemic cells and resulted in prolonged survival of curcumin-treated mice. Overall, our results indicate that ER stress induction in NT1-M CLL might represent a new therapeutic opportunity for these high-risk CLL patients and improve the therapeutic effect of drugs currently used in CLL.
Collapse
Affiliation(s)
- Estevão Carlos Silva Barcelos
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo, Vitória, Brazil
| | - Chiara Rompietti
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Francesco Maria Adamo
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Erica Dorillo
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Filomena De Falco
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Beatrice Del Papa
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Stefano Baldoni
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Manuel Nogarotto
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Angela Esposito
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Silvia Capoccia
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Clelia Geraci
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Daniele Sorcini
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Arianna Stella
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Roberta Arcaleni
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Valentina Tini
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | | | - Emanuela Rosati
- Department of Medicine and Surgery, Biosciences and Medical Embryology Section, University of Perugia, Perugia, Italy
| | - Paolo Sportoletti
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| |
Collapse
|
105
|
Majerciak V, Zhou T, Kruhlak M, Zheng ZM. RNA helicase DDX6 and scaffold protein GW182 in P-bodies promote biogenesis of stress granules. Nucleic Acids Res 2023; 51:9337-9355. [PMID: 37427791 PMCID: PMC10516652 DOI: 10.1093/nar/gkad585] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 06/05/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023] Open
Abstract
Two prominent cytoplasmic RNA granules, ubiquitous RNA-processing bodies (PB) and inducible stress granules (SG), regulate mRNA translation and are intimately related. In this study, we found that arsenite (ARS)-induced SG formed in a stepwise process is topologically and mechanically linked to PB. Two essential PB components, GW182 and DDX6, are repurposed under stress to play direct but distinguishable roles in SG biogenesis. By providing scaffolding activities, GW182 promotes the aggregation of SG components to form SG bodies. DEAD-box helicase DDX6 is also essential for the proper assembly and separation of PB from SG. DDX6 deficiency results in the formation of irregularly shaped 'hybrid' PB/SG granules with accumulated components of both PB and SG. Wild-type DDX6, but not its helicase mutant E247A, can rescue the separation of PB from SG in DDX6KO cells, indicating a requirement of DDX6 helicase activity for this process. DDX6 activity in biogenesis of both PB and SG in the cells under stress is further modulated by its interaction with two protein partners, CNOT1 and 4E-T, of which knockdown affects the formation of both PB and also SG. Together, these data highlight a new functional paradigm between PB and SG biogenesis during the stress.
Collapse
Affiliation(s)
- Vladimir Majerciak
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Tongqing Zhou
- Structural Biology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J Kruhlak
- CCR Confocal Microscopy Core Facility, Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| |
Collapse
|
106
|
Pioltine EM, Costa CB, Franchi FF, dos Santos PH, Nogueira MFG. Tauroursodeoxycholic Acid Supplementation in In Vitro Culture of Indicine Bovine Embryos: Molecular and Cellular Effects on the In Vitro Cryotolerance. Int J Mol Sci 2023; 24:14060. [PMID: 37762363 PMCID: PMC10531190 DOI: 10.3390/ijms241814060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
During embryo development, the endoplasmic reticulum (ER) acts as an important site for protein biosynthesis; however, in vitro culture (IVC) can negatively affect ER homeostasis. Therefore, the aim of our study was to evaluate the effects of the supplementation of tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor, in the IVC of bovine embryos. Two experiments were carried out: Exp. 1: an evaluation of blastocyst rate, hatching kinetics, and gene expression of hatched embryos after being treated with different concentrations of TUDCA (50, 200, or 1000 μM) in the IVC; Exp. 2: an evaluation of the re-expansion, hatching, and gene expression of hatched embryos previously treated with 200 µM of TUDCA at IVC and submitted to vitrification. There was no increase in the blastocyst and hatched blastocyst rates treated with TUDCA in the IVC. However, embryos submitted to vitrification after treatment with 200 µM of TUDCA underwent an increased hatching rate post-warming together with a down-regulation in the expression of ER stress-related genes and the accumulation of lipids. In conclusion, this work showed that the addition of TUDCA during in vitro culture can improve the cryotolerance of the bovine blastocyst through the putative modulation of ER and oxidative stress.
Collapse
Affiliation(s)
- Elisa Mariano Pioltine
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
| | - Camila Bortoliero Costa
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
- Laboratory of Embryonic Micromanipulation, Department of Biological Sciences, School of Sciences and Languages, São Paulo State University (UNESP), Assis 19806-900, Brazil
| | - Fernanda Fagali Franchi
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
| | - Priscila Helena dos Santos
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
| | - Marcelo Fábio Gouveia Nogueira
- Multi-User Laboratory of Phytomedicines Pharmacology, and Biotechnology (PhitoPharmaTec), Department of Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-000, Brazil
- Laboratory of Embryonic Micromanipulation, Department of Biological Sciences, School of Sciences and Languages, São Paulo State University (UNESP), Assis 19806-900, Brazil
| |
Collapse
|
107
|
Un B, Cetinkaya-Un B, Akpolat M, Andic F, Yazir Y. The effects of human amnion membrane-derived mesenchymal stem cells conditioned medium on ionizing radiation-induced premature ovarian failure and endoplasmic reticulum stress-related apoptosis mechanism. Eur J Obstet Gynecol Reprod Biol 2023; 288:191-197. [PMID: 37566962 DOI: 10.1016/j.ejogrb.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 05/07/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023]
Abstract
OBJECTIVES Premature ovarian failure (POF) is defined as the cessation of menstrual periods for at least 4-6 months before the age of 40 years, accompanied by FSH values measuring over 40 IU/L for a month. Radiation therapy, one of the cancer treatment methods, is known to accelerate ovarian aging by reducing and eliminating the number of primordial follicles in the ovarian follicle pool. Ionizing radiation has been reported to cause POF. The objective of this study is to investigate the impact of mesenchymal stem cell conditioned medium (hAMSCs-CM), which is isolated from the amniotic membrane of human placenta, on premature ovarian failure (POF) caused by whole-body irradiation. The study will focus on the ER stress and apoptosis mechanisms in the process. STUDY DISAYN A POF model was created by exposing rats to 7 Gy of whole-body irradiation. Serum-free hAMSCs-CM were then administered via the tail vein. Follicle count was performed on the ovaries, and immunohistochemistry was used to determine the expressions of GRP78, CHOP, IRE-1, caspase-12, caspase-9, caspase-3. TUNEL was also carried out, and levels of serum FSH, LH, E2, AMH, and oxidative stress marker 8-OHdG were measured. RESULTS AND CONCLUSION The application of hAMSCs-CM has been found to have a positive impact on follicles affected by radiation. After treatment, the number of primordial, primary, secondary, and graafian follicles, which had previously decreased due to radiation, showed an increase. Furthermore, the number of atretic follicles, which had been increasing due to radiation, showed a decrease. ER is one of the targets affected by ionizing radiation. After ionizing radiation, the expressions of ER stress-related markers and apoptosis markers increased in the ovary. After hAMSCs-CM administration, the expressions of these markers and number of TUNEL-positive cells decreased. Following irradiation, anti-mullerian hormone (AMH) and estradiol (E2) levels decreased, while follicle stimulating hormone (FSH) and luteinizing hormone (LH) levels increased. After administration of hAMSCs-CM, AMH and E2 levels increased, while FSH and LH levels decreased. Amnion membrane-derived mesenchymal stem cell conditioned medium can play a therapeutic role in ionizing radiation-induced premature ovarian failure by reducing endoplasmic reticulum stress and apoptosis.
Collapse
Affiliation(s)
- Burak Un
- Department of Obstetrics and Gynecology, Adana City Training and Research Hospital, 01370 Adana, Turkey; Department of Histology and Embryology, Medicine Faculty, Zonguldak Bulent Ecevit University, 67600 Zonguldak, Turkey.
| | - Busra Cetinkaya-Un
- IVF Unit, Adana City Training and Research Hospital, 01370 Adana, Turkey; Department of Histology and Embryology, Medicine Faculty, Zonguldak Bulent Ecevit University, 67600 Zonguldak, Turkey.
| | - Meryem Akpolat
- Department of Histology and Embryology, Medicine Faculty, Zonguldak Bulent Ecevit University, 67600 Zonguldak, Turkey.
| | - Fundagul Andic
- Department of Radiation Oncology, Medicine Faculty, Cukurova University, 01380 Adana, Turkey.
| | - Yusufhan Yazir
- Department of Histology and Embryology, Medicine Faculty, Kocaeli University, 41001 Kocaeli, Turkey.
| |
Collapse
|
108
|
Cheng D, Zhou T, Liu H, Li L, Xuan Y, Huang L, Liu Y, Zhang X, Wei W, Wu H. MANF inhibits Sjögren's syndrome salivary gland epithelial cell apoptosis and antigen expression of Ro52/SSA through endoplasmic reticulum stress/autophagy pathway. Int Immunopharmacol 2023; 122:110582. [PMID: 37393840 DOI: 10.1016/j.intimp.2023.110582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/13/2023] [Accepted: 06/25/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Sjögren's syndrome (SS) is a typical autoimmune disease characterized by lymphocyte infiltration accompanied by the production of Ro52/SSA and La/SSB autoantibodies against whole body ribonucleoprotein particles. The release of type I IFN can induce endoplasmic reticulum stress (ERS) in submandibular gland cells. ERS not only produces a large number of Ro52/SSA antigens and changes their location, but also down-regulates autophagy and increases apoptosis. METHOD We collected human submandibular gland tissue samples, established an Experimental Sjögren's syndrome (ESS) mouse model, and used submandibular gland cells to test whether Mesencephalic astrocyte-derived neurotrophic factor (MANF) could reverse ERS-induced autophagy downregulation and reduce apoptosis and Ro52/SSA antigen expression. RESULT It was found that MANF could reduce lymphocyte infiltration and the proportion of CD4+ T cell subsets in the salivary glands, reduce the phosphorylation of AKT and mTOR proteins and the expression of ERS-related proteins, and increase the expression of autophagy proteins. We also found that MANF can reduce the expression of Ro52/SSA antigen on the cell membrane and reduce apoptosis. CONCLUSION In short, we found that MANF can activate autophagy, inhibit apoptosis and reduce the expression of Ro52/SSA by regulating the AKT/mTOR/LC3B signaling pathway. The above results suggest that MANF may be a protective factor against SS.
Collapse
Affiliation(s)
- Danqian Cheng
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Tongtong Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Hui Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Lijun Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Yuhao Xuan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Lijun Huang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Yuqi Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Xiao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China; Anhui Provincial Institute of Translation Medicine, Hefei 230032, China
| | - Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei 230032, Anhui, China; Anhui Provincial Institute of Translation Medicine, Hefei 230032, China.
| |
Collapse
|
109
|
Lamichhane PP, Samir P. Cellular Stress: Modulator of Regulated Cell Death. BIOLOGY 2023; 12:1172. [PMID: 37759572 PMCID: PMC10525759 DOI: 10.3390/biology12091172] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Cellular stress response activates a complex program of an adaptive response called integrated stress response (ISR) that can allow a cell to survive in the presence of stressors. ISR reprograms gene expression to increase the transcription and translation of stress response genes while repressing the translation of most proteins to reduce the metabolic burden. In some cases, ISR activation can lead to the assembly of a cytoplasmic membraneless compartment called stress granules (SGs). ISR and SGs can inhibit apoptosis, pyroptosis, and necroptosis, suggesting that they guard against uncontrolled regulated cell death (RCD) to promote organismal homeostasis. However, ISR and SGs also allow cancer cells to survive in stressful environments, including hypoxia and during chemotherapy. Therefore, there is a great need to understand the molecular mechanism of the crosstalk between ISR and RCD. This is an active area of research and is expected to be relevant to a range of human diseases. In this review, we provided an overview of the interplay between different cellular stress responses and RCD pathways and their modulation in health and disease.
Collapse
Affiliation(s)
| | - Parimal Samir
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
110
|
Arabkari V, Sultana A, Barua D, Webber M, Smith T, Gupta A, Gupta S. UPR-Induced miR-616 Inhibits Human Breast Cancer Cell Growth and Migration by Targeting c-MYC. Int J Mol Sci 2023; 24:13034. [PMID: 37685841 PMCID: PMC10487498 DOI: 10.3390/ijms241713034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/10/2023] Open
Abstract
C/EBP homologous protein (CHOP), also known as growth arrest and DNA damage-inducible protein 153 (GADD153), belongs to the CCAAT/enhancer-binding protein (C/EBP) family. CHOP expression is induced by unfolded protein response (UPR), and sustained CHOP activation acts as a pivotal trigger for ER stress-induced apoptosis. MicroRNA-616 is located within an intron of the CHOP gene. However, the regulation of miR-616 expression during UPR and its function in breast cancer is not clearly understood. Here we show that the expression of miR-616 and CHOP (host gene of miR-616) is downregulated in human breast cancer. Both miR-5p/-3p arms of miR-616 are expressed with levels of the 5p arm higher than the 3p arm. During conditions of ER stress, the expression of miR-616-5p and miR-616-3p arms was concordantly increased primarily through the PERK pathway. Our results show that ectopic expression of miR-616 significantly suppressed cell proliferation and colony formation, whereas knockout of miR-616 increased it. We found that miR-616 represses c-MYC expression via binding sites located in its protein coding region. Furthermore, we show that miR-616 exerted growth inhibitory effects on cells by suppressing c-MYC expression. Our results establish a new role for the CHOP locus by providing evidence that miR-616 can inhibit cell proliferation by targeting c-MYC. In summary, our results suggest a dual function for the CHOP locus, where CHOP protein and miR-616 can cooperate to inhibit cancer progression.
Collapse
Affiliation(s)
- Vahid Arabkari
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland; (V.A.); (A.S.); (D.B.); (M.W.)
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Afrin Sultana
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland; (V.A.); (A.S.); (D.B.); (M.W.)
| | - David Barua
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland; (V.A.); (A.S.); (D.B.); (M.W.)
| | - Mark Webber
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland; (V.A.); (A.S.); (D.B.); (M.W.)
| | - Terry Smith
- Molecular Diagnostic Research Group, College of Science, University of Galway, H91 TK33 Galway, Ireland;
| | - Ananya Gupta
- Discipline of Physiology, School of Medicine, University of Galway, H91 TK33 Galway, Ireland;
| | - Sanjeev Gupta
- Discipline of Pathology, Cancer Progression and Treatment Research Group, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland; (V.A.); (A.S.); (D.B.); (M.W.)
| |
Collapse
|
111
|
Wang H, Pan X, Xiang X, Zhang Y, Chen J, Wen S, Wang J, Gao R, Yang J, Zhi Y, Wen S, Zheng Y, Li T, Ai H, He X, Lu Y, Zhu Y, Li C, Chen Y, Shi G. CRISPR screen identifies the role of RBBP8 in mediating unfolded protein response induced liver damage through regulating protein synthesis. Cell Death Dis 2023; 14:531. [PMID: 37591836 PMCID: PMC10435451 DOI: 10.1038/s41419-023-06046-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Unfolded protein response (UPR) maintains the endoplasmic reticulum (ER) homeostasis, survival, and physiological function of mammalian cells. However, how cells adapt to ER stress under physiological or disease settings remains largely unclear. Here by a genome-wide CRISPR screen, we identified that RBBP8, an endonuclease involved in DNA damage repair, is required for ATF4 activation under ER stress in vitro. RNA-seq analysis suggested that RBBP8 deletion led to impaired cell cycle progression, retarded proliferation, attenuated ATF4 activation, and reduced global protein synthesis under ER stress. Mouse tissue analysis revealed that RBBP8 was highly expressed in the liver, and its expression is responsive to ER stress by tunicamycin intraperitoneal injection. Hepatocytes with RBBP8 inhibition by adenovirus-mediated shRNA were resistant to tunicamycin (Tm)-induced liver damage, cell death, and ER stress response. To study the pathological role of RBBP8 in regulating ATF4 activity, we illustrated that both RBBP8 and ATF4 were highly expressed in liver cancer tissues compared with healthy controls and highly expressed in Ki67-positive proliferating cells within the tumors. Interestingly, overexpression of RBBP8 in vitro promoted ATF4 activation under ER stress, and RBBP8 expression showed a positive correlation with ATF4 expression in liver cancer tissues by co-immunostaining. Our findings provide new insights into the mechanism of how cells adapt to ER stress through the crosstalk between the nucleus and ER and how tumor cells survive under chemotherapy or other anticancer treatments, which suggests potential therapeutic strategies against liver disease by targeting DNA damage repair, UPR or protein synthesis.
Collapse
Affiliation(s)
- Heting Wang
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuya Pan
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoxin Xiang
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yang Zhang
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, USA
- Cancer Biology Program/Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, USA
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shiyi Wen
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jin Wang
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rong Gao
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jifeng Yang
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yaping Zhi
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Siying Wen
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yubao Zheng
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ting Li
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Heying Ai
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuemin He
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Lu
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yanhua Zhu
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, USA.
- Cancer Biology Program/Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, USA.
| | - Yanming Chen
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Guojun Shi
- Department of Endocrinology and Metabolism, Medical Center for Comprehensive Weight Control, Guangdong Provincial Key Laboratory of Diabetology, Guangzhou Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
112
|
Oshitari T. Neurovascular Cell Death and Therapeutic Strategies for Diabetic Retinopathy. Int J Mol Sci 2023; 24:12919. [PMID: 37629100 PMCID: PMC10454228 DOI: 10.3390/ijms241612919] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Diabetic retinopathy (DR) is a major complication of diabetes and a leading cause of blindness worldwide. DR was recently defined as a neurovascular disease associated with tissue-specific neurovascular impairment of the retina in patients with diabetes. Neurovascular cell death is the main cause of neurovascular impairment in DR. Thus, neurovascular cell protection is a potential therapy for preventing the progression of DR. Growing evidence indicates that a variety of cell death pathways, such as apoptosis, necroptosis, ferroptosis, and pyroptosis, are associated with neurovascular cell death in DR. These forms of regulated cell death may serve as therapeutic targets for ameliorating the pathogenesis of DR. This review focuses on these cell death mechanisms and describes potential therapies for the treatment of DR that protect against neurovascular cell death.
Collapse
Affiliation(s)
- Toshiyuki Oshitari
- Department of Ophthalmology and Visual Science, Chiba University Graduate School of Medicine, Inohana 1-8-1, Chuo-ku, Chiba 260-8670, Japan; ; Tel.: +81-43-226-2124; Fax: +81-43-224-4162
- Department of Ophthalmology, School of Medicine, International University of Health and Welfare, 4-3 Kozunomori, Narita 286-8686, Japan
| |
Collapse
|
113
|
Trendel J, Boileau E, Jochem M, Dieterich C, Krijgsveld J. PEPseq quantifies transcriptome-wide changes in protein occupancy and reveals selective translational repression after translational stress. Nucleic Acids Res 2023; 51:e79. [PMID: 37395449 PMCID: PMC10415142 DOI: 10.1093/nar/gkad557] [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] [Received: 12/29/2022] [Revised: 05/24/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023] Open
Abstract
Post-transcriptional gene regulation is accomplished by the interplay of the transcriptome with RNA-binding proteins, which occurs in a dynamic manner in response to altered cellular conditions. Recording the combined occupancy of all proteins binding to the transcriptome offers the opportunity to interrogate if a particular treatment leads to any interaction changes, pointing to sites in RNA that undergo post-transcriptional regulation. Here, we establish a method to monitor protein occupancy in a transcriptome-wide fashion by RNA sequencing. To this end, peptide-enhanced pull-down for RNA sequencing (or PEPseq) uses metabolic RNA labelling with 4-thiouridine (4SU) for light-induced protein-RNA crosslinking, and N-hydroxysuccinimide (NHS) chemistry to isolate protein-crosslinked RNA fragments across all long RNA biotypes. We use PEPseq to investigate changes in protein occupancy during the onset of arsenite-induced translational stress in human cells and reveal an increase of protein interactions in the coding region of a distinct set of mRNAs, including mRNAs coding for the majority of cytosolic ribosomal proteins. We use quantitative proteomics to demonstrate that translation of these mRNAs remains repressed during the initial hours of recovery after arsenite stress. Thus, we present PEPseq as a discovery platform for the unbiased investigation of post-transcriptional regulation.
Collapse
Affiliation(s)
- Jakob Trendel
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Etienne Boileau
- Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| | - Marco Jochem
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Dieterich
- Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany
| | - Jeroen Krijgsveld
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| |
Collapse
|
114
|
Shi S, Ding C, Zhu S, Xia F, Buscho SE, Li S, Motamedi M, Liu H, Zhang W. PERK Inhibition Suppresses Neovascularization and Protects Neurons During Ischemia-Induced Retinopathy. Invest Ophthalmol Vis Sci 2023; 64:17. [PMID: 37566408 PMCID: PMC10424802 DOI: 10.1167/iovs.64.11.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Purpose Retinal ischemia is a common cause of a variety of eye diseases, such as retinopathy of prematurity, diabetic retinopathy, and vein occlusion. Protein kinase RNA-activated-like endoplasmic reticulum (ER) kinase (PERK), one of the main ER stress sensor proteins, has been involved in many diseases. In this study, we investigated the role of PERK in ischemia-induced retinopathy using a mouse model of oxygen-induced retinopathy (OIR). Methods OIR was induced by subjecting neonatal pups to 70% oxygen at postnatal day 7 (P7) followed by returning to room air at P12. GSK2606414, a selective PERK inhibitor, was orally administrated to pups right after they were returned to room air once daily until 1 day before sample collection. Western blot, immunostaining, and quantitative PCR were used to assess PERK phosphorylation, retinal changes, and signaling pathways in relation to PERK inhibition. Results PERK phosphorylation was prominently increased in OIR retinas, which was inhibited by GSK2606414. Concomitantly, PERK inhibition significantly reduced retinal neovascularization (NV) and retinal ganglion cell (RGC) loss, restored astrocyte network, and promoted revascularization. Furthermore, PERK inhibition downregulated the recruitment/proliferation of mononuclear phagocytes but did not affect OIR-upregulated canonical angiogenic pathways. Conclusions Our results demonstrate that PERK is involved in ischemia-induced retinopathy and its inhibition using GSK2606414 could offer an effective therapeutic intervention aimed at alleviating retinal NV while preventing neuron loss during retinal ischemia.
Collapse
Affiliation(s)
- Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Chun Ding
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Shuang Zhu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Seth E. Buscho
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Shengguo Li
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
- Departments of Neurobiology, University of Texas Medical Branch, Galveston, Texas, United States
| |
Collapse
|
115
|
Perea V, Cole C, Lebeau J, Dolina V, Baron KR, Madhavan A, Kelly JW, Grotjahn DA, Wiseman RL. PERK signaling promotes mitochondrial elongation by remodeling membrane phosphatidic acid. EMBO J 2023; 42:e113908. [PMID: 37306086 PMCID: PMC10390871 DOI: 10.15252/embj.2023113908] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Endoplasmic reticulum (ER) stress and mitochondrial dysfunction are linked in the onset and pathogenesis of numerous diseases. This has led to considerable interest in defining the mechanisms responsible for regulating mitochondria during ER stress. The PERK signaling arm of the unfolded protein response (UPR) has emerged as a prominent ER stress-responsive signaling pathway that regulates diverse aspects of mitochondrial biology. Here, we show that PERK activity promotes adaptive remodeling of mitochondrial membrane phosphatidic acid (PA) to induce protective mitochondrial elongation during acute ER stress. We find that PERK activity is required for ER stress-dependent increases in both cellular PA and YME1L-dependent degradation of the intramitochondrial PA transporter PRELID1. These two processes lead to the accumulation of PA on the outer mitochondrial membrane where it can induce mitochondrial elongation by inhibiting mitochondrial fission. Our results establish a new role for PERK in the adaptive remodeling of mitochondrial phospholipids and demonstrate that PERK-dependent PA regulation adapts organellar shape in response to ER stress.
Collapse
Affiliation(s)
- Valerie Perea
- Department of Molecular MedicineScripps ResearchLa JollaCAUSA
| | | | - Justine Lebeau
- Department of Molecular MedicineScripps ResearchLa JollaCAUSA
| | - Vivian Dolina
- Department of Molecular MedicineScripps ResearchLa JollaCAUSA
| | - Kelsey R Baron
- Department of Molecular MedicineScripps ResearchLa JollaCAUSA
| | | | - Jeffery W Kelly
- Department of ChemistryScripps ResearchLa JollaCAUSA
- Skaggs Institute for Chemical BiologyScripps ResearchLa JollaCAUSA
| | - Danielle A Grotjahn
- Department of Integrative, Structural, and Computational BiologyScripps ResearchLa JollaCAUSA
| | - R Luke Wiseman
- Department of Molecular MedicineScripps ResearchLa JollaCAUSA
| |
Collapse
|
116
|
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: 29] [Impact Index Per Article: 14.5] [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
|
117
|
Pakarinen E, Lindholm P. CDNF and MANF in the brain dopamine system and their potential as treatment for Parkinson's disease. Front Psychiatry 2023; 14:1188697. [PMID: 37555005 PMCID: PMC10405524 DOI: 10.3389/fpsyt.2023.1188697] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/23/2023] [Indexed: 08/10/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by gradual loss of midbrain dopamine neurons, leading to impaired motor function. Preclinical studies have indicated cerebral dopamine neurotrophic factor (CDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF) to be potential therapeutic molecules for the treatment of PD. CDNF was proven to be safe and well tolerated when tested in Phase I-II clinical trials in PD patients. Neuroprotective and neurorestorative effects of CDNF and MANF were demonstrated in animal models of PD, where they promoted the survival of dopamine neurons and improved motor function. However, biological roles of endogenous CDNF and MANF proteins in the midbrain dopamine system have been less clear. In addition to extracellular trophic activities, CDNF/MANF proteins function intracellularly in the endoplasmic reticulum (ER), where they modulate protein homeostasis and protect cells against ER stress by regulating the unfolded protein response (UPR). Here, our aim is to give an overview of the biology of endogenous CDNF and MANF in the brain dopamine system. We will discuss recent studies on CDNF and MANF knockout animal models, and effects of CDNF and MANF in preclinical models of PD. To elucidate possible roles of CDNF and MANF in human biology, we will review CDNF and MANF tissue expression patterns and regulation of CDNF/MANF levels in human diseases. Finally, we will discuss novel findings related to the molecular mechanism of CDNF and MANF action in ER stress, UPR, and inflammation, all of which are mechanisms potentially involved in the pathophysiology of PD.
Collapse
Affiliation(s)
| | - Päivi Lindholm
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
118
|
Zhao Y, Lin H, Ma J, Cui Y. Botulinum toxin type A activates protective autophagy by modulating endoplasmic reticulum stress in hypoxia/reoxygenation-treated endothelial cells. Heliyon 2023; 9:e17907. [PMID: 37483799 PMCID: PMC10362318 DOI: 10.1016/j.heliyon.2023.e17907] [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: 01/01/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/25/2023] Open
Abstract
Botulinum toxin type A (BTXA) previously protected endothelial cells in free skin flaps from ischemia/reperfusion injury by inducing autophagy. Endoplasmic reticulum (ER) stress-autophagy activation may have a role in BTXA-antagonized ischemia/reperfusion damage in human dermal microvascular endothelial cells (HDMECs), however, this has yet to be proven. HDMECs were pretreated with BTXA at various concentrations for 12 h before being subjected to hypoxia and reoxygenation (H/R). Cell Count Kit 8 (CCK8) and Western blot (WB) data showed that H/R treatment significantly increased the expression of ER stress (GRP78, CHOP) and autophagy (LC3II/I, Beclin-1) proteins. The optimal BTXA pretreatment concentration was 1.6 U/mL, which resulted in the highest levels of cell survival and expression of ER stress and autophagy. Following pretreatment with 1.6 U/mL BTXA and the addition of the ER stress inducer Thapsigargin (Tg), the ER stress inhibitor 4-phenylbutyrate (4-PBA), and the inhibitor of autophagy Bafilomycin A1 (Baf A1), respectively, HDMECs were then subjected to H/R treatment. Cell survival and the percentage of ethynyldeoxyuridine-labeled cells in the BTXA pretreatment groups were reduced by the addition of Tg, 4-PBA, and Baf A1. While the expression of GRP78, CHOP, and LC3 was upregulated by Tg and Baf A1, it was downregulated by 4-PBA. The findings showed that ER stress produced by BTXA pretreatment triggers protective autophagy and protects HDMECs from H/R damage. There were no cytoprotective effects from either excessive activation or reduction of ER stress. Our results are consistent with the notion that autophagy and moderate ER stress are critical for cellular longevity and, consequently, functional integrity and may represent a potential therapeutic target.
Collapse
|
119
|
Davies JP, Sivadas A, Keller KR, Wojcikiewicz RJ, Plate L. SARS-CoV-2 Nonstructural Proteins 3 and 4 tune the Unfolded Protein Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.22.537917. [PMID: 37162862 PMCID: PMC10168236 DOI: 10.1101/2023.04.22.537917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Coronaviruses (CoV), including SARS-CoV-2, modulate host proteostasis through activation of stress-responsive signaling pathways such as the Unfolded Protein Response (UPR), which remedies misfolded protein accumulation by attenuating translation and increasing protein folding capacity. While CoV nonstructural proteins (nsps) are essential for infection, little is known about the role of nsps in modulating the UPR. We characterized the impact of SARS-CoV-2 nsp4, a key driver of replication, on the UPR using quantitative proteomics to sensitively detect pathway-wide upregulation of effector proteins. We find nsp4 preferentially activates the ATF6 and PERK branches of the UPR. Previously, we found an N-terminal truncation of nsp3 (nsp3.1) can suppress pharmacological ATF6 activation. To determine how nsp3.1 and nsp4 tune the UPR, their co-expression demonstrated that nsp3.1 suppresses nsp4-mediated PERK, but not ATF6 activation. Re-analysis of SARS-CoV-2 infection proteomics data revealed time-dependent activation of PERK targets early in infection, which subsequently fades. This temporal regulation suggests a role for nsp3 and nsp4 in tuning the PERK pathway to attenuate host translation beneficial for viral replication while avoiding later apoptotic signaling caused by chronic activation. This work furthers our understanding of CoV-host proteostasis interactions and highlights the power of proteomic methods for systems-level analysis of the UPR.
Collapse
Affiliation(s)
| | - Athira Sivadas
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
| | | | | | - Lars Plate
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
- Department of Chemistry, Vanderbilt University, Nashville, TN
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
120
|
Perea V, Baron KR, Dolina V, Aviles G, Rosarda JD, Guo X, Kampmann M, Wiseman RL. Pharmacologic Activation of a Compensatory Integrated Stress Response Kinase Promotes Mitochondrial Remodeling in PERK-deficient Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.11.532186. [PMID: 36945406 PMCID: PMC10029010 DOI: 10.1101/2023.03.11.532186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
Abstract
The integrated stress response (ISR) comprises the eIF2α kinases PERK, GCN2, HRI, and PKR, which induce translational and transcriptional signaling in response to diverse insults. Deficiencies in PERK signaling lead to mitochondrial dysfunction and contribute to the pathogenesis of numerous diseases. We define the potential for pharmacologic activation of compensatory eIF2α kinases to rescue ISR signaling and promote mitochondrial adaptation in PERK-deficient cells. We show that the HRI activator BtdCPU and GCN2 activator halofuginone promote ISR signaling and rescue ER stress sensitivity in PERK-deficient cells. However, BtdCPU induces mitochondrial depolarization, leading to mitochondrial fragmentation and activation of the OMA1-DELE1-HRI signaling axis. In contrast, halofuginone promotes mitochondrial elongation and adaptive mitochondrial respiration, mimicking regulation induced by PERK. This shows halofuginone can compensate for deficiencies in PERK signaling and promote adaptive mitochondrial remodeling, highlighting the potential for pharmacologic ISR activation to mitigate mitochondrial dysfunction and motivating the pursuit of highly-selective ISR activators.
Collapse
Affiliation(s)
- Valerie Perea
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Authors contributed equally
| | - Kelsey R. Baron
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
- Authors contributed equally
| | - Vivian Dolina
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Giovanni Aviles
- Department of Biophysics and Biochemistry and Institute for Neurodegenerative Diseases, UCSF, San Francisco, CA 94158
| | - Jessica D. Rosarda
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Xiaoyan Guo
- Department of Biophysics and Biochemistry and Institute for Neurodegenerative Diseases, UCSF, San Francisco, CA 94158
- Department of Genetics and Genome Sciences, University of Connecticut Health, Farmington, CT 06030
| | - Martin Kampmann
- Department of Biophysics and Biochemistry and Institute for Neurodegenerative Diseases, UCSF, San Francisco, CA 94158
| | - R. Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
121
|
Li B, Yang H, Wu H, Wang H, Ye Z, Wu Y, Chen Y, Tang H. Hydroquinone-induced endoplasmic reticulum stress affects TK6 cell autophagy and apoptosis via ATF6-mTOR. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 37148176 DOI: 10.1002/tox.23814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/23/2023] [Accepted: 04/16/2023] [Indexed: 05/08/2023]
Abstract
Hydroquinone (HQ), one of the main active metabolites of benzene in vivo, 7is commonly used as a surrogate for benzene in in vitro studies and has been shown to be cytotoxic. The aim of this study was to investigate the role of endoplasmic reticulum stress (ERS) in HQ-induced autophagy and apoptosis in human lymphoblastoid cells (TK6) and how activating transcription factor 6 (ATF-6) is involved. We treated TK6 cells with HQ to establish a cytotoxicity model and found that HQ induced cellular ERS, autophagy and apoptosis by Western blot, flow cytometry and transmission electron microscopy. In addition, inhibition of both reactive oxygen species (ROS) and ERS inhibited cellular autophagy and apoptosis, suggesting that ERS may be induced by ROS, which in turn affects autophagy and apoptosis. Our study also found that HQ could inhibit ATF6 expression and mTOR activation. Knockdown of ATF6 enhanced autophagy and apoptosis levels and further inhibited mTOR activation; activation of ATF6 by AA147 enhanced cellular activity, suggesting that ATF6 may affect cellular autophagy and apoptosis through mTOR. In conclusion, our data suggest that ROS mediated ERS may promote autophagy and apoptosis by inhibiting ATF6-mTOR pathway after HQ treatment of TK6 cells.
Collapse
Affiliation(s)
- Boxin Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| | - Hui Yang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| | - Haipeng Wu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| | - Huanhuan Wang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| | - Zhongming Ye
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| | - Yao Wu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| | - Yuting Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
122
|
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: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
123
|
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: 60] [Impact Index Per Article: 30.0] [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
|
124
|
Matai L, Slack FJ. MicroRNAs in Age-Related Proteostasis and Stress Responses. Noncoding RNA 2023; 9:26. [PMID: 37104008 PMCID: PMC10143298 DOI: 10.3390/ncrna9020026] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/28/2023] Open
Abstract
Aging is associated with the accumulation of damaged and misfolded proteins through a decline in the protein homeostasis (proteostasis) machinery, leading to various age-associated protein misfolding diseases such as Huntington's or Parkinson's. The efficiency of cellular stress response pathways also weakens with age, further contributing to the failure to maintain proteostasis. MicroRNAs (miRNAs or miRs) are a class of small, non-coding RNAs (ncRNAs) that bind target messenger RNAs at their 3'UTR, resulting in the post-transcriptional repression of gene expression. From the discovery of aging roles for lin-4 in C. elegans, the role of numerous miRNAs in controlling the aging process has been uncovered in different organisms. Recent studies have also shown that miRNAs regulate different components of proteostasis machinery as well as cellular response pathways to proteotoxic stress, some of which are very important during aging or in age-related pathologies. Here, we present a review of these findings, highlighting the role of individual miRNAs in age-associated protein folding and degradation across different organisms. We also broadly summarize the relationships between miRNAs and organelle-specific stress response pathways during aging and in various age-associated diseases.
Collapse
Affiliation(s)
| | - Frank J. Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
125
|
He D, Tao L, Cai B, Chen X, Wang Y, Li S, Liao C, Chen Y, Chen J, Liu Z, Wu Y. eIF2α incites photoreceptor cell and retina damage by all-trans-retinal. J Biol Chem 2023; 299:104686. [PMID: 37031820 DOI: 10.1016/j.jbc.2023.104686] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/11/2023] Open
Abstract
Dry age-related macular degeneration (AMD) and recessive Stargardt's disease (STGD1) lead to irreversible blindness in humans. The accumulation of all-trans-retinal (atRAL) induced by chaos in visual cycle is closely associated with retinal atrophy in dry AMD and STGD1, but its critical downstream signaling molecules remain ambiguous. Here, we reported that activation of eukaryotic translation initiation factor 2α (eIF2α) by atRAL promoted retinal degeneration and photoreceptor loss through activating c-Jun N-terminal kinase (JNK) signaling-dependent apoptosis and gasdermin E (GSDME)-mediated pyroptosis. We determined that eIF2α activation by atRAL in photoreceptor cells resulted from endoplasmic reticulum (ER) homeostasis disruption caused at least in part by reactive oxygen species (ROS) production, and it activated JNK signaling independent of and dependent on activating transcription factor 4 (ATF4) and the ATF4/transcription factor C/EBP homologous protein (CHOP) axis. CHOP overexpression induced apoptosis of atRAL-loaded photoreceptor cells through activating JNK signaling rather than inhibiting the expression of anti-apoptotic gene Bcl2. JNK activation by eIF2α facilitated photoreceptor cell apoptosis caused by atRAL via caspase-3 activation and DNA damage. Additionally, we demonstrated that eIF2α was activated in neural retina of light-exposed Abca4-/-Rdh8-/- mice, a model that shows severe defects in atRAL clearance and displays primary features of human dry AMD and STGD1. Of note, inhibition of eIF2α activation by salubrinal effectively ameliorated retinal degeneration and photoreceptor apoptosis in Abca4-/-Rdh8-/- mice upon light exposure. The results of this study suggest that eIF2α is an important target to develop drug therapies for the treatment of dry AMD and STGD1.
Collapse
Affiliation(s)
- Danxue He
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lei Tao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Binxiang Cai
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiangjun Chen
- Eye Center of the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, China; Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kaixuan Road, Hangzhou, China
| | - Yan Wang
- Department of Ophthalmology, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| | - Shiying Li
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Chunyan Liao
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuling Chen
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jingmeng Chen
- School of Medicine, Xiamen University, Xiamen, China
| | - Zuguo Liu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Eye Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Shenzhen Research Institute of Xiamen University, Shenzhen, China; Xiamen Eye Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
126
|
Fernández-Alfara M, Sibilio A, Martin J, Tusquets Uxó E, Malumbres M, Alcalde V, Chanes V, Cañellas-Socias A, Palomo-Ponce S, Batlle E, Méndez R. Antitumor T-cell function requires CPEB4-mediated adaptation to chronic endoplasmic reticulum stress. EMBO J 2023; 42:e111494. [PMID: 36919984 PMCID: PMC10152139 DOI: 10.15252/embj.2022111494] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 03/16/2023] Open
Abstract
Tumor growth is influenced by a complex network of interactions between multiple cell types in the tumor microenvironment (TME). These constrained conditions trigger the endoplasmic reticulum (ER) stress response, which extensively reprograms mRNA translation. When uncontrolled over time, chronic ER stress impairs the antitumor effector function of CD8 T lymphocytes. How cells promote adaptation to chronic stress in the TME without the detrimental effects of the terminal unfolded protein response (UPR) is unknown. Here, we find that, in effector CD8 T lymphocytes, RNA-binding protein CPEB4 constitutes a new branch of the UPR that allows cells to adapt to sustained ER stress, yet remains decoupled from the terminal UPR. ER stress, induced during CD8 T-cell activation and effector function, triggers CPEB4 expression. CPEB4 then mediates chronic stress adaptation to maintain cellular fitness, allowing effector molecule production and cytotoxic activity. Accordingly, this branch of the UPR is required for the antitumor effector function of T lymphocytes, and its disruption in these cells exacerbates tumor growth.
Collapse
Affiliation(s)
- Marcos Fernández-Alfara
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Annarita Sibilio
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Judit Martin
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Elsa Tusquets Uxó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marina Malumbres
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Victor Alcalde
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Verónica Chanes
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Adrià Cañellas-Socias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sergio Palomo-Ponce
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Raúl Méndez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
127
|
Zhao N, Gao Y, Jia H, Jiang X. Anti-apoptosis effect of traditional Chinese medicine in the treatment of cerebral ischemia-reperfusion injury. Apoptosis 2023; 28:702-729. [PMID: 36892639 DOI: 10.1007/s10495-023-01824-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2023] [Indexed: 03/10/2023]
Abstract
Cerebral ischemia, one of the leading causes of neurological dysfunction of brain cells, muscle dysfunction, and death, brings great harm and challenges to individual health, families, and society. Blood flow disruption causes decreased glucose and oxygen, insufficient to maintain normal brain tissue metabolism, resulting in intracellular calcium overload, oxidative stress, neurotoxicity of excitatory amino acids, and inflammation, ultimately leading to neuronal cell necrosis, apoptosis, or neurological abnormalities. This paper summarizes the specific mechanism of cell injury that apoptosis triggered by reperfusion after cerebral ischemia, the related proteins involved in apoptosis, and the experimental progress of herbal medicine treatment through searching, analyzing, and summarizing the PubMed and Web Of Science databases, which includes active ingredients of herbal medicine, prescriptions, Chinese patent medicines, and herbal extracts, providing a new target or new strategy for drug treatment, and providing a reference for future experimental directions and using them to develop suitable small molecule drugs for clinical application. With the research of anti-apoptosis as the core, it is important to find highly effective, low toxicity, safe and cheap compounds from natural plants and animals with abundant resources to prevent and treat Cerebral ischemia/reperfusion (I/R) injury (CIR) and solve human suffering. In addition, understanding and summarizing the apoptotic mechanism of cerebral ischemia-reperfusion injury, the microscopic mechanism of CIR treatment, and the cellular pathways involved will help to develop new drugs.
Collapse
Affiliation(s)
- Nan Zhao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yuhe Gao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Hongtao Jia
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xicheng Jiang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China.
| |
Collapse
|
128
|
Sassano ML, van Vliet AR, Vervoort E, Van Eygen S, Van den Haute C, Pavie B, Roels J, Swinnen JV, Spinazzi M, Moens L, Casteels K, Meyts I, Pinton P, Marchi S, Rochin L, Giordano F, Felipe-Abrio B, Agostinis P. PERK recruits E-Syt1 at ER-mitochondria contacts for mitochondrial lipid transport and respiration. J Cell Biol 2023; 222:e202206008. [PMID: 36821088 PMCID: PMC9998969 DOI: 10.1083/jcb.202206008] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/07/2022] [Accepted: 01/19/2023] [Indexed: 02/24/2023] Open
Abstract
The integrity of ER-mitochondria appositions ensures transfer of ions and phospholipids (PLs) between these organelles and exerts crucial effects on mitochondrial bioenergetics. Malfunctions within the ER-mitochondria contacts altering lipid trafficking homeostasis manifest in diverse pathologies, but the molecular effectors governing this process remain ill-defined. Here, we report that PERK promotes lipid trafficking at the ER-mitochondria contact sites (EMCS) through a non-conventional, unfolded protein response-independent, mechanism. PERK operates as an adaptor for the recruitment of the ER-plasma membrane tether and lipid transfer protein (LTP) Extended-Synaptotagmin 1 (E-Syt1), within the EMCS. In resting cells, the heterotypic E-Syt1-PERK interaction endorses transfer of PLs between the ER and mitochondria. Weakening the E-Syt1-PERK interaction or removing the lipid transfer SMP-domain of E-Syt1, compromises mitochondrial respiration. Our findings unravel E-Syt1 as a PERK interacting LTP and molecular component of the lipid trafficking machinery of the EMCS, which critically maintains mitochondrial homeostasis and fitness.
Collapse
Affiliation(s)
- Maria Livia Sassano
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Alexander R. van Vliet
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ellen Vervoort
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Sofie Van Eygen
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Chris Van den Haute
- Research Group for Neurobiology and Gene Therapy, Department of Neuroscience, Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | | | - Joris Roels
- VIB-bioimaging Center UGent, Ghent, Belgium
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marco Spinazzi
- Neuromuscular Reference Center, CHU Angers, Angers, France
| | - Leen Moens
- Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Kristina Casteels
- Woman and Child, Department for Development and Regeneration, KU Leuven, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Isabelle Meyts
- Laboratory for Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | | | | | - Blanca Felipe-Abrio
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| |
Collapse
|
129
|
Pre-ischaemic Treatment with Enriched Environment Alleviates Acute Neuronal Injury by Inhibiting Endoplasmic Reticulum Stress-dependent Autophagy and Apoptosis. Neuroscience 2023; 513:14-27. [PMID: 36549603 DOI: 10.1016/j.neuroscience.2022.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
Enriched environment (EE) is effective in preventing cerebral ischemia-reperfusion (I/R) injury. However, little is known about the mechanism underlying the neuroprotection of EE preprocessing. Endoplasmic reticulum (ER) stress has been demonstrated to be extensively involved in I/R injury. We aimed to investigate the potential regulatory mechanism of ER stress in the neuroprotection of pre-ischemic EE. Rats were subjected to middle cerebral artery occlusion (MCAO) or sham surgery after 4 weeks of exposure in standard or enriched environments. We found that EE pretreatment alleviates acute neuronal injury after MCAO, as shown by reduced infarct volume and neurological deficit score. The expression of ER stress-related proteins, markers of autophagy, and apoptosis were detected to investigate the underlying mechanism. Our results showed that pre-ischemic EE inhibited the ER stress, as evidenced by the inactivation of activating transcription factor 6 (ATF6), protein kinase RNA (PKR)-like ER kinase (PERK), and inositol-requiring enzyme 1 (IRE1) pathways. Moreover, the rats reared in EE were detected with lower autophagic activity and apoptosis levels. The decrease in activating transcription factor 4 (ATF4), C/EBP homologous protein (CHOP), and phospho-c-Jun N-terminal kinases (p-JNK) expression suggested EE pretreatment might inhibit autophagy and apoptosis via modulating ER stress-mediated PERK-ATF4-CHOP and IRE1-JNK signal pathways, which provides a new idea for the prevention of the deleterious cerebral and functional consequences of ischemic stroke.
Collapse
|
130
|
Waisner H, Grieshaber B, Saud R, Henke W, Stephens EB, Kalamvoki M. SARS-CoV-2 Harnesses Host Translational Shutoff and Autophagy To Optimize Virus Yields: the Role of the Envelope (E) Protein. Microbiol Spectr 2023; 11:e0370722. [PMID: 36622177 PMCID: PMC9927098 DOI: 10.1128/spectrum.03707-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/07/2022] [Indexed: 01/10/2023] Open
Abstract
The SARS-CoV-2 virion is composed of four structural proteins: spike (S), nucleocapsid (N), membrane (M), and envelope (E). E spans the membrane a single time and is the smallest, yet most enigmatic of the structural proteins. E is conserved among coronaviruses and has an essential role in virus-mediated pathogenesis. We found that ectopic expression of E had deleterious effects on the host cell as it activated stress responses, leading to LC3 lipidation and phosphorylation of the translation initiation factor eIF2α that resulted in host translational shutoff. During infection E is highly expressed, although only a small fraction is incorporated into virions, suggesting that E activity is regulated and harnessed by the virus to its benefit. Consistently, we found that proteins from heterologous viruses, such as the γ1 34.5 protein of herpes simplex virus 1, prevented deleterious effects of E on the host cell and allowed for E protein accumulation. This observation prompted us to investigate whether other SARS-CoV-2 structural proteins regulate E. We found that the N and M proteins enabled E protein accumulation, whereas S did not. While γ1 34.5 protein prevented deleterious effects of E on the host cells, it had a negative effect on SARS-CoV-2 replication. The negative effect of γ1 34.5 was most likely associated with failure of SARS-CoV-2 to divert the translational machinery and with deregulation of autophagy. Overall, our data suggest that SARS-CoV-2 causes stress responses and subjugates these pathways, including host protein synthesis (phosphorylated eIF2α) and autophagy, to support optimal virus replication. IMPORTANCE In late 2019, a new β-coronavirus, SARS-CoV-2, entered the human population causing a pandemic that has resulted in over 6 million deaths worldwide. Although closely related to SARS-CoV, the mechanisms of SARS-CoV-2 pathogenesis are not fully understood. We found that ectopic expression of the SARS-CoV-2 E protein had detrimental effects on the host cell, causing metabolic alterations, including shutoff of protein synthesis and mobilization of cellular resources through autophagy activation. Coexpression of E with viral proteins known to subvert host antiviral responses such as autophagy and translational inhibition, either from SARS-CoV-2 or from heterologous viruses, increased cell survival and E protein accumulation. However, such factors were found to negatively impact SARS-CoV-2 infection, as autophagy contributes to formation of viral membrane factories and translational control offers an advantage for viral gene expression. Overall, SARS-CoV-2 has evolved mechanisms to harness host functions that are essential for virus replication.
Collapse
Affiliation(s)
- Hope Waisner
- University of Kansas Medical Center, Department of Microbiology, Molecular Genetics, and Immunology, Kansas City, Kansas, USA
| | - Brandon Grieshaber
- University of Kansas Medical Center, Department of Microbiology, Molecular Genetics, and Immunology, Kansas City, Kansas, USA
| | - Rabina Saud
- University of Kansas Medical Center, Department of Microbiology, Molecular Genetics, and Immunology, Kansas City, Kansas, USA
| | - Wyatt Henke
- University of Kansas Medical Center, Department of Microbiology, Molecular Genetics, and Immunology, Kansas City, Kansas, USA
| | - Edward B. Stephens
- University of Kansas Medical Center, Department of Microbiology, Molecular Genetics, and Immunology, Kansas City, Kansas, USA
| | - Maria Kalamvoki
- University of Kansas Medical Center, Department of Microbiology, Molecular Genetics, and Immunology, Kansas City, Kansas, USA
| |
Collapse
|
131
|
Bola S, Subramanian P, Calzia D, Dahl A, Panfoli I, Funk RHW, Roehlecke C. Analysis of Electric Field Stimulation in Blue Light Stressed 661W Cells. Int J Mol Sci 2023; 24:ijms24043433. [PMID: 36834840 PMCID: PMC9965974 DOI: 10.3390/ijms24043433] [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: 01/05/2023] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/11/2023] Open
Abstract
Though electrical stimulation is used as a therapeutic approach to treat retinal and spinal injuries, many protective mechanisms at cellular level have not been elucidated. We performed a detailed analysis of cellular events in blue light (Li) stressed 661W cells, which were subjected to direct current electric field (EF) stimulation. Our findings revealed that EF stimulation induced protective effects in 661W cells from Li-induced stress by multiple defense mechanisms, such as increase in mitochondrial activity, gain in mitochondrial potential, increase in superoxide levels, and the activation of unfolded protein response (UPR) pathways, all leading to an enhanced cell viability and decreased DNA damage. Here, our genetic screen results revealed the UPR pathway to be a promising target to ameliorate Li-induced stress by EF stimulation. Thus, our study is important for a knowledgeable transfer of EF stimulation into clinical application.
Collapse
Affiliation(s)
- Sharanya Bola
- Institute of Anatomy, TU Dresden, D-01304 Dresden, Germany
| | - Pallavi Subramanian
- Institute of Clinical Chemistry and Laboratory Medicine, TU Dresden, D-01069 Dresden, Germany
| | - Daniela Calzia
- Department of Pharmacy—DIFAR, Biochemistry and Physiology Lab., University of Genoa, 16126 Genova, Italy
| | - Andreas Dahl
- Deep Sequencing Group SFB 655, Biotechnology Center, TU Dresden, D-01069 Dresden, Germany
| | - Isabella Panfoli
- Department of Pharmacy—DIFAR, Biochemistry and Physiology Lab., University of Genoa, 16126 Genova, Italy
| | - Richard H. W. Funk
- Institute of Anatomy, TU Dresden, D-01304 Dresden, Germany
- Correspondence:
| | - Cora Roehlecke
- Institute of Anatomy, TU Dresden, D-01304 Dresden, Germany
| |
Collapse
|
132
|
Shen WB, Wang B, Yao R, Goetzinger KR, Wu S, Gao H, Yang P. Obesity impacts placental function through activation of p-IRE1a-XBP1s signaling. Front Cell Dev Biol 2023; 11:1023327. [PMID: 36819099 PMCID: PMC9929362 DOI: 10.3389/fcell.2023.1023327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Maternal obesity is associated with a variety of obstetrical outcomes including stillbirth, preeclampsia, and gestational diabetes, and increases the risk of fetuses for congenital heart defects. Obesity during pregnancy represents a major contribution to metabolic dysregulation, which not only plays a key role in the pathogenesis of adverse outcome but also can potently induce endoplasmic reticulum (ER) stress. However, the mechanism associating such an obesogenic metabolic environment and adverse pregnancy outcomes has remained poorly understood. In this study, we aimed to determine whether the ER stress pathways (also named unfolded protein response (UPR)) were activated in the placenta by obesity. We collected placenta from the obese pregnancy (n = 12) and non-obese pregnancy (n = 12) following delivery by Caesarean-section at term. The specimens were assessed with immunocytochemistry staining and RT-QPCR. Our results revealed that in the obese placenta, p-IRE1α and XBP1s were significantly increased, CHOP and nine UPR chaperone genes were upregulated, including GRP95, PDIA6, Calnexin, p58IPK, SIL-1, EDEM, Herp, GRP58 and Calreticulin. However, Perk and BiP are not activated in the obese placenta. Our data suggest that upregulated p-IRE1α and XBP1s signaling, and UPR chaperone genes may play an important role in maternal obesity-induced placental pathology. In conclusion, this is the first report on ER stress and UPR activation in the placenta of maternal obesity. Our findings represent the first step in the understanding of one of the key ER signaling pathways, also referred to IRE1α-XBP1, in placental pathophysiology affected by obesity, which may be an important mechanism accounting for the observed higher maternal and perinatal risks.
Collapse
Affiliation(s)
- Wei-Bin Shen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bingbing Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ruofan Yao
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Katherine R. Goetzinger
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sheng Wu
- Center for Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University School of Medicine, Philadelphia, PA, United States
| | - Haijun Gao
- Departmentof Physiology and Biophysics, Howard University College of Medicine, Washington, DC, United States
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States,*Correspondence: Peixin Yang,
| |
Collapse
|
133
|
Mann JP, Duan X, Patel S, Tábara LC, Scurria F, Alvarez-Guaita A, Haider A, Luijten I, Page M, Protasoni M, Lim K, Virtue S, O'Rahilly S, Armstrong M, Prudent J, Semple RK, Savage DB. A mouse model of human mitofusin-2-related lipodystrophy exhibits adipose-specific mitochondrial stress and reduced leptin secretion. eLife 2023; 12:e82283. [PMID: 36722855 PMCID: PMC9937658 DOI: 10.7554/elife.82283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/30/2023] [Indexed: 02/02/2023] Open
Abstract
Mitochondrial dysfunction has been reported in obesity and insulin resistance, but primary genetic mitochondrial dysfunction is generally not associated with these, arguing against a straightforward causal relationship. A rare exception, recently identified in humans, is a syndrome of lower body adipose loss, leptin-deficient severe upper body adipose overgrowth, and insulin resistance caused by the p.Arg707Trp mutation in MFN2, encoding mitofusin 2. How the resulting selective form of mitochondrial dysfunction leads to tissue- and adipose depot-specific growth abnormalities and systemic biochemical perturbation is unknown. To address this, Mfn2R707W/R707W knock-in mice were generated and phenotyped on chow and high fat diets. Electron microscopy revealed adipose-specific mitochondrial morphological abnormalities. Oxidative phosphorylation measured in isolated mitochondria was unperturbed, but the cellular integrated stress response was activated in adipose tissue. Fat mass and distribution, body weight, and systemic glucose and lipid metabolism were unchanged, however serum leptin and adiponectin concentrations, and their secretion from adipose explants were reduced. Pharmacological induction of the integrated stress response in wild-type adipocytes also reduced secretion of leptin and adiponectin, suggesting an explanation for the in vivo findings. These data suggest that the p.Arg707Trp MFN2 mutation selectively perturbs mitochondrial morphology and activates the integrated stress response in adipose tissue. In mice, this does not disrupt most adipocyte functions or systemic metabolism, whereas in humans it is associated with pathological adipose remodelling and metabolic disease. In both species, disproportionate effects on leptin secretion may relate to cell autonomous induction of the integrated stress response.
Collapse
Affiliation(s)
- Jake P Mann
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Xiaowen Duan
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Satish Patel
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Luis Carlos Tábara
- Medical Research Council Mitochondrial Biology Unit, University of CambridgeCambridgeUnited Kingdom
| | - Fabio Scurria
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Anna Alvarez-Guaita
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Afreen Haider
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Ineke Luijten
- Centre for Cardiovascular Science, University of EdinburghEdinburghUnited Kingdom
| | | | - Margherita Protasoni
- Medical Research Council Mitochondrial Biology Unit, University of CambridgeCambridgeUnited Kingdom
| | - Koini Lim
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Sam Virtue
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | | | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of CambridgeCambridgeUnited Kingdom
| | - Robert K Semple
- Centre for Cardiovascular Science, University of EdinburghEdinburghUnited Kingdom
- MRC Human Genetics Unit, University of EdinburghEdinburghUnited Kingdom
| | - David B Savage
- Wellcome Trust-MRC Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
134
|
Parwani K, Mandal P. Role of advanced glycation end products and insulin resistance in diabetic nephropathy. Arch Physiol Biochem 2023; 129:95-107. [PMID: 32730131 DOI: 10.1080/13813455.2020.1797106] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
Abstract
Metabolic syndrome (MetS), i.e. a cluster of physiological and biochemical abnormalities can lead to diabetic nephropathy (DN). Insulin resistance, impaired fasting glucose are the main signs and symptoms of MetS. Excess sugar can induce various substantial structural changes like formation of advanced glycation end products (AGEs). AGEs are formed due to reaction of reducing sugars with amino groups of proteins, lipids and nucleic acids. AGEs when bound to the receptor for advanced glycation end products (RAGE) activate increased production of pro-inflammatory markers like interleukin-6 (IL-6), tumour necrosis factor alpha (TNF-α) along with induction of endoplasmic reticulum (ER) stress. Accumulation of AGEs, enhanced reactive oxygen species (ROS) generation and activation of protein kinase C (PKC), are considered to induce glomerular hypertrophy, podocyte apoptosis, therefore contributing to the development and progression of DN. In this review, we decipher different biochemical and physiological factors that link AGEs and DN.
Collapse
Affiliation(s)
- Kirti Parwani
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Gujarat 388421, India
| | - Palash Mandal
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, Gujarat 388421, India
| |
Collapse
|
135
|
Yung HW, Zhao X, Glover L, Burrin C, Pang PC, Jones CJ, Gill C, Duhig K, Olovsson M, Chappell LC, Haslam SM, Dell A, Burton GJ, Charnock-Jones DS. Perturbation of placental protein glycosylation by endoplasmic reticulum stress promotes maladaptation of maternal hepatic glucose metabolism. iScience 2023; 26:105911. [PMID: 36660474 PMCID: PMC9843443 DOI: 10.1016/j.isci.2022.105911] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/30/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Placental hormones orchestrate maternal metabolic adaptations to support pregnancy. We hypothesized that placental ER stress, which characterizes early-onset pre-eclampsia (ePE), compromises glycosylation, reducing hormone bioactivity and these maladaptations predispose the mother to metabolic disease in later life. We demonstrate ER stress reduces the complexity and sialylation of trophoblast protein N-glycosylation, while aberrant glycosylation of vascular endothelial growth factor reduced its bioactivity. ER stress alters the expression of 66 of the 146 genes annotated with "protein glycosylation" and reduces the expression of sialyltransferases. Using mouse placental explants, we show ER stress promotes the secretion of mis-glycosylated glycoproteins. Pregnant mice carrying placentas with junctional zone-specific ER stress have reduced blood glucose, anomalous hepatic glucose metabolism, increased cellular stress and elevated DNA methyltransferase 3A. Using pregnancy-specific glycoproteins as a readout, we also demonstrate aberrant glycosylation of placental proteins in women with ePE, thus providing a mechanistic link between ePE and subsequent maternal metabolic disorders.
Collapse
Affiliation(s)
- Hong Wa Yung
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Luke Glover
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Charlotte Burrin
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Poh-Choo Pang
- Department of Life Sciences, Imperial College London, London, UK
| | - Carolyn J.P. Jones
- Maternal and Fetal Health Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Carolyn Gill
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Kate Duhig
- Maternal and Fetal Health Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Matts Olovsson
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | - Lucy C. Chappell
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Stuart M. Haslam
- Department of Life Sciences, Imperial College London, London, UK
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, UK
| | - Graham J. Burton
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - D. Stephen Charnock-Jones
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge CB2 0SW, UK
| |
Collapse
|
136
|
Song Y, Yin D, Zhang Z, Chi L. Research progress of treatment of functional dyspepsia with traditional Chinese medicine compound based on cell signal pathway. Front Pharmacol 2023; 13:1089231. [PMID: 36699059 PMCID: PMC9868459 DOI: 10.3389/fphar.2022.1089231] [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: 11/04/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Functional dyspepsia (FD) is the most common clinical gastrointestinal disease, with complex and prolonged clinical symptoms. The prevalence of FD is increasing year by year, seriously affecting the quality of life of patients. The main causes of FD are related to abnormal gastrointestinal dynamics, increased visceral sensitivity, Helicobacter pylori (HP) infection, intestinal flora disturbance and psychological factors. A review of the relevant literature reveals that the mechanisms of traditional Chinese medicine (TCM) in the treatment of FD mainly involve the following pathways:5-HT signal pathway, AMPK signal pathway,C-kit signal pathway, CRF signal pathway, PERK signal pathway,NF-κB signal pathway. Based on a holistic concept, TCM promotes gastrointestinal motility, regulates visceral sensitivity and alleviates gastrointestinal inflammation through multiple signal pathways, reflecting the advantages of multi-level, multi-pathway and multi-targeted treatment of FD.
Collapse
Affiliation(s)
- Yujiao Song
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| | - Defei Yin
- Digestive System Department II, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| | - Zhenyi Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| | - Lili Chi
- Digestive System Department II, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China,*Correspondence: Yujiao Song, ; Defei Yin, ; Zhenyi Zhang, ; Lili Chi,
| |
Collapse
|
137
|
Bahamondes Lorca VA, Wu S. Ultraviolet Light, Unfolded Protein Response and Autophagy †. Photochem Photobiol 2023; 99:498-508. [PMID: 36591940 DOI: 10.1111/php.13777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/22/2022] [Indexed: 01/03/2023]
Abstract
The endoplasmic reticulum (ER) plays an important role in the regulation of protein synthesis. Alterations in the folding capacity of the ER induce stress, which activates three ER sensors that mediate the unfolded protein response (UPR). Components of the pathways regulated by these sensors have been shown to regulate autophagy. The last corresponds to a mechanism of self-eating and recycling important for proper cell maintenance. Ultraviolet radiation (UV) is an external damaging stimulus that is known for inducing oxidative stress, and DNA, lipid and protein damage. Many controversies exist regarding the role of UV-inducing ER stress or autophagy. However, a connection between the three of them has not been addressed. In this review, we will discuss the contradictory theories regarding the relationships between UV radiation with the induction of ER stress and autophagy, as well as hypothetic connections between UV, ER stress and autophagy.
Collapse
Affiliation(s)
- Verónica A Bahamondes Lorca
- Edison Biotechnology Institute, Ohio University, Athens, OH.,Departamento de Tecnología Médica, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Shiyong Wu
- Edison Biotechnology Institute, Ohio University, Athens, OH.,Department of Chemistry and Biochemistry, Ohio University, Athens, OH
| |
Collapse
|
138
|
Li W, Wang Y. Stress granules: potential therapeutic targets for infectious and inflammatory diseases. Front Immunol 2023; 14:1145346. [PMID: 37205103 PMCID: PMC10185834 DOI: 10.3389/fimmu.2023.1145346] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Eukaryotic cells are stimulated by external pressure such as that derived from heat shock, oxidative stress, nutrient deficiencies, or infections, which induce the formation of stress granules (SGs) that facilitates cellular adaptation to environmental pressures. As aggregated products of the translation initiation complex in the cytoplasm, SGs play important roles in cell gene expression and homeostasis. Infection induces SGs formation. Specifically, a pathogen that invades a host cell leverages the host cell translation machinery to complete the pathogen life cycle. In response, the host cell suspends translation, which leads to SGs formation, to resist pathogen invasion. This article reviews the production and function of SGs, the interaction between SGs and pathogens, and the relationship between SGs and pathogen-induced innate immunity to provide directions for further research into anti-infection and anti-inflammatory disease strategies.
Collapse
Affiliation(s)
- Wenyuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yao Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- *Correspondence: Yao Wang,
| |
Collapse
|
139
|
Celik C, Lee SYT, Yap WS, Thibault G. Endoplasmic reticulum stress and lipids in health and diseases. Prog Lipid Res 2023; 89:101198. [PMID: 36379317 DOI: 10.1016/j.plipres.2022.101198] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022]
Abstract
The endoplasmic reticulum (ER) is a complex and dynamic organelle that regulates many cellular pathways, including protein synthesis, protein quality control, and lipid synthesis. When one or multiple ER roles are dysregulated and saturated, the ER enters a stress state, which, in turn, activates the highly conserved unfolded protein response (UPR). By sensing the accumulation of unfolded proteins or lipid bilayer stress (LBS) at the ER, the UPR triggers pathways to restore ER homeostasis and eventually induces apoptosis if the stress remains unresolved. In recent years, it has emerged that the UPR works intimately with other cellular pathways to maintain lipid homeostasis at the ER, and so does at cellular levels. Lipid distribution, along with lipid anabolism and catabolism, are tightly regulated, in part, by the ER. Dysfunctional and overwhelmed lipid-related pathways, independently or in combination with ER stress, can have reciprocal effects on other cellular functions, contributing to the development of diseases. In this review, we summarize the current understanding of the UPR in response to proteotoxic stress and LBS and the breadth of the functions mitigated by the UPR in different tissues and in the context of diseases.
Collapse
Affiliation(s)
- Cenk Celik
- School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Wei Sheng Yap
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore.
| |
Collapse
|
140
|
Waad Sadiq Z, Brioli A, Al-Abdulla R, Çetin G, Schütt J, Murua Escobar H, Krüger E, Ebstein F. Immunogenic cell death triggered by impaired deubiquitination in multiple myeloma relies on dysregulated type I interferon signaling. Front Immunol 2023; 14:982720. [PMID: 36936919 PMCID: PMC10018035 DOI: 10.3389/fimmu.2023.982720] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 02/06/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Proteasome inhibition is first line therapy in multiple myeloma (MM). The immunological potential of cell death triggered by defects of the ubiquitin-proteasome system (UPS) and subsequent perturbations of protein homeostasis is, however, less well defined. Methods In this paper, we applied the protein homeostasis disruptors bortezomib (BTZ), ONX0914, RA190 and PR619 to various MM cell lines and primary patient samples to investigate their ability to induce immunogenic cell death (ICD). Results Our data show that while BTZ treatment triggers sterile type I interferon (IFN) responses, exposure of the cells to ONX0914 or RA190 was mostly immunologically silent. Interestingly, inhibition of protein de-ubiquitination by PR619 was associated with the acquisition of a strong type I IFN gene signature which relied on key components of the unfolded protein and integrated stress responses including inositol-requiring enzyme 1 (IRE1), protein kinase R (PKR) and general control nonderepressible 2 (GCN2). The immunological relevance of blocking de-ubiquitination in MM was further reflected by the ability of PR619-induced apoptotic cells to facilitate dendritic cell (DC) maturation via type I IFN-dependent mechanisms. Conclusion Altogether, our findings identify de-ubiquitination inhibition as a promising strategy for inducing ICD of MM to expand current available treatments.
Collapse
Affiliation(s)
- Zeinab Waad Sadiq
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Annamaria Brioli
- Klinik und Poliklinik für Innere Medizin C, Universitätsmedizin Greifswald, Greifswald, Germany
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Ruba Al-Abdulla
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Gonca Çetin
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Jacqueline Schütt
- Klinik und Poliklinik für Innere Medizin C, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Elke Krüger
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Frédéric Ebstein
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
141
|
Kawaguchi Y, Hagiwara D, Tsumura T, Miyata T, Kobayashi T, Sugiyama M, Onoue T, Yasuda Y, Iwama S, Suga H, Banno R, Grinevich V, Arima H. Knockdown of endoplasmic reticulum chaperone BiP leads to the death of parvocellular AVP/CRH neurons in mice. J Neuroendocrinol 2023; 35:e13223. [PMID: 36535753 DOI: 10.1111/jne.13223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022]
Abstract
Arginine vasopressin (AVP) is expressed in both magnocellular (magnAVP) and parvocellular AVP (parvAVP) neurons of the paraventricular nucleus, and AVP colocalizes with corticotropin-releasing hormone (CRH) only in the parvocellular neurons. The immunoglobulin heavy chain binding protein (BiP) is a major endoplasmic reticulum (ER) chaperone which regulates the unfolded protein response under ER stress. We previously demonstrated that knockdown of BiP in magnAVP neurons exacerbated ER stress, which resulted in the autophagy-associated cell death of magnAVP neurons. Using the same approach, in the present study we examined the role of BiP in mouse parvAVP/CRH neurons. Our data demonstrate that BiP is expressed in mouse parvAVP/CRH neurons under nonstress conditions and is upregulated in proportion to the increase in CRH expression after adrenalectomy. For BiP knockdown in parvAVP/CRH neurons, we utilized a viral approach in combination with shRNA interference. Knockdown of BiP expression induced ER stress in parvAVP/CRH neurons, as reflected by the expression of C/EBP homologous protein. Furthermore, BiP knockdown led to the loss of parvAVP/CRH neurons after 4 weeks. In summary, our results demonstrate that BiP plays a pivotal role in parvAVP/CRH neurons, which function as neuroendocrine cells producing a large number of secretory proteins.
Collapse
Affiliation(s)
- Yohei Kawaguchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuro Tsumura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Miyata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshinori Yasuda
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, Japan
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
142
|
Park G, Xu K, Chea L, Kim K, Safarta L, Song KH, Wu J, Park S, Min H, Hiramatsu N, Han J, Lin JH. Neurodegeneration risk factor, EIF2AK3 (PERK), influences tau protein aggregation. J Biol Chem 2022; 299:102821. [PMID: 36563857 PMCID: PMC9852698 DOI: 10.1016/j.jbc.2022.102821] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/12/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Tauopathies are neurodegenerative diseases caused by pathologic misfolded tau protein aggregation in the nervous system. Population studies implicate EIF2AK3 (eukaryotic translation initiation factor 2 alpha kinase 3), better known as PERK (protein kinase R-like endoplasmic reticulum kinase), as a genetic risk factor in several tauopathies. PERK is a key regulator of intracellular proteostatic mechanisms-unfolded protein response and integrated stress response. Previous studies found that tauopathy-associated PERK variants encoded functional hypomorphs with reduced signaling in vitro. But, it remained unclear how altered PERK activity led to tauopathy. Here, we chemically or genetically modulated PERK signaling in cell culture models of tau aggregation and found that PERK pathway activation prevented tau aggregation, whereas inhibition exacerbated tau aggregation. In primary tauopathy patient brain tissues, we found that reduced PERK signaling correlated with increased tau neuropathology. We found that tauopathy-associated PERK variants targeted the endoplasmic reticulum luminal domain; and two of these variants damaged hydrogen bond formation. Our studies support that PERK activity protects against tau aggregation and pathology. This may explain why people carrying hypomorphic PERK variants have increased risk for developing tauopathies. Finally, our studies identify small-molecule augmentation of PERK signaling as an attractive therapeutic strategy to treat tauopathies by preventing tau pathology.
Collapse
Affiliation(s)
- Goonho Park
- Department of Pathology, Stanford University, Palo Alto, California, USA,VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Ke Xu
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | - Leon Chea
- Department of Pathology, Stanford University, Palo Alto, California, USA,VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Kyle Kim
- Department of Pathology, Stanford University, Palo Alto, California, USA,VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Lance Safarta
- VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Keon-Hyoung Song
- Department of Pathology, Stanford University, Palo Alto, California, USA,VA Palo Alto Healthcare System, Palo Alto, California, USA,Department of Pharmaceutical Engineering, Soonchunhyang University, Asan, Republic of Korea
| | - Jian Wu
- Department of Pharmacology, UC San Diego, La Jolla, California, USA
| | - Soyoung Park
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Hyejung Min
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | - Nobuhiko Hiramatsu
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | - Jaeseok Han
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Jonathan H. Lin
- Department of Pathology, Stanford University, Palo Alto, California, USA,VA Palo Alto Healthcare System, Palo Alto, California, USA,For correspondence: Jonathan H. Lin
| |
Collapse
|
143
|
Yu W, Ilyas I, Hu X, Xu S, Yu H. Therapeutic potential of paeoniflorin in atherosclerosis: A cellular action and mechanism-based perspective. Front Immunol 2022; 13:1072007. [PMID: 36618414 PMCID: PMC9811007 DOI: 10.3389/fimmu.2022.1072007] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies have shown that the incidence, prevalence and mortality of atherosclerotic cardiovascular disease (ASCVD) are increasing globally. Atherosclerosis is characterized as a chronic inflammatory disease which involves inflammation and immune dysfunction. P. lactiflora Pall. is a plant origin traditional medicine that has been widely used for the treatment of various diseases for more than a millennium in China, Japan and Korean. Paeoniflorin is a bioactive monomer extracted from P. lactiflora Pall. with anti-atherosclerosis effects. In this article, we comprehensively reviewed the potential therapeutic effects and molecular mechanism whereby paeoniflorin protects against atherosclerosis from the unique angle of inflammation and immune-related pathway dysfunction in vascular endothelial cells, smooth muscle cells, monocytes, macrophages, platelets and mast cells. Paeoniflorin, with multiple protective effects in atherosclerosis, has the potential to be used as a promising therapeutic agent for the treatment of atherosclerosis and its complications. We conclude with a detailed discussion of the challenges and future perspective of paeoniflorin in translational cardiovascular medicine.
Collapse
Affiliation(s)
- Wei Yu
- School of Materials Science and Engineering, Hefei University of Technology, Hefei, Anhui, China,Center for Drug Research and Development, Anhui Renovo Pharmaceutical Co., Ltd, Center for Drug Research and Development, Hefei, Anhui, China
| | - Iqra Ilyas
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xuerui Hu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hui Yu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Materials Laminating Fabrication and Interfacial Controlling Technology, Hebei University of Technology, Tianjin, China,*Correspondence: Hui Yu,
| |
Collapse
|
144
|
Singh N, Romick-Rosendale L, Watanabe-Chailland M, Privette Vinnedge LM, Komurov K. Drug resistance mechanisms create targetable proteostatic vulnerabilities in Her2+ breast cancers. PLoS One 2022; 17:e0256788. [PMID: 36480552 PMCID: PMC9731458 DOI: 10.1371/journal.pone.0256788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/22/2022] [Indexed: 12/13/2022] Open
Abstract
Oncogenic kinase inhibitors show short-lived responses in the clinic due to high rate of acquired resistance. We previously showed that pharmacologically exploiting oncogene-induced proteotoxic stress can be a viable alternative to oncogene-targeted therapy. Here, we performed extensive analyses of the transcriptomic, metabolomic and proteostatic perturbations during the course of treatment of Her2+ breast cancer cells with a Her2 inhibitor covering the drug response, resistance, relapse and drug withdrawal phases. We found that acute Her2 inhibition, in addition to blocking mitogenic signaling, leads to significant decline in the glucose uptake, and shutdown of glycolysis and of global protein synthesis. During prolonged therapy, compensatory overexpression of Her3 allows for the reactivation of mitogenic signaling pathways, but fails to re-engage the glucose uptake and glycolysis, resulting in proteotoxic ER stress, which maintains the protein synthesis block and growth inhibition. Her3-mediated cell proliferation under ER stress during prolonged Her2 inhibition is enabled due to the overexpression of the eIF2 phosphatase GADD34, which uncouples protein synthesis block from the ER stress response to allow for active cell growth. We show that this imbalance in the mitogenic and proteostatic signaling created during the acquired resistance to anti-Her2 therapy imposes a specific vulnerability to the inhibition of the endoplasmic reticulum quality control machinery. The latter is more pronounced in the drug withdrawal phase, where the de-inhibition of Her2 creates an acute surge in the downstream signaling pathways and exacerbates the proteostatic imbalance. Therefore, the acquired resistance mechanisms to oncogenic kinase inhibitors may create secondary vulnerabilities that could be exploited in the clinic.
Collapse
Affiliation(s)
- Navneet Singh
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Lindsey Romick-Rosendale
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Miki Watanabe-Chailland
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Lisa M. Privette Vinnedge
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Kakajan Komurov
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| |
Collapse
|
145
|
Riesenberg BP, Hunt EG, Tennant MD, Hurst KE, Andrews AM, Leddy LR, Neskey DM, Hill EG, Rivera GOR, Paulos CM, Gao P, Thaxton JE. Stress-Mediated Attenuation of Translation Undermines T-cell Activity in Cancer. Cancer Res 2022; 82:4386-4399. [PMID: 36126165 PMCID: PMC9722626 DOI: 10.1158/0008-5472.can-22-1744] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/06/2022] [Accepted: 09/15/2022] [Indexed: 01/24/2023]
Abstract
Protein synthesis supports robust immune responses. Nutrient competition and global cell stressors in the tumor microenvironment (TME) may impact protein translation in T cells and antitumor immunity. Using human and mouse tumors, we demonstrated here that protein translation in T cells is repressed in solid tumors. Reduced glucose availability to T cells in the TME led to activation of the unfolded protein response (UPR) element eIF2α (eukaryotic translation initiation factor 2 alpha). Genetic mouse models revealed that translation attenuation mediated by activated p-eIF2α undermines the ability of T cells to suppress tumor growth. Reprograming T-cell metabolism was able to alleviate p-eIF2α accumulation and translational attenuation in the TME, allowing for sustained protein translation. Metabolic and pharmacological approaches showed that proteasome activity mitigates induction of p-eIF2α to support optimal antitumor T-cell function, protecting from translation attenuation and enabling prolonged cytokine synthesis in solid tumors. Together, these data identify a new therapeutic avenue to fuel the efficacy of tumor immunotherapy. SIGNIFICANCE Proteasome function is a necessary cellular component for endowing T cells with tumor killing capacity by mitigating translation attenuation resulting from the unfolded protein response induced by stress in the tumor microenvironment.
Collapse
Affiliation(s)
- Brian P. Riesenberg
- Immunotherapy Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill; Chapel Hill, NC 27514; USA
| | - Elizabeth G. Hunt
- Immunotherapy Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill; Chapel Hill, NC 27514; USA,Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill; Chapel Hill, NC 27514; USA
| | - Megan D. Tennant
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425; USA
| | - Katie E. Hurst
- Immunotherapy Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill; Chapel Hill, NC 27514; USA
| | - Alex M. Andrews
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; USA
| | - Lee R. Leddy
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; USA
| | - David M. Neskey
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; USA
| | - Elizabeth G. Hill
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; USA,Department of Public Health Sciences, Hollings Cancer Center Biostatistics Shared Resource; Director, Medical University of South Carolina, Charleston, SC 29425; USA
| | - Guillermo O. Rangel Rivera
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425; USA,Department of Surgery and Microbiology & Immunology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322; USA
| | - Chrystal M. Paulos
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425; USA,Department of Surgery and Microbiology & Immunology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322; USA
| | - Peng Gao
- Department of Medicine, Metabolomics Core Facility; Director, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611; USA
| | - Jessica E. Thaxton
- Immunotherapy Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill; Chapel Hill, NC 27514; USA,Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill; Chapel Hill, NC 27514; USA,Correspondence: Dr. Jessica Thaxton, Department of Cell Biology & Physiology, Immunotherapy Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC 27514, 919-966-4913,
| |
Collapse
|
146
|
Kamble VS, Pachpor TA, Khandagale SB, Wagh VV, Khare SP. Translation initiation and dysregulation of initiation factors in rare diseases. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
147
|
Cabral AJ, Costello DC, Farny NG. The enigma of ultraviolet radiation stress granules: Research challenges and new perspectives. Front Mol Biosci 2022; 9:1066650. [PMID: 36533077 PMCID: PMC9751325 DOI: 10.3389/fmolb.2022.1066650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/17/2022] [Indexed: 01/16/2025] Open
Abstract
Stress granules (SGs) are non-membrane bound cytoplasmic condensates that form in response to a variety of different stressors. Canonical SGs are thought to have a cytoprotective role, reallocating cellular resources during stress by activation of the integrated stress response (ISR) to inhibit translation and avoid apoptosis. However, different stresses result in compositionally distinct, non-canonical SG formation that is likely pro-apoptotic, though the exact function(s) of both SGs subtypes remain unclear. A unique non-canonical SG subtype is triggered upon exposure to ultraviolet (UV) radiation. While it is generally agreed that UV SGs are bona fide SGs due to their dependence upon the core SG nucleating protein Ras GTPase-activating protein-binding protein 1 (G3BP1), the localization of other key components of UV SGs are unknown or under debate. Further, the dynamics of UV SGs are not known, though unique properties such as cell cycle dependence have been observed. This Perspective compiles the available information on SG subtypes and on UV SGs in particular in an attempt to understand the formation, dynamics, and function of these mysterious stress-specific complexes. We identify key gaps in knowledge related to UV SGs, and examine the unique aspects of their formation. We propose that more thorough knowledge of the distinct properties of UV SGs will lead to new avenues of understanding of the function of SGs, as well as their roles in disease.
Collapse
Affiliation(s)
| | | | - Natalie G. Farny
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
148
|
de la Calle CM, Shee K, Yang H, Lonergan PE, Nguyen HG. The endoplasmic reticulum stress response in prostate cancer. Nat Rev Urol 2022; 19:708-726. [PMID: 36168057 DOI: 10.1038/s41585-022-00649-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/09/2022]
Abstract
In order to proliferate in unfavourable conditions, cancer cells can take advantage of the naturally occurring endoplasmic reticulum-associated unfolded protein response (UPR) via three highly conserved signalling arms: IRE1α, PERK and ATF6. All three arms of the UPR have key roles in every step of tumour progression: from cancer initiation to tumour growth, invasion, metastasis and resistance to therapy. At present, no cure for metastatic prostate cancer exists, as targeting the androgen receptor eventually results in treatment resistance. New research has uncovered an important role for the UPR in prostate cancer tumorigenesis and crosstalk between the UPR and androgen receptor signalling pathways. With an improved understanding of the mechanisms by which cancer cells exploit the endoplasmic reticulum stress response, targetable points of vulnerability can be uncovered.
Collapse
Affiliation(s)
- Claire M de la Calle
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin Shee
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Heiko Yang
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Peter E Lonergan
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, St. James's Hospital, Dublin, Ireland
- Department of Surgery, Trinity College, Dublin, Ireland
| | - Hao G Nguyen
- Department of Urology, Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
149
|
Martelli A, Omrani M, Zarghooni M, Citi V, Brogi S, Calderone V, Sureda A, Lorzadeh S, da Silva Rosa SC, Grabarek BO, Staszkiewicz R, Los MJ, Nabavi SF, Nabavi SM, Mehrbod P, Klionsky DJ, Ghavami S. New Visions on Natural Products and Cancer Therapy: Autophagy and Related Regulatory Pathways. Cancers (Basel) 2022; 14:5839. [PMID: 36497321 PMCID: PMC9738256 DOI: 10.3390/cancers14235839] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Macroautophagy (autophagy) has been a highly conserved process throughout evolution and allows cells to degrade aggregated/misfolded proteins, dysfunctional or superfluous organelles and damaged macromolecules, in order to recycle them for biosynthetic and/or energetic purposes to preserve cellular homeostasis and health. Changes in autophagy are indeed correlated with several pathological disorders such as neurodegenerative and cardiovascular diseases, infections, cancer and inflammatory diseases. Conversely, autophagy controls both apoptosis and the unfolded protein response (UPR) in the cells. Therefore, any changes in the autophagy pathway will affect both the UPR and apoptosis. Recent evidence has shown that several natural products can modulate (induce or inhibit) the autophagy pathway. Natural products may target different regulatory components of the autophagy pathway, including specific kinases or phosphatases. In this review, we evaluated ~100 natural compounds and plant species and their impact on different types of cancers via the autophagy pathway. We also discuss the impact of these compounds on the UPR and apoptosis via the autophagy pathway. A multitude of preclinical findings have shown the function of botanicals in regulating cell autophagy and its potential impact on cancer therapy; however, the number of related clinical trials to date remains low. In this regard, further pre-clinical and clinical studies are warranted to better clarify the utility of natural compounds and their modulatory effects on autophagy, as fine-tuning of autophagy could be translated into therapeutic applications for several cancers.
Collapse
Affiliation(s)
- Alma Martelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Marzieh Omrani
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983969411, Iran
| | - Maryam Zarghooni
- Department of Laboratory Medicine & Pathobiology, University of Toronto Alumna, Toronto, ON M5S 3J3, Canada
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Antoni Sureda
- Research Group in Community Nutrition, Oxidative Stress and Health Research Institute of the Balearic Islands (IdISBa), University of Balearic Islands, 07122 Palma de Mallorca, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Simone C. da Silva Rosa
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Beniamin Oscar Grabarek
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine in Zabrze, Academy of Silesia, 41-800 Zabrze, Poland
- Department of Gynaecology and Obstetrics, Faculty of Medicine in Zabrze, Academy of Silesia, 41-800 Zabrze, Poland
- GynCentrum, Laboratory of Molecular Biology and Virology, 40-851 Katowice, Poland
| | - Rafał Staszkiewicz
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine in Zabrze, Academy of Silesia, 41-800 Zabrze, Poland
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, 30-901 Krakow, Poland
| | - Marek J. Los
- Biotechnology Centre, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Seyed Fazel Nabavi
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite 62760-000, Brazil
| | - Seyed Mohammad Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre, Via Cortenocera, 82030 San Salvatore Telesino, Italy
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Faculty of Medicine in Zabrze, Academia of Silesia, 41-800 Zabrze, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
150
|
Recovery Sleep Immediately after Prolonged Sleep Deprivation Stimulates the Transcription of Integrated Stress Response-Related Genes in the Liver of Male Rats. Clocks Sleep 2022; 4:623-632. [PMID: 36412581 PMCID: PMC9680379 DOI: 10.3390/clockssleep4040048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
Sleep loss induces performance impairment and fatigue. The reactivation of human herpesvirus-6, which is related to the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α), is one candidate for use as an objective biomarker of fatigue. Phosphorylated eIF2α is a key regulator in integrated stress response (ISR), an intracellular stress response system. However, the relation between sleep/sleep loss and ISR is unclear. The purpose of the current study was to evaluate the effect of prolonged sleep deprivation and recovery sleep on ISR-related gene expression in rat liver. Eight-week-old male Sprague-Dawley rats were subjected to a 96-hour sleep deprivation using a flowerpot technique. The rats were sacrificed, and the liver was collected immediately or 6 or 72 h after the end of the sleep deprivation. RT-qPCR was used to analyze the expression levels of ISR-related gene transcripts in the rat liver. The transcript levels of the Atf3, Ddit3, Hmox-1, and Ppp15a1r genes were markedly increased early in the recovery sleep period after the termination of sleep deprivation. These results indicate that both activation and inactivation of ISRs in the rat liver occur simultaneously in the early phase of recovery sleep.
Collapse
|