1
|
Liu J, Chen K, Wu W, Pang Z, Zhu D, Yan X, Wang B, Qiu J, Fang Z. GRP78 exerts antiviral function against influenza A virus infection by activating the IFN/JAK-STAT signaling. Virology 2024; 600:110249. [PMID: 39303344 DOI: 10.1016/j.virol.2024.110249] [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: 03/18/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Influenza is an acute viral respiratory infection that causes mild to severe illness in humans and animals. Current studies show that glucose-regulated protein 78 (GRP78) can exert crucial functions during viral infection; however, the mechanism by which GRP78 regulates influenza A virus (IAV) infection remains unclear. In the present study, we found that IAV infection increased GRP78 expression. Overexpression of GRP78 significantly inhibited IAV replication, as indicated by reduced viral mRNA levels, protein levels, and viral titers. Mechanistically, Type I interferon (IFN) response signaling is upregulated during IAV infection by GRP78. Further study showed that GRP78 interacts with tyrosine kinase 2 (TYK2) and enhances its phosphorylation, thereby activating downstream STAT1/2 and antiviral IFN-stimulated gene (ISG) expression. Collectively, these results demonstrate an important mechanism by which GRP78 exerts in innate antiviral effect in IAV infection. This mechanism could be used as a therapeutic target for anti-influenza treatment.
Collapse
Affiliation(s)
- Jiaxin Liu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Kanghong Chen
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Wenjiao Wu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zefen Pang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Dandong Zhu
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Xiukui Yan
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Bangqi Wang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| | - Jianxiang Qiu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| | - Zhixin Fang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
2
|
Li Y, Li C, Zhao C, Wu J, Zhu Y, Wang F, Zhong J, Yan Y, Jin Y, Dong W, Chen J, Yang X, Zhou J, Hu B. Coronavirus M protein promotes mitophagy over virophagy by recruiting PDPK1 to phosphorylate SQSTM1 at T138. Nat Commun 2024; 15:8927. [PMID: 39414765 PMCID: PMC11484861 DOI: 10.1038/s41467-024-53100-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Autophagy plays a dual role in coronavirus infection, facilitating the elimination of either proviral components (virophagy) or antiviral factors such as mitochondria (mitophagy), leading to complex mechanisms of immune evasion. Understanding the mechanisms that govern the switch between the autophagic degradation of deleterious or beneficial substrates in coronavirus infection is crucial for developing precise drug targets to treat virus-induced diseases. However, this switch remains largely unknown. Using a dual split-fluorescence assay, we identify PDPK1 as a negative regulator of innate immunity, directing the transition from virophagy to mitophagy through the phosphorylation of SQSTM1 at T138. Remarkably, a PDPK1-targeting peptide inhibits the replication of various RNA viruses by restoring innate immunity through enhanced virophagy and suppressed mitophagy, thereby protecting female mice from lethal infections. These findings underscore the detrimental role of PDPK1 in innate immunity by orchestrating the shift from virophagy to mitophagy, positioning PDPK1 as a promising pharmacological target for effectively combating a broad spectrum of virus infections.
Collapse
Affiliation(s)
- Yahui Li
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Chunyan Li
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Chenchen Zhao
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Jiayu Wu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Ya Zhu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Fei Wang
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Jiepeng Zhong
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yan Yan
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yulan Jin
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Weiren Dong
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Jinyang Chen
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Xianghong Yang
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University, Hangzhou, China.
| | - Boli Hu
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China.
| |
Collapse
|
3
|
Lallai V, Lam TT, Garcia-Milian R, Chen YC, Fowler JP, Manca L, Piomelli D, Williams K, Nairn AC, Fowler CD. Proteomic Profile of Circulating Extracellular Vesicles in the Brain after Δ9-Tetrahydrocannabinol Inhalation. Biomolecules 2024; 14:1143. [PMID: 39334909 PMCID: PMC11430348 DOI: 10.3390/biom14091143] [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: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Given the increasing use of cannabis in the US, there is an urgent need to better understand the drug's effects on central signaling mechanisms. Extracellular vesicles (EVs) have been identified as intercellular signaling mediators that contain a variety of cargo, including proteins. Here, we examined whether the main psychoactive component in cannabis, Δ9-tetrahydrocannabinol (THC), alters EV protein signaling dynamics in the brain. We first conducted in vitro studies, which found that THC activates signaling in choroid plexus epithelial cells, resulting in transcriptional upregulation of the cannabinoid 1 receptor and immediate early gene c-fos, in addition to the release of EVs containing RNA cargo. Next, male and female rats were examined for the effects of either acute or chronic exposure to aerosolized ('vaped') THC on circulating brain EVs. Cerebrospinal fluid was extracted from the brain, and EVs were isolated and processed with label-free quantitative proteomic analyses via high-resolution tandem mass spectrometry. Interestingly, circulating EV-localized proteins were differentially expressed based on acute or chronic THC exposure in a sex-specific manner. Taken together, these findings reveal that THC acts in the brain to modulate circulating EV signaling, thereby providing a novel understanding of how exogenous factors can regulate intercellular communication in the brain.
Collapse
Affiliation(s)
- Valeria Lallai
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
| | - TuKiet T. Lam
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT 06511, USA
| | - Rolando Garcia-Milian
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Bioinformatics Support Hub, Harvey Cushing/John Whitney Medical Library, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yen-Chu Chen
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - James P. Fowler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - Letizia Manca
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - Daniele Piomelli
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA;
| | - Kenneth Williams
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Angus C. Nairn
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA
| | - Christie D. Fowler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
| |
Collapse
|
4
|
Monaghan RM, Naylor RW, Flatman D, Kasher PR, Williams SG, Keavney BD. FLT4 causes developmental disorders of the cardiovascular and lymphovascular systems via pleiotropic molecular mechanisms. Cardiovasc Res 2024; 120:1164-1176. [PMID: 38713105 PMCID: PMC11368125 DOI: 10.1093/cvr/cvae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 05/08/2024] Open
Abstract
AIMS Rare, deleterious genetic variants in FLT4 are associated with Tetralogy of Fallot (TOF), the most common cyanotic congenital heart disease. The distinct genetic variants in FLT4 are also an established cause of Milroy disease, the most prevalent form of primary hereditary lymphoedema. The phenotypic features of these two conditions are non-overlapping, implying pleiotropic cellular mechanisms during development. METHODS AND RESULTS In this study, we show that FLT4 variants identified in patients with TOF, when expressed in primary human endothelial cells, cause aggregation of FLT4 protein in the perinuclear endoplasmic reticulum, activating proteostatic and metabolic signalling, whereas lymphoedema-associated FLT4 variants and wild-type (WT) FLT4 do not. FLT4 TOF variants display characteristic gene expression profiles in key developmental signalling pathways, revealing a role for FLT4 in cardiogenesis distinct from its role in lymphatic development. Inhibition of proteostatic signalling abrogates these effects, identifying potential avenues for therapeutic intervention. Depletion of flt4 in zebrafish caused cardiac phenotypes of reduced heart size and altered heart looping. These phenotypes were rescued with coinjection of WT human FLT4 mRNA, but incompletely or not at all by mRNA harbouring FLT4 TOF variants. CONCLUSION Taken together, we identify a pathogenic mechanism for FLT4 variants predisposing to TOF that is distinct from the known dominant negative mechanism of Milroy-causative variants. FLT4 variants give rise to conditions of the two circulatory subdivisions of the vascular system via distinct developmental pleiotropic molecular mechanisms.
Collapse
Affiliation(s)
- Richard M Monaghan
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, 5th Floor, AV Hill Building, Oxford Road, Manchester, M13 9NT, UK
| | - Richard W Naylor
- Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PN, UK
| | - Daisy Flatman
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Paul R Kasher
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Simon G Williams
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, 5th Floor, AV Hill Building, Oxford Road, Manchester, M13 9NT, UK
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, 5th Floor, AV Hill Building, Oxford Road, Manchester, M13 9NT, UK
- Manchester Heart Institute, Manchester University NHS Foundation Trust, Oxford Road, M13 9WL, UK
| |
Collapse
|
5
|
Silva NSM, Siebeneichler B, Oliveira CS, Dores-Silva PR, Borges JC. The regulation of the thermal stability and affinity of the HSPA5 (Grp78/BiP) by clients and nucleotides is modulated by domains coupling. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1872:141034. [PMID: 39009203 DOI: 10.1016/j.bbapap.2024.141034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
The HSPA5 protein (BiP/Grp78) serves as a pivotal chaperone in maintaining cellular protein quality control. As a member of the human HSP70 family, HSPA5 comprises two distinct domains: a nucleotide-binding domain (NBD) and a peptide-binding domain (PBD). In this study, we investigated the interdomain interactions of HSPA5, aiming to elucidate how these domains regulate its function as a chaperone. Our findings revealed that HSPA5-FL, HSPA5-T, and HSPA5-N exhibit varying affinities for ATP and ADP, with a noticeable dependency on Mg2+ for optimal interactions. Interestingly, in ADP assays, the presence of the metal ion seems to enhance NBD binding only for HSPA5-FL and HSPA5-T. Moreover, while the truncation of the C-terminus does not significantly impact the thermal stability of HSPA5, experiments involving MgATP underscore its essential role in mediating interactions and nucleotide hydrolysis. Thermal stability assays further suggested that the NBD-PBD interface enhances the stability of the NBD, more pronounced for HSPA5 than for the orthologous HSPA1A, and prevents self-aggregation through interdomain coupling. Enzymatic analyses indicated that the presence of PBD enhances NBD ATPase activity and augments its nucleotide affinity. Notably, the intrinsic chaperone activity of the PBD is dependent on the presence of the NBD, potentially due to the propensity of the PBD for self-oligomerization. Collectively, our data highlight the pivotal role of allosteric mechanisms in modulating thermal stability, nucleotide interaction, and ATPase activity of HSPA5, underscoring its significance in protein quality control within cellular environments.
Collapse
Affiliation(s)
- Noeli S M Silva
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, SP, Brazil.
| | - Bruna Siebeneichler
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, SP, Brazil; Exact and Technology Sciences Center, Federal University of São Carlos, São Carlos, SP 13560-970, Brazil
| | - Carlos S Oliveira
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, SP, Brazil
| | - Paulo R Dores-Silva
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, SP, Brazil
| | - Júlio C Borges
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, SP, Brazil.
| |
Collapse
|
6
|
Johnson N, Pattinson C, Burgoyne K, Hijazi K, Houssen WE, Milne BF. SARS-CoV-2 Spike Protein-Derived Cyclic Peptides as Modulators of Spike Interaction with GRP78. Chembiochem 2024; 25:e202300789. [PMID: 38613462 PMCID: PMC11497264 DOI: 10.1002/cbic.202300789] [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: 11/22/2023] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/15/2024]
Abstract
The human glucose-regulated protein GRP78 is a human chaperone that translocactes to the cell surface when cells are under stress. Theoretical studies suggested it could be involved in SARS-CoV-2 virus entry to cells. In this work, we used in vitro surface plasmon resonance-based assays to show that human GRP78 indeed binds to SARS-CoV-2 spike protein. We have designed and synthesised cyclic peptides based on the loop structure of amino acids 480-488 of the SARS-CoV-2 spike protein S1 domain from the Wuhan and Omicron variants and showed that both peptides bind to GRP78. Consistent with the greater infectiousness of the Omicron variant, the Omicron-derived peptide displays slower dissociation from the target protein. Both peptides significantly inhibit the binding of wild-type S1 protein to the human protein GRP78 suggesting that further development of these cyclic peptide motifs may provide a viable route to novel anti-SARS-CoV-2 agents.
Collapse
Affiliation(s)
- Nicholas Johnson
- Institute of Medical SciencesUniversity of AberdeenAshgrove Road WestAberdeenAB25 2ZDUK
| | - Craig Pattinson
- School of MedicineMedical Sciences and NutritionUniversity of AberdeenAberdeenAB25 2ZDUK
| | - Kate Burgoyne
- School of MedicineMedical Sciences and NutritionUniversity of AberdeenAberdeenAB25 2ZDUK
| | - Karolin Hijazi
- School of MedicineMedical Sciences and NutritionUniversity of AberdeenAberdeenAB25 2ZDUK
| | - Wael E. Houssen
- Institute of Medical SciencesUniversity of AberdeenAshgrove Road WestAberdeenAB25 2ZDUK
- Department of ChemistryUniversity of AberdeenMeston WalkAberdeenAB24 3UEUK
| | - Bruce F. Milne
- Department of ChemistryUniversity of AberdeenMeston WalkAberdeenAB24 3UEUK
- CFisUCDepartment of PhysicsUniversity of CoimbraRua Larga3004-516CoimbraPortugal
| |
Collapse
|
7
|
Xiong Z, Wang Q, Pei S, Zheng H, Wang W. CircRNA_0088196 Regulates Trophoblast Proliferation and Apoptosis in Preeclampsia Through the miR-379-5p/HSPA5 Axis. Biochem Genet 2024; 62:1742-1761. [PMID: 37698761 DOI: 10.1007/s10528-023-10506-6] [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: 06/05/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023]
Abstract
Existing research has confirmed the dysregulation of circular RNA (circRNA) in a wide variety of human diseases. Thus, in this study, we explored the potential mechanism of circRNA_0088196 in preeclampsia (PE). We performed quantitative real-time PCR to examine circRNA_0088196 expression and verified the function of circRNA_0088196 in vitro using CCK-8, TUNEL, flow cytometry, and Western blotting analyses. Additionally, we studied the mechanism using dual-luciferase reporter gene experiments. The results of our research revealed the up-regulation of circRNA_0088196 in PE patients' placentas and Heat Shock 70 kDa Protein 5 (HSPA5)-stimulated trophoblast (HTR-8/SVneo) cells. An investigation of the mechanism also showed that there was a binding between miR-379-5p and circRNA_0088196. Additionally, circRNA_0088196 inhibited HTR-8/SVneo cell proliferation and promoted cell apoptosis via the miR-337-3p/HSPA5 axis, thereby facilitating PE. In vivo experiments indicated that circRNA_0088196 regulated HTR-8/SVneo cell production through miR-379-5p. Overall, the findings of this study illustrate that circRNA_0088196 interference promotes cell apoptosis and inhibits HTR-8/SVneo proliferation via the miR-379-5p/HSPA5 axis, thereby accelerating the development of PE.
Collapse
Affiliation(s)
- Zhihui Xiong
- Department of Obstetrics, Tongde Hospital of Zhejiang Provincial, Hangzhou, 310012, China
| | - Qingmin Wang
- Department of Obstetrics, Tongde Hospital of Zhejiang Provincial, Hangzhou, 310012, China
| | - Shuping Pei
- Department of Obstetrics, Tongde Hospital of Zhejiang Provincial, Hangzhou, 310012, China
| | - Huiling Zheng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, 310005, China.
| | - Wei Wang
- Department of Obstetrics, Tongde Hospital of Zhejiang Provincial, Hangzhou, 310012, China.
| |
Collapse
|
8
|
Han C, Xie Z, Lv Y, Liu D, Chen R. Direct interaction of the molecular chaperone GRP78/BiP with the Newcastle disease virus hemagglutinin-neuraminidase protein plays a vital role in viral attachment to and infection of culture cells. Front Immunol 2023; 14:1259237. [PMID: 37920471 PMCID: PMC10619984 DOI: 10.3389/fimmu.2023.1259237] [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: 07/15/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023] Open
Abstract
Introduction Glucose Regulated Proteins/Binding protein (GRP78/Bip), a representative molecular chaperone, effectively influences and actively participates in the replication processes of many viruses. Little is known, however, about the functional involvement of GRP78 in the replication of Newcastle disease virus (NDV) and the underlying mechanisms. Methods The method of this study are to establish protein interactomes between host cell proteins and the NDV Hemagglutinin-neuraminidase (HN) protein, and to systematically investigate the regulatory role of the GRP78-HN protein interaction during the NDV replication cycle. Results Our study revealed that GRP78 is upregulated during NDV infection, and its direct interaction with HN is mediated by the N-terminal 326 amino acid region. Knockdown of GRP78 by small interfering RNAs (siRNAs) significantly suppressed NDV infection and replication. Conversely, overexpression of GRP78 resulted in a significant increase in NDV replication, demonstrating its role as a positive regulator in the NDV replication cycle. We further showed that the direct interaction between GRP78 and HN protein enhanced the attachment of NDV to cells, and masking of GRP78 expressed on the cell surface with specific polyclonal antibodies (pAbs) inhibited NDV attachment and replication. Discussion These findings highlight the essential role of GRP78 in the adsorption stage during the NDV infection cycle, and, importantly, identify the critical domain required for GRP78-HN interaction, providing novel insights into the molecular mechanisms involved in NDV replication and infection.
Collapse
Affiliation(s)
- Chenxin Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Ziwei Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Yadi Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| | - Dingxiang Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
- Integrative Microbiology Research Centre, South China Agricultural University, Guangzhou, China
| | - Ruiai Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Zhaoqing Branch Centre of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, China
| |
Collapse
|
9
|
Shin J, Shimomura I. COVID-19, Obesity, and GRP78: Unraveling the Pathological Link. J Obes Metab Syndr 2023; 32:183-196. [PMID: 37752707 PMCID: PMC10583770 DOI: 10.7570/jomes23053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/16/2023] [Accepted: 09/22/2023] [Indexed: 09/28/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, driven by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to an unprecedented global surge in infections and fatalities. Notably, obesity has emerged as an important susceptibility factor for COVID-19; however, the pathological mechanisms for this remain poorly understood. Recent studies proposed a role for glucose-regulated protein 78 (GRP78), a protein implicated in both obesity and metabolic syndrome, which may function as a binding partner and/or co-receptor for SARS-CoV-2. Given its crucial involvement in diverse biological processes, GRP78 likely plays a major role in multiple facets of the viral life cycle and the pathology of COVID-19. This perspective review discusses the potential contributions of GRP78 to the dynamics of SARS-CoV-2 infection and pathology, particularly in the context of obesity. The primary objective is to facilitate a deeper understanding of the pathogenesis of COVID-19. Through this exploration, we aim to illuminate the complex interactions underpinning the nexus of COVID-19, obesity, and GRP78, ultimately paving the way for informed therapeutic strategies and preventive measures.
Collapse
Affiliation(s)
- Jihoon Shin
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
10
|
Li T, Fu J, Cheng J, Elfiky AA, Wei C, Fu J. New progresses on cell surface protein HSPA5/BiP/GRP78 in cancers and COVID-19. Front Immunol 2023; 14:1166680. [PMID: 37275848 PMCID: PMC10232979 DOI: 10.3389/fimmu.2023.1166680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/10/2023] [Indexed: 06/07/2023] Open
Abstract
Heat-shock-protein family A (Hsp70) member 5 (HSPA5), aliases GRP78 or BiP, is a protein encoded with 654 amino acids by the HSPA5 gene located on human chromosome 9q33.3. When the endoplasmic reticulum (ER) was stressed, HSPA5 translocated to the cell surface, the mitochondria, and the nucleus complexed with other proteins to execute its functions. On the cell surface, HSPA5/BiP/GRP78 can play diverse functional roles in cell viability, proliferation, apoptosis, attachments, and innate and adaptive immunity regulations, which lead to various diseases, including cancers and coronavirus disease 2019 (COVID-19). COVID-19 is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, which caused the pandemic since the first outbreak in late December 2019. HSPA5, highly expressed in the malignant tumors, likely plays a critical role in SARS-CoV-2 invasion/attack in cancer patients via tumor tissues. In the current study, we review the newest research progresses on cell surface protein HSPA5 expressions, functions, and mechanisms for cancers and SARS-CoV-2 invasion. The therapeutic and prognostic significances and prospects in cancers and COVID-19 disease by targeting HSPA5 are also discussed. Targeting HSPA5 expression by natural products may imply the significance in clinical for both anti-COVID-19 and anti-cancers in the future.
Collapse
Affiliation(s)
- Ting Li
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jiewen Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Abdo A. Elfiky
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Chunli Wei
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
11
|
Rehati A, Abuduaini B, Liang Z, Chen D, He F. Identification of heat shock protein family A member 5 (HSPA5) targets involved in nonalcoholic fatty liver disease. Genes Immun 2023:10.1038/s41435-023-00205-y. [PMID: 37156995 DOI: 10.1038/s41435-023-00205-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/27/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
Heat shock protein family A (Hsp70) member 5 (HSPA5) is an endoplasmic reticulum chaperone, which regulates cell metabolism, particularly lipid metabolism. While HSPA5's role in regulating cell function is well described, HSPA5 binding to RNA and its biological function in nonalcoholic fatty liver disease (NAFLD) is still lacking. In the present study, the ability of HSPA5 to modulate alternative splicing (AS) of cellular genes was assessed using Real-Time PCR on 89 NAFLD-associated genes. RNA immunoprecipitation coupled to RNA sequencing (RIP-Seq) assays were also performed to identify cellular mRNAs bound by HSPA5. We obtained the HSPA5-bound RNA profile in HeLa cells and peak calling analysis revealed that HSPA5 binds to coding genes and lncRNAs. Moreover, RIP-Seq assays demonstrated that HSPA5 immunoprecipitates specific cellular mRNAs such as EGFR, NEAT1, LRP1 and TGFß1, which are important in the pathology of NAFLD. Finally, HSPA5 binding sites may be associated with splicing sites. We used the HOMER algorithm to search for motifs enriched in coding sequence (CDs) peaks, which identified over-representation of the AGAG motif in both sets of immunoprecipitated peaks. HSPA5 regulated genes at the 5'UTR alternative splicing and introns and in an AG-rich sequence-dependent manner. We propose that the HSPA5-AGAG interaction might play an important role in regulating alternative splicing of NAFLD-related genes. This report is the first to demonstrate that HSPA5 regulated pre-RNA alternative splicing, stability, or translation and affected target protein(s) via binding to lncRNA and mRNA linked to NAFLD.
Collapse
Affiliation(s)
- Aliya Rehati
- Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Buzukela Abuduaini
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University, 393 South Li Yu Shan Road, Urumqi, 830054, Xinjiang, China.
| | - Zhao Liang
- Department of General Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Dong Chen
- ABLife BioBigData Institute, Wuhan, 430075, Hubei, China
| | - Fangping He
- Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China.
| |
Collapse
|
12
|
Trinh QD, Pham NTK, Takada K, Ushijima H, Komine-Aizawa S, Hayakawa S. Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives. Int J Mol Sci 2023; 24:ijms24076489. [PMID: 37047462 PMCID: PMC10095195 DOI: 10.3390/ijms24076489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) is a pleiotropic growth factor playing various roles in the human body including cell growth and development. More functions of TGF-β1 have been discovered, especially its roles in viral infection. TGF-β1 is abundant at the maternal-fetal interface during pregnancy and plays an important function in immune tolerance, an essential key factor for pregnancy success. It plays some critical roles in viral infection in pregnancy, such as its effects on the infection and replication of human cytomegalovirus in syncytiotrophoblasts. Interestingly, its role in the enhancement of Zika virus (ZIKV) infection and replication in first-trimester trophoblasts has recently been reported. The above up-to-date findings have opened one of the promising approaches to studying the mechanisms of viral infection during pregnancy with links to corresponding congenital syndromes. In this article, we review our current and recent advances in understanding the roles of TGF-β1 in viral infection. Our discussion focuses on viral infection during pregnancy, especially in the first trimester. We highlight the mutual roles of viral infection and TGF-β1 in specific contexts and possible functions of the Smad pathway in viral infection, with a special note on ZIKV infection. In addition, we discuss promising approaches to performing further studies on this topic.
Collapse
Affiliation(s)
- Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
13
|
Lee D, Lee Y, Hye Shin S, Min Choi S, Hyeon Lee S, Jeong S, Jang S, Kee JM. A simple protein histidine kinase activity assay for high-throughput inhibitor screening. Bioorg Chem 2023; 130:106232. [DOI: 10.1016/j.bioorg.2022.106232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022]
|
14
|
Ha DP, Huang B, Wang H, Rangel DF, Van Krieken R, Liu Z, Samanta S, Neamati N, Lee AS. Targeting GRP78 suppresses oncogenic KRAS protein expression and reduces viability of cancer cells bearing various KRAS mutations. Neoplasia 2022; 33:100837. [PMID: 36162331 PMCID: PMC9516447 DOI: 10.1016/j.neo.2022.100837] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/01/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022]
Abstract
KRAS is the most commonly mutated oncogene in human cancers with limited therapeutic options, thus there is a critical need to identify novel targets and inhibiting agents. The 78-kDa glucose-regulated protein GRP78, which is upregulated in KRAS cancers, is an essential chaperone and the master regulator of the unfolded protein response (UPR). Following up on our recent discoveries that GRP78 haploinsufficiency suppresses both KRASG12D-driven pancreatic and lung tumorigenesis, we seek to determine the underlying mechanisms. Here, we report that knockdown of GRP78 via siRNA reduced oncogenic KRAS protein level in human lung, colon, and pancreatic cancer cells bearing various KRAS mutations. This effect was at the post-transcriptional level and is independent of proteasomal degradation or autophagy. Moreover, targeting GRP78 via small molecule inhibitors such as HA15 and YUM70 with anti-cancer activities while sparing normal cells significantly suppressed oncogenic KRAS expression in vitro and in vivo, associating with onset of apoptosis and loss of viability in cancer cells bearing various KRAS mutations. Collectively, our studies reveal that GRP78 is a previously unidentified regulator of oncogenic KRAS expression, and, as such, augments the other anti-cancer activities of GRP78 small molecule inhibitors to potentially achieve general, long-term suppression of mutant KRAS-driven tumorigenesis.
Collapse
Affiliation(s)
- Dat P Ha
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center
| | - Bo Huang
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center
| | - Han Wang
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center
| | - Daisy Flores Rangel
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center
| | - Richard Van Krieken
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center
| | - Ze Liu
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center
| | - Soma Samanta
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA; USC Norris Comprehensive Cancer Center.
| |
Collapse
|
15
|
Dent P, Booth L, Roberts JL, Poklepovic A, Martinez J, Cridebring D, Reiman EM. AR12 increases BAG3 expression which is essential for Tau and APP degradation via LC3-associated phagocytosis and macroautophagy. Aging (Albany NY) 2022; 14:8221-8242. [PMID: 36227739 PMCID: PMC9648812 DOI: 10.18632/aging.204337] [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: 08/17/2022] [Accepted: 10/05/2022] [Indexed: 11/26/2022]
Abstract
We defined the mechanisms by which the chaperone ATPase inhibitor AR12 and the multi-kinase inhibitor neratinib interacted to reduce expression of Tau and amyloid-precursor protein (APP) in microglia and neuronal cells. AR12 and neratinib interacted to increase the phosphorylation of eIF2A S51 and the expression of BAG3, Beclin1 and ATG5, and in parallel, enhanced autophagosome formation and autophagic flux. Knock down of BAG3, Beclin1 or ATG5 abolished autophagosome formation and significantly reduced degradation of p62, LAMP2, Tau, APP, and GRP78 (total and plasma membrane). Knock down of Rubicon, a key component of LC3-associated phagocytosis (LAP), significantly reduced autophagosome formation but not autophagic flux and prevented degradation of Tau, APP, and cell surface GRP78, but not ER-localized GRP78. Knock down of Beclin1, ATG5 or Rubicon or over-expression of GRP78 prevented the significant increase in eIF2A phosphorylation. Knock down of eIF2A prevented the increase in BAG3 expression and significantly reduced autophagosome formation, autophagic flux, and it prevented Tau and APP degradation. We conclude that AR12 has the potential to reduce Tau and APP levels in neurons and microglia via the actions of LAP, endoplasmic reticulum stress signaling and macroautophagy. We hypothesize that the initial inactivation of GRP78 catalytic function by AR12 facilitates an initial increase in eIF2A phosphorylation which in turn is essential for greater levels of eIF2A phosphorylation, greater levels of BAG3 and macroautophagy and eventually leading to significant amounts of APP/Tau degradation.
Collapse
Affiliation(s)
- Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jane L Roberts
- Department of Surgery, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jennifer Martinez
- National Institute of Environmental Health Sciences, Inflammation and Autoimmunity Group, Triangle Park, Durham, NC 27709, USA
| | - Derek Cridebring
- Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA
| | - Eric M Reiman
- Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA.,Banner Alzheimer's Institute, Phoenix, AZ 85006, USA
| |
Collapse
|
16
|
Cui X, Wang L, Lü Y, Yue C. Development and research progress of anti-drug resistant fungal drugs. J Infect Public Health 2022; 15:986-1000. [PMID: 35981408 DOI: 10.1016/j.jiph.2022.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022] Open
Abstract
With the widespread use of immunosuppressive agents and the increase in patients with severe infections, the incidence of fungal infections worldwide has increased year by year. The fungal pathogens Candida albicans, Cryptococcus neoformans and Aspergillus fumigatus cause a total of more than 1 million deaths each year. Long-term use of antifungal drugs can easily lead to fungal resistance, and the prevalence of drug-resistant fungi is a major global health challenge. In order to effectively control global fungal infections, there is an urgent need for new drugs that can exert effective antifungal activity and overcome drug resistance. We must promote the discovery of new antifungal targets and drugs, and find effective ways to control drug-resistant fungi through different ways, so as to reduce the threat of drug-resistant fungi to human life, health and safety. In the past few years, certain progress has been made in the research and development of antifungal drugs. In addition to summarizing some of the antifungal drugs currently approved by the FDA, this review also focuses on potential antifungal drugs, the repositioned drugs, and drugs that can treat drug-resistant bacteria and fungal infections, and provide new ideas for the development of antifungal drugs in the future.
Collapse
Affiliation(s)
- Xiangyi Cui
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Lanlin Wang
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Yuhong Lü
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| | - Changwu Yue
- Key Laboratory of Microbial Drugs Innovation and Transformation of Yan'an, School of Basic Medicine, Yan'an University, Yan'an 716000, Shaanxi, China; Shaanxi Engineering & Technological Research Center for Conversation & Utilization of Regional Biological Resources,Yan'an University, NO.580 Shengdi Road, Baota District, Yan'an 716000, Shaanxi, China.
| |
Collapse
|
17
|
Shin J, Toyoda S, Fukuhara A, Shimomura I. GRP78, a Novel Host Factor for SARS-CoV-2: The Emerging Roles in COVID-19 Related to Metabolic Risk Factors. Biomedicines 2022; 10:biomedicines10081995. [PMID: 36009544 PMCID: PMC9406123 DOI: 10.3390/biomedicines10081995] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 11/28/2022] Open
Abstract
The outbreak of coronavirus disease 19 (COVID-19), caused by the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in an unprecedented amount of infection cases and deaths, leading to the global health crisis. Despite many research efforts, our understanding of COVID-19 remains elusive. Recent studies have suggested that cell surface glucose-regulated protein 78 (GRP78) acts as a host co-receptor for SARS-CoV-2 infection and is related to COVID-19 risks, such as older age, obesity, and diabetes. Given its significance in a wide range of biological processes, such as protein homeostasis and cellular signaling, GRP78 might also play an important role in various stages of the viral life cycle and pathology of SARS-CoV-2. In this perspective, we explore the emerging and potential roles of GRP78 in SARS-CoV-2 infection. Additionally, we discuss the association with COVID-19 risks and symptoms. We hope this review article will be helpful to understand COVID-19 pathology and promote attention and study of GRP78 from many clinical and basic research fields.
Collapse
Affiliation(s)
- Jihoon Shin
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Diabetes Care Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Correspondence:
| | - Shinichiro Toyoda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsunori Fukuhara
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Department of Adipose Management, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
18
|
Sadeghi N, Tavalaee M, Shahverdi A, Sengupta P, Leisegang K, Saleh R, Agarwal A, Nasr-Esfahani MH. Vulnerability of The Male Reproductive System to SARS-CoV-2 Invasion: Potential Role for The Endoplasmic Reticulum Chaperone Grp78/HSPA5/BiP. CELL JOURNAL 2022; 24:427-433. [PMID: 36093801 PMCID: PMC9468718 DOI: 10.22074/cellj.2022.8312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Indexed: 11/04/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) may adversely affect male reproductive tissues and male<br />fertility. This concern is elicited by the higher susceptibility and mortality rate of men to the SARS-CoV-2 mediated coronavirus disease-19 (COVID-19), compared to the women. SARS-CoV-2 enters host cells after binding to a functional receptor named angiotensin-converting enzyme-2 (ACE2) and then replicates in the host cells and gets released into the plasma. SARS-CoVs use the endoplasmic reticulum (ER) as a site for viral protein synthesis and processing, as well as glucose-regulated protein 78 (Grp78) is a key ER chaperone involved in protein folding by preventing newly synthesized proteins from aggregation.<br />Therefore, we analyzed Grp78 expression in various human organs, particularly male reproductive organs, using Broad<br />Institute Cancer Cell Line Encyclopedia (CCLE), the Genotype-Tissue Expression (GTEx), and Human Protein Atlas online<br />datasets. Grp78 is expressed in male reproductive tissues such as the testis, epididymis, prostate, and seminal vesicle. It can facilitate the coronavirus entry into the male reproductive tract, providing an opportunity for its replication. This link between the SARS-CoV-2 and the Grp78 protein could become a therapeutic target to mitigate its harmful effects on male fertility.
Collapse
Affiliation(s)
- Niloofar Sadeghi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan,
Iran,Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Marziyeh Tavalaee
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan,
Iran
| | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR,
Tehran, Iran
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Bandar Saujana Putra, Malaysia
| | - Kristian Leisegang
- School of Natural Medicine, Faculty of Community and Health Sciences, University of the Western Cape, Cape Town, South Africa
| | - Ramadan Saleh
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag, Egypt,Ajyal IVF Center, Ajyal Hospital, Sohag, Egypt
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA,P.O.Box: 8165131378Department of Animal BiotechnologyReproductive Biomedicine Research CenterRoyan Institute for BiotechnologyACECRIsfahanIran
Emails:,
| | - Mohammad Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan,
Iran,P.O.Box: 8165131378Department of Animal BiotechnologyReproductive Biomedicine Research CenterRoyan Institute for BiotechnologyACECRIsfahanIran
Emails:,
| |
Collapse
|
19
|
HSPA5 Inhibitor Meliorate DSS-Induced Colitis through HSPA1A/CHIP. DISEASE MARKERS 2022; 2022:7115181. [PMID: 35872700 PMCID: PMC9300310 DOI: 10.1155/2022/7115181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/27/2022] [Indexed: 12/01/2022]
Abstract
Objective Ulcerative colitis (UC) is closely related to immune response, in which Treg cells (Tregs) suppress the autoimmune response of effector T cells to maintain homeostasis. As a marker of endoplasmic reticulum stress (ERS), HSPA5 was highly expressed in the colon tissue of UC patients. This study is aimed at evaluating the therapeutic effect of HSPA5 inhibitor (HA15) on dextran sulfate sodium- (DSS-) induced ulcerative colitis in mice and explored the effect and related mechanism of HSPA5 inhibitor on the differentiation and function of Tregs. Methods Thirty-two C57BL/6 mice were randomly divided into four groups (8 mice per group): normal control group, DSS model group, HSPA5 inhibitor (HA15) group (intraperitoneal injection), and dexamethasone (DXM) group (intraperitoneal injection). Except for the blank control group, the other groups were induced with 3% DSS for 7 days and then given corresponding intervention therapy for 7 days. Results The disease activity index (DAI) score, colon length, histopathological changes, and scores of DSS-induced mice show that HA15 could significantly improve the degree of inflammation in ulcerative colitis. Moreover, HA15 can better inhibit the expression of HSPA5, HSPA1A, and CHIP in the colon and increase the level of FOXP3 mRNA. Finally, the content of Treg cells and the levels of IL-10 and TGF-β1 were significantly increased, and the levels of IL-6 were significantly reduced. Conclusions HA15 can improve the differentiation and function of Treg cells by inhibiting the HSPA1A/CHIP pathway, thereby improving ulcerative colitis. Therefore, inhibiting the expression of HSPA5 may serve as a new approach to treat ulcerative colitis.
Collapse
|
20
|
He W, Gao Y, Zhou J, Shi Y, Xia D, Shen HM. Friend or Foe? Implication of the autophagy-lysosome pathway in SARS-CoV-2 infection and COVID-19. Int J Biol Sci 2022; 18:4690-4703. [PMID: 35874956 PMCID: PMC9305279 DOI: 10.7150/ijbs.72544] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/24/2022] [Indexed: 12/14/2022] Open
Abstract
There is increasing amount of evidence indicating the close interplays between the replication cycle of SARS-CoV-2 and the autophagy-lysosome pathway in the host cells. While autophagy machinery is known to either assist or inhibit the viral replication process, the reciprocal effects of the SARS-CoV-2 on the autophagy-lysosome pathway have also been increasingly appreciated. More importantly, despite the disappointing results from the clinical trials of chloroquine and hydroxychloroquine in treatment of COVID-19, there is still ongoing effort in discovering new therapeutics targeting the autophagy-lysosome pathway. In this review, we provide an update-to-date summary of the interplays between the autophagy-lysosome pathway in the host cells and the pathogen SARS-CoV-2 at the molecular level, to highlight the prognostic value of autophagy markers in COVID-19 patients and to discuss the potential of developing novel therapeutic strategies for COVID-19 by targeting the autophagy-lysosome pathway. Thus, understanding the nature of such interactions between SARS-CoV-2 and the autophagy-lysosome pathway in the host cells is expected to provide novel strategies in battling against this global pandemic.
Collapse
Affiliation(s)
- Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yuan Gao
- Faculty of Health Sciences, University of Macau, Macau, China
- Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101 Beijing, China
| | - Jing Zhou
- Department of Physiology, School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi Province, China
| | - Yi Shi
- Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, 100101 Beijing, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health, Department of Gynecologic Oncology of Women's Hospital; Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
21
|
Lee SY, Wong PF, Jamal J, Roebuck MM. Naturally-derived endoplasmic reticulum stress inhibitors for osteoarthritis? Eur J Pharmacol 2022; 922:174903. [DOI: 10.1016/j.ejphar.2022.174903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/22/2022] [Accepted: 03/17/2022] [Indexed: 01/15/2023]
|
22
|
Shahriari-Felordi M, Alikhani HK, Hashemian SMR, Hassan M, Vosough M. Mini review ATF4 and GRP78 as novel molecular targets in ER-Stress modulation for critical COVID-19 patients. Mol Biol Rep 2022; 49:1545-1549. [PMID: 35028855 PMCID: PMC8758217 DOI: 10.1007/s11033-021-07071-9] [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: 10/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in more than 4.4 million deaths worldwide as of August 24, 2021. Viral infections such as SARS-CoV2 are associated with endoplasmic reticulum (ER) stress and also increased the level of reactive oxygen species. Activating transcription factor 4 (ATF4) is preferentially translated under integrated stress conditions and controls the genes involved in protein homeostasis, amino acid transport and metabolism, and also protection from oxidative stress. The GRP78, regulated either directly or indirectly by ATF4, is an essential chaperone in the ER and overexpressed and appears on the surface of almost all cells during stress and function as a SARS-CoV2 receptor. In this mini-review article, we briefly discuss the effects of SARS-CoV2 infection on the ER stress, and then the stress modulator functions of ATF4 and GRP78 as novel therapeutic targets were highlighted. Finally, the effects of GRP78 inhibitory components as potential factors for targeted therapies for COVID-19 critical cases were discussed.
Collapse
Affiliation(s)
- Mahtab Shahriari-Felordi
- Department of Molecular Cell Biology-Genetics, Faculty of Basic Sciences and Advanced Technologies in biology, University of Science and Culture, Tehran, Iran.,Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hani Keshavarz Alikhani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyed-Mohammad Reza Hashemian
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
23
|
Dent P. Cell Signaling and Translational Developmental Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC7538147 DOI: 10.1016/b978-0-12-820472-6.00002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The relationships between drug pharmacodynamics and subsequent changes in cellular signaling processes are complex. Many in vitro cell signaling studies often use drug concentrations above physiologically safe drug levels achievable in a patient's plasma. Drug companies develop agents to inhibit or modify the activities of specific target enzymes, often without a full consideration that their compounds have additional unknown targets. These two negative sequelae, when published together, become impediments against successful developmental therapeutics and translation because this data distorts our understanding of signaling mechanisms and reduces the probability of successfully translating drug-based concepts from the bench to the bedside. This article will discuss cellular signaling in isolation and as it relates to extant single and combined therapeutic drug interventions. This will lead to a hypothetical series standardized sequential approaches describing a rigorous concept to drug development and clinical translation.
Collapse
|
24
|
Law JN, Akers K, Tasnina N, Santina CMD, Deutsch S, Kshirsagar M, Klein-Seetharaman J, Crovella M, Rajagopalan P, Kasif S, Murali TM. Interpretable network propagation with application to expanding the repertoire of human proteins that interact with SARS-CoV-2. Gigascience 2021; 10:giab082. [PMID: 34966926 PMCID: PMC8716363 DOI: 10.1093/gigascience/giab082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/21/2021] [Accepted: 11/28/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Network propagation has been widely used for nearly 20 years to predict gene functions and phenotypes. Despite the popularity of this approach, little attention has been paid to the question of provenance tracing in this context, e.g., determining how much any experimental observation in the input contributes to the score of every prediction. RESULTS We design a network propagation framework with 2 novel components and apply it to predict human proteins that directly or indirectly interact with SARS-CoV-2 proteins. First, we trace the provenance of each prediction to its experimentally validated sources, which in our case are human proteins experimentally determined to interact with viral proteins. Second, we design a technique that helps to reduce the manual adjustment of parameters by users. We find that for every top-ranking prediction, the highest contribution to its score arises from a direct neighbor in a human protein-protein interaction network. We further analyze these results to develop functional insights on SARS-CoV-2 that expand on known biology such as the connection between endoplasmic reticulum stress, HSPA5, and anti-clotting agents. CONCLUSIONS We examine how our provenance-tracing method can be generalized to a broad class of network-based algorithms. We provide a useful resource for the SARS-CoV-2 community that implicates many previously undocumented proteins with putative functional relationships to viral infection. This resource includes potential drugs that can be opportunistically repositioned to target these proteins. We also discuss how our overall framework can be extended to other, newly emerging viruses.
Collapse
Affiliation(s)
- Jeffrey N Law
- Interdisciplinary Ph.D. Program in Genetics, Bioinformatics, and Computational Biology, Virginia Tech, Blacksburg, VA 24061, USA
| | - Kyle Akers
- Interdisciplinary Ph.D. Program in Genetics, Bioinformatics, and Computational Biology, Virginia Tech, Blacksburg, VA 24061, USA
| | - Nure Tasnina
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, USA
| | | | - Shay Deutsch
- Department of Mathematics, University of California, Los Angeles, CA 90095, USA
| | | | | | - Mark Crovella
- Department of Computer Science, Boston University, Boston, MA 02215, USA
| | | | - Simon Kasif
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
25
|
Tsao N, Chang YC, Hsieh SY, Li TC, Chiu CC, Yu HH, Hsu TC, Kuo CF. AR-12 Has a Bactericidal Activity and a Synergistic Effect with Gentamicin against Group A Streptococcus. Int J Mol Sci 2021; 22:ijms222111617. [PMID: 34769046 PMCID: PMC8583967 DOI: 10.3390/ijms222111617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/24/2021] [Accepted: 10/24/2021] [Indexed: 12/30/2022] Open
Abstract
Streptococcus pyogenes (group A Streptococcus (GAS) is an important human pathogen that can cause severe invasive infection, such as necrotizing fasciitis and streptococcal toxic shock syndrome. The mortality rate of streptococcal toxic shock syndrome ranges from 20% to 50% in spite of antibiotics administration. AR-12, a pyrazole derivative, has been reported to inhibit the infection of viruses, intracellular bacteria, and fungi. In this report, we evaluated the bactericidal activities and mechanisms of AR-12 on GAS infection. Our in vitro results showed that AR-12 dose-dependently reduced the GAS growth, and 2.5 μg/mL of AR-12 significantly killed GAS within 2 h. AR-12 caused a remarkable reduction in nucleic acid and protein content of GAS. The expression of heat shock protein DnaK and streptococcal exotoxins was also inhibited by AR-12. Surveys of the GAS architecture by scanning electron microscopy revealed that AR-12-treated GAS displayed incomplete septa and micro-spherical structures protruding out of cell walls. Moreover, the combination of AR-12 and gentamicin had a synergistic antibacterial activity against GAS replication for both in vitro and in vivo infection. Taken together, these novel findings obtained in this study may provide a new therapeutic strategy for invasive GAS infection.
Collapse
Affiliation(s)
- Nina Tsao
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
- Department of Biological Science and Technology, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (H.-H.Y.); (T.-C.H.)
| | - Ya-Chu Chang
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
| | - Sung-Yuan Hsieh
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu 300024, Taiwan;
| | - Tang-Chi Li
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
| | - Ching-Chen Chiu
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (N.T.); (Y.-C.C.); (T.-C.L.); (C.-C.C.)
| | - Hai-Han Yu
- Department of Biological Science and Technology, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (H.-H.Y.); (T.-C.H.)
| | - Tzu-Ching Hsu
- Department of Biological Science and Technology, College of Medical Science and Technology, I-Shou University, Kaohsiung 824005, Taiwan; (H.-H.Y.); (T.-C.H.)
| | - Chih-Feng Kuo
- School of Medicine, I-Shou University, Kaohsiung 824005, Taiwan
- Department of Nursing, College of Medicine, I-Shou University, Kaohsiung 824005, Taiwan
- Correspondence: ; Tel.: +886-7-6151100 (ext. 7967)
| |
Collapse
|
26
|
Abstract
Viruses are intracellular parasites that subvert the functions of their host cells to accomplish their infection cycle. The endoplasmic reticulum (ER)-residing chaperone proteins are central for the achievement of different steps of the viral cycle, from entry and replication to assembly and exit. The most abundant ER chaperones are GRP78 (78-kDa glucose-regulated protein), GRP94 (94-kDa glucose-regulated protein), the carbohydrate or lectin-like chaperones calnexin (CNX) and calreticulin (CRT), the protein disulfide isomerases (PDIs), and the DNAJ chaperones. This review will focus on the pleiotropic roles of ER chaperones during viral infection. We will cover their essential role in the folding and quality control of viral proteins, notably viral glycoproteins which play a major role in host cell infection. We will also describe how viruses co-opt ER chaperones at various steps of their infectious cycle but also in order to evade immune responses and avoid apoptosis. Finally, we will discuss the different molecules targeting these chaperones and the perspectives in the development of broad-spectrum antiviral drugs.
Collapse
|
27
|
Huang SSY, Toufiq M, Saraiva LR, Van Panhuys N, Chaussabel D, Garand M. Transcriptome and Literature Mining Highlight the Differential Expression of ERLIN1 in Immune Cells during Sepsis. BIOLOGY 2021; 10:755. [PMID: 34439987 PMCID: PMC8389572 DOI: 10.3390/biology10080755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/20/2022]
Abstract
Sepsis results from the dysregulation of the host immune system. This highly variable disease affects 19 million people globally, and accounts for 5 million deaths annually. In transcriptomic datasets curated from public repositories, we observed a consistent upregulation (3.26-5.29 fold) of ERLIN1-a gene coding for an ER membrane prohibitin and a regulator of inositol 1, 4, 5-trisphosphate receptors and sterol regulatory element-binding proteins-under septic conditions in healthy neutrophils, monocytes, and whole blood. In vitro expression of the ERLIN1 gene and proteins was measured by stimulating the whole blood of healthy volunteers to a combination of lipopolysaccharide and peptidoglycan. Septic stimulation induced a significant increase in ERLIN1 expression; however, ERLIN1 was differentially expressed among the immune blood cell subsets. ERLIN1 was uniquely increased in whole blood neutrophils, and confirmed in the differentiated HL60 cell line. The scarcity of ERLIN1 in sepsis literature indicates a knowledge gap between the functions of ERLIN1, calcium homeostasis, and cholesterol and fatty acid biosynthesis, and sepsis. In combination with experimental data, we bring forth the hypothesis that ERLIN1 is variably modulated among immune cells in response to cellular perturbations, and has implications for ER functions and/or ER membrane protein components during sepsis.
Collapse
Affiliation(s)
- Susie S. Y. Huang
- Research Department, Sidra Medicine, Doha 26999, Qatar; (M.T.); (L.R.S.); (N.V.P.); (D.C.)
| | - Mohammed Toufiq
- Research Department, Sidra Medicine, Doha 26999, Qatar; (M.T.); (L.R.S.); (N.V.P.); (D.C.)
| | - Luis R. Saraiva
- Research Department, Sidra Medicine, Doha 26999, Qatar; (M.T.); (L.R.S.); (N.V.P.); (D.C.)
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha 34110, Qatar
| | - Nicholas Van Panhuys
- Research Department, Sidra Medicine, Doha 26999, Qatar; (M.T.); (L.R.S.); (N.V.P.); (D.C.)
| | - Damien Chaussabel
- Research Department, Sidra Medicine, Doha 26999, Qatar; (M.T.); (L.R.S.); (N.V.P.); (D.C.)
| | - Mathieu Garand
- Research Department, Sidra Medicine, Doha 26999, Qatar; (M.T.); (L.R.S.); (N.V.P.); (D.C.)
| |
Collapse
|
28
|
Dent P, Booth L, Roberts JL, Poklepovic A, Cridebring D, Reiman EM. Inhibition of heat shock proteins increases autophagosome formation, and reduces the expression of APP, Tau, SOD1 G93A and TDP-43. Aging (Albany NY) 2021; 13:17097-17117. [PMID: 34252884 PMCID: PMC8312464 DOI: 10.18632/aging.203297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022]
Abstract
Aberrant expression and denaturation of Tau, amyloid-beta and TDP-43 can lead to cell death and is a major component of pathologies such as Alzheimer’s Disease (AD). AD neurons exhibit a reduced ability to form autophagosomes and degrade proteins via autophagy. Using genetically manipulated colon cancer cells we determined whether drugs that directly inhibit the chaperone ATPase activity or cause chaperone degradation and endoplasmic reticulum stress signaling leading to macroautophagy could reduce the levels of these proteins. The antiviral chaperone ATPase inhibitor AR12 reduced the ATPase activities and total expression of GRP78, HSP90, and HSP70, and of Tau, Tau 301L, APP, APP692, APP715, SOD1 G93A and TDP-43. In parallel, it increased the phosphorylation of ATG13 S318 and eIF2A S51 and caused eIF2A-dependent autophagosome formation and autophagic flux. Knock down of Beclin1 or ATG5 prevented chaperone, APP and Tau degradation. Neratinib, used to treat HER2+ breast cancer, reduced chaperone levels and expression of Tau and APP via macroautophagy, and neratinib interacted with AR12 to cause further reductions in protein levels. The autophagy-regulatory protein ATG16L1 is expressed as two isoforms, T300 or A300: Africans trend to express T300 and Europeans A300. We observed higher basal expression of Tau in T300 cells when compared to isogenic A300 cells. ATG16L1 isoform expression did not alter basal levels of HSP90, HSP70 or HSP27, however, basal levels of GRP78 were reduced in A300 cells. The abilities of both AR12 and neratinib to stimulate ATG13 S318 and eIF2A S51 phosphorylation and autophagic flux was also reduced in A300 cells. Our data support further evaluation of AR12 and neratinib in neuronal cells as repurposed treatments for AD.
Collapse
Affiliation(s)
- Paul Dent
- Department of Biochemistry and Molecular Biology, Richmond, VA 23298, USA
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Richmond, VA 23298, USA
| | - Jane L Roberts
- Department of Pharmacology and Toxicology, Richmond, VA 23298, USA
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Derek Cridebring
- Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, USA
| | - Eric M Reiman
- Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, USA.,Banner Alzheimer's Institute, Phoenix, AZ 85006, USA
| |
Collapse
|
29
|
Bath PM, Coleman CM, Gordon AL, Lim WS, Webb AJ. Nitric oxide for the prevention and treatment of viral, bacterial, protozoal and fungal infections. F1000Res 2021; 10:536. [PMID: 35685687 PMCID: PMC9171293 DOI: 10.12688/f1000research.51270.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Although the antimicrobial potential of nitric oxide (NO) is widely published, it is little used clinically. NO is a key signalling molecule modulating vascular, neuronal, inflammatory and immune responses. Endogenous antimicrobial activity is largely mediated by high local NO concentrations produced by cellular inducible nitric oxide synthase, and by derivative reactive nitrogen oxide species including peroxynitrite and S-nitrosothiols. NO may be taken as dietary substrate (inorganic nitrate, L-arginine), and therapeutically as gaseous NO, and transdermal, sublingual, oral, intranasal and intravenous nitrite or nitrate. Numerous preclinical studies have demonstrated that NO has generic static and cidal activities against viruses (including β-coronaviruses such as SARS-CoV-2), bacteria, protozoa and fungi/yeasts in vitro. Therapeutic effects have been seen in animal models in vivo, and phase II trials have demonstrated that NO donors can reduce microbial infection. Nevertheless, excess NO, as occurs in septic shock, is associated with increased morbidity and mortality. In view of the dose-dependent positive and negative effects of NO, safety and efficacy trials of NO and its donors are needed for assessing their role in the prevention and treatment of infections. Trials should test dietary inorganic nitrate for pre- or post-exposure prophylaxis and gaseous NO or oral, topical or intravenous nitrite and nitrate for treatment of mild-to-severe infections, including due to SARS-CoV-2 (COVID-19). This review summarises the evidence base from in vitro, in vivo and early phase clinical studies of NO activity in viral, bacterial, protozoal and fungal infections.
Collapse
Affiliation(s)
- Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
- Stroke, Nottingham University Hospitals NHS Trust, Nottingham, Notts, NG7 2UH, UK
| | - Christopher M Coleman
- Division of Infection, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
| | - Adam L Gordon
- Unit of Injury, Inflammation and Recovery Sciences, University of Nottingham, Derby, Derbyshire, DE22 3NE, UK
- NIHR Applied Research Collaboration-East Midlands (ARC-EM), Nottingham, Notts, UK
| | - Wei Shen Lim
- Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Andrew J Webb
- Clinical Pharmacology, School of Cardiovascular Medicine & Sciences, Kings College London British Heart Foundation Centre of Research Excellence, St Thomas' Hospital, London, SE1 7EH, UK
| |
Collapse
|
30
|
Downregulation of Tim-1 inhibits the proliferation, migration and invasion of glioblastoma cells via the miR-133a/TGFBR1 axis and the restriction of Wnt/β-catenin pathway. Cancer Cell Int 2021; 21:347. [PMID: 34225723 PMCID: PMC8256541 DOI: 10.1186/s12935-021-02036-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/21/2021] [Indexed: 01/11/2023] Open
Abstract
Background Glioblastoma remains one of the most lethal brain cancers. T-cell immunoglobulin and mucin domain 1 (Tim-1) is associated with various immune diseases. The molecular mechanism of Tim-1 in regulating glioblastoma cell proliferation, invasion, and migration is still unknown. Moreover, it has shown that miR-133a plays an important role in glioblastoma. However, little is known about the interaction between Tim-1 and miR-133a in glioblastoma. Methods Tim-1 expression in glioblastoma and normal brain tissues was detected by qPCR, Western Blot and IHC. After Tim-1 knockdown in U251 and U87 cells, genes showing significantly differential expression, along with the significant differential miRNAs were analyzed using RNA-seq analysis. The binding sites were verified using dual-luciferase reporter gene assay. U251 and U87 cells were allocated into the small harpin-negative control (sh-NC), sh-Tim-1, sh-Tim-1 + inhibitor NC, and sh-Tim-1 + miR-133a inhibitor group. Cell proliferation, migration, and invasion were determined by CCK-8, flow cytometry, wound-healing and Transwell assays, respectively. Next, U251 and U87 cells were allocated into the mimic NC, miR-133a mimic, miR-133a mimic + pcDNA3.1, and miR-133a mimic + pcDNA3.1-TGFBR1 groups, followed by the detection of cell proliferation, migration, and invasion. Western blot was used to identify the expression of vital kinases in the Wnt/β-catenin pathway. Results Tim-1 was highly expressed in glioblastoma tissues compared with that in normal brain tissues. RNA-seq analysis showed that Tim-1 knockdown could lead to the downregulation of TGFBR1 and the upregulation of miR-133a. The binding sites between TGFBR1 and miR-133a were confirmed. Tim-1 knockdown impaired the invasion, migration, proliferation of U251 and U87 cells, which could be reversed by miR-133a downregulation. miR-133a upregulation inhibited the proliferation, invasion, and migration of U251 and U87 cells, which could be reversed by TGFBR1 upregulation. Tim-1 knockdown and miR-133a upregulation could inhibit the activation of the Wnt/β-catenin pathway, while the elevation of TGFBR1 showed opposite effects. Conclusion Tim-1 knockdown inhibited glioblastoma cell proliferation, invasion, and migration through the miR-133a/TGFBR1 axis and restrained the activation of the Wnt/β-catenin pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02036-1.
Collapse
|
31
|
Bath PM, Coleman CM, Gordon AL, Lim WS, Webb AJ. Nitric oxide for the prevention and treatment of viral, bacterial, protozoal and fungal infections. F1000Res 2021; 10:536. [PMID: 35685687 PMCID: PMC9171293 DOI: 10.12688/f1000research.51270.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 12/18/2023] Open
Abstract
Although the antimicrobial potential of nitric oxide (NO) is widely published, it is little used clinically. NO is a key signalling molecule modulating vascular, neuronal, inflammatory and immune responses. Endogenous antimicrobial activity is largely mediated by high local NO concentrations produced by cellular inducible nitric oxide synthase, and by derivative reactive nitrogen oxide species including peroxynitrite and S-nitrosothiols. NO may be taken as dietary substrate (inorganic nitrate, L-arginine), and therapeutically as gaseous NO, and transdermal, sublingual, oral, intranasal and intravenous nitrite or nitrate. Numerous preclinical studies have demonstrated that NO has generic static and cidal activities against viruses (including β-coronaviruses such as SARS-CoV-2), bacteria, protozoa and fungi/yeasts in vitro. Therapeutic effects have been seen in animal models in vivo, and phase II trials have demonstrated that NO donors can reduce microbial infection. Nevertheless, excess NO, as occurs in septic shock, is associated with increased morbidity and mortality. In view of the dose-dependent positive and negative effects of NO, safety and efficacy trials of NO and its donors are needed for assessing their role in the prevention and treatment of infections. Trials should test dietary inorganic nitrate for pre- or post-exposure prophylaxis and gaseous NO or oral, topical or intravenous nitrite and nitrate for treatment of mild-to-severe infections, including due to SARS-CoV-2 (COVID-19). This review summarises the evidence base from in vitro, in vivo and early phase clinical studies of NO activity in viral, bacterial, protozoal and fungal infections.
Collapse
Affiliation(s)
- Philip M. Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
- Stroke, Nottingham University Hospitals NHS Trust, Nottingham, Notts, NG7 2UH, UK
| | - Christopher M. Coleman
- Division of Infection, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, Notts, NG7 2UH, UK
| | - Adam L. Gordon
- Unit of Injury, Inflammation and Recovery Sciences, University of Nottingham, Derby, Derbyshire, DE22 3NE, UK
- NIHR Applied Research Collaboration-East Midlands (ARC-EM), Nottingham, Notts, UK
| | - Wei Shen Lim
- Respiratory Medicine, Nottingham University Hospitals NHS Trust, Nottingham, NG5 1PB, UK
| | - Andrew J. Webb
- Clinical Pharmacology, School of Cardiovascular Medicine & Sciences, Kings College London British Heart Foundation Centre of Research Excellence, St Thomas' Hospital, London, SE1 7EH, UK
| |
Collapse
|
32
|
Sun B, Tan D, Pan D, Baker MR, Liang Z, Wang Z, Lei J, Liu S, Hu CY, Li QX. Dihydromyricetin Imbues Antiadipogenic Effects on 3T3-L1 Cells via Direct Interactions with 78-kDa Glucose-Regulated Protein. J Nutr 2021; 151:1717-1725. [PMID: 33830233 DOI: 10.1093/jn/nxab057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/15/2021] [Accepted: 02/11/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Obesity is among the most serious public health problems worldwide, with few safe pharmaceutical interventions. Natural products have become an important source of potential anti-obesity therapeutics. Dihydromyricetin (DHM) exerts antidiabetic effects. The biochemical target of DHM, however, has been unknown. It is crucial to identify the biochemical target of DHM for elucidating its physiological function and therapeutic value. OBJECTIVES The objective of this study was to identify the biochemical target of DHM. METHODS An abundant antiadipogenic flavanonol was extracted from the herbal plant Ampelopsis grossedentata through bioassay-guided fractionation and characterized with high-resolution LC-MS and 1H and 13C nuclear magnetic resonance. Antiadipogenic experiments were done with mouse 3T3-L1 preadipocytes. A biochemical target of the chemical of interest was identified with drug affinity responsive target stability assay. Direct interactions between the chemical of interest and the protein target in vitro were predicted with molecular docking and subsequently confirmed with surface plasmon resonance. Expression levels of peroxisome proliferator-activated receptor γ (PPARγ), which is associated with 78-kDa glucose-regulated protein (GRP78), were measured with real-time qPCR. RESULTS DHM was isolated, purified, and structurally characterized. Cellular studies showed that DHM notably reduced intracellular oil droplet formation in 3T3-L1 cells with a median effective concentration of 294 μM (i.e., 94 μg/mL). DHM targeted the ATP binding site of GRP78, which is associated with adipogenesis. An equilibrium dissociation constant between DHM and GRP78 was 21.8 μM. In 3T3-L1 cells upon treatment with DHM at 50 μM (i.e., 16 μg/mL), the expression level of PPARγ was downregulated to 53.9% of the solvent vehicle control's level. CONCLUSIONS DHM targets GRP78 in vitro. DHM is able to reduce lipid droplet formation in 3T3-L1 cells through a mode of action that is plausibly associated with direct interactions between GRP78 and DHM, which is a step forward in determining potential applications of DHM as an anti-obesity agent.
Collapse
Affiliation(s)
- Binmei Sun
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA.,College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Deguan Tan
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA.,Institute of Tropical Bioscience and Biotechnology, Ministry of Agriculture Key Laboratory of Tropical Crops Biology and Genetic Resources and Hainan Academy of Tropical Agricultural Resource, Chinese Academy of Tropical Agriculural Sciences, Haikou, China
| | - Dongjin Pan
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA.,Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Margaret R Baker
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Zhibin Liang
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Zhizheng Wang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, China
| | - Jianjun Lei
- College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Shaoqun Liu
- College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Ching Yuan Hu
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
33
|
Fu J, Wei C, He J, Zhang L, Zhou J, Balaji KS, Shen S, Peng J, Sharma A, Fu J. Evaluation and characterization of HSPA5 (GRP78) expression profiles in normal individuals and cancer patients with COVID-19. Int J Biol Sci 2021; 17:897-910. [PMID: 33767597 PMCID: PMC7975696 DOI: 10.7150/ijbs.54055] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
HSPA5 (BiP, GRP78) has been reported as a potential host-cell receptor for SARS-Cov-2, but its expression profiles on different tissues including tumors, its susceptibility to SARS-Cov-2 virus and severity of its adverse effects on malignant patients are unclear. In the current study, HSPA5 has been found to be expressed ubiquitously in normal tissues and significantly increased in 14 of 31 types of cancer tissues. In lung cancer, mRNA levels of HSPA5 were 253-fold increase than that of ACE2. Meanwhile, in both malignant tumors and matched normal samples across almost all cancer types, mRNA levels of HSPA5 were much higher than those of ACE2. Higher expression of HSPA5 significantly decreased patient overall survival (OS) in 7 types of cancers. Moreover, systematic analyses found that 7.15% of 5,068 COVID-19 cases have malignant cancer coincidental situations, and the rate of severe events of COVID-19 patients with cancers present a higher trend than that for all COVID-19 patients, showing a significant difference (33.33% vs 16.09%, p<0.01). Collectively, these data imply that the tissues with high HSPA5 expression, not low ACE2 expression, are susceptible to be invaded by SARS-CoV-2. Taken together, this study not only indicates the clinical significance of HSPA5 in COVID-19 disease and cancers, but also provides potential clues for further medical treatments and managements of COVID-19 patients.
Collapse
Affiliation(s)
- Jiewen Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Chunli Wei
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Jiayue He
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Lianmei Zhang
- Department of Pathology, the Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu, China
| | - Ju Zhou
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | | | - Shiyi Shen
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Jiangzhou Peng
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510000, China
| | - Amrish Sharma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston 77030, Texas, USA
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| |
Collapse
|
34
|
Booth LA, Roberts JL, Dent P. The role of cell signaling in the crosstalk between autophagy and apoptosis in the regulation of tumor cell survival in response to sorafenib and neratinib. Semin Cancer Biol 2020; 66:129-139. [PMID: 31644944 PMCID: PMC7167338 DOI: 10.1016/j.semcancer.2019.10.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 09/23/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms by which tumor cells survive or die following therapeutic interventions are complex. There are three broadly defined categories of cell death processes: apoptosis (Type I), autophagic cell death (Type II), and necrosis (Type III). In hematopoietic tumor cells, the majority of toxic stimuli cause these cells to undergo a death process called apoptosis; apoptosis specifically involves the cleavage of DNA into large defined pieces and their subsequent localization in vesicles. Thus, 'pure' apoptosis largely lacks inflammatory potential. In carcinomas, however, the mechanisms by which tumor cells ultimately die are considerably more complex. Although the machinery of apoptosis is engaged by toxic stimuli, other processes such as autophagy ("self-eating") and replicative cell death can lead to observations that do not simplistically correspond to any of the individual Type I-III formalized death categories. The 'hybrid' forms of cell death observed in carcinoma cells result in cellular materials being released into the extracellular space without packaging, which promotes inflammation, potentially leading to the accelerated re-growth of surviving tumor cells by macrophages. Drugs as single agents or in combinations can simultaneously initiate signaling via both apoptotic and autophagic pathways. Based on the tumor type and its oncogene drivers, as well as the drug(s) being used and the duration and intensity of the autophagosome signal, apoptosis and autophagy have the potential to act in concert to kill or alternatively that the actions of either pathway can act to suppress signaling by the other pathway. And, there also is evidence that autophagic flux, by causing lysosomal protease activation, with their subsequent release into the cytosol, can directly mediate killing. This review will discuss the interactive biology between apoptosis and autophagy in carcinoma cells. Finally, the molecular actions of the FDA-approved drugs neratinib and sorafenib, and how they enhance both apoptotic and toxic autophagic processes, alone or in combination with other agents, is discussed in a bench-to-bedside manner.
Collapse
Affiliation(s)
- Laurence A Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, 401 College St, Richmond, VA 23298, United States.
| |
Collapse
|
35
|
Rayner JO, Roberts RA, Kim J, Poklepovic A, Roberts JL, Booth L, Dent P. AR12 (OSU-03012) suppresses GRP78 expression and inhibits SARS-CoV-2 replication. Biochem Pharmacol 2020; 182:114227. [PMID: 32966814 PMCID: PMC7502229 DOI: 10.1016/j.bcp.2020.114227] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
AR12 is a derivative of celecoxib which no-longer acts against COX2 but instead inhibits the ATPase activity of multiple chaperone proteins, in particular GRP78. GRP78 acts as a sensor of endoplasmic reticulum stress and is an essential chaperone required for the life cycle of all mammalian viruses. We and others previously demonstrated in vitro and in vivo that AR12 increases autophagosome formation and autophagic flux, enhances virus protein degradation, preventing virus reproduction, and prolonging the survival of infected animals. In this report, we determined whether AR12 could act against SARS-CoV-2. In a dose-dependent fashion AR12 inhibited SARS-CoV-2 spike protein expression in transfected or infected cells. AR12 suppressed the production of infectious virions via autophagosome formation, which was also associated with degradation of GRP78. After AR12 exposure, the colocalization of GRP78 with spike protein was reduced. Knock down of eIF2α prevented AR12-induced spike degradation and knock down of Beclin1 or ATG5 caused the spike protein to localize in LAMP2+ vesicles without apparent degradation. HCT116 cells expressing ATG16L1 T300, found in the majority of persons of non-European descent, particularly from Africa, expressed greater amounts of GRP78 and SARS-CoV-2 receptor angiotensin converting enzyme 2 compared to ATG16L1 A300, predominantly found in Europeans, suggestive that ATG16L1 T300 expression may be associated with a greater ability to be infected and to reproduce SARS-CoV-2. In conclusion, our findings demonstrate that AR12 represents a clinically relevant anti-viral drug for the treatment of SARS-CoV-2.
Collapse
Affiliation(s)
- Jonathan O Rayner
- Department of Microbiology and Immunology, Laboratory of Infectious Diseases, University of South Alabama, Mobile, AL 36688-0002, United States
| | - Rosemary A Roberts
- Department of Microbiology and Immunology, Laboratory of Infectious Diseases, University of South Alabama, Mobile, AL 36688-0002, United States
| | - Jin Kim
- Department of Microbiology and Immunology, Laboratory of Infectious Diseases, University of South Alabama, Mobile, AL 36688-0002, United States
| | - Andrew Poklepovic
- Departments of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0035, United States
| | - Jane L Roberts
- Departments of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298-0035, United States
| | - Laurence Booth
- Departments of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, United States
| | - Paul Dent
- Departments of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298-0035, United States.
| |
Collapse
|
36
|
Gao F, Fan H. Heat shock protein 5 and inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2020; 28:802-806. [DOI: 10.11569/wcjd.v28.i16.802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease is a kind of chronic recurrent intestinal inflammatory disease whose occurrence and development are affected by the integrity of the mucosal barrier. As the main component of the mucosal barrier, intestinal epithelial cells mainly include Paneth cells, goblet cells, etc. Heat shock protein 5 is a key factor for endoplasmic reticulum stress, and it affects the survival and apoptosis of intestinal epithelial cells mainly through endoplasmic reticulum stress pathways, and then participates in the process of inflammatory bowel disease.
Collapse
Affiliation(s)
- Fei Gao
- Department of Integrated Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Heng Fan
- Department of Integrated Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
37
|
Ha DP, Lee AS. Insulin-like growth factor 1-receptor signaling stimulates GRP78 expression through the PI3K/AKT/mTOR/ATF4 axis. Cell Signal 2020; 75:109736. [PMID: 32805346 DOI: 10.1016/j.cellsig.2020.109736] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/06/2020] [Accepted: 08/09/2020] [Indexed: 01/09/2023]
Abstract
GRP78, a major molecular chaperone, is critical for the folding and maturation of membrane and secretory proteins and serves as the master regulator of the unfolded protein response. Thus, GRP78 is frequently upregulated in highly proliferative cells to cope with elevated protein synthesis and metabolic stress. IGF-1 is a potent regulator of cell growth, metabolism and survival. Previously we discovered that GRP78 is a novel downstream target of IGF-1 signaling by utilizing mouse embryonic fibroblast model systems where the IGF-1 receptor (IGF-1R) was either overexpressed (R+) or knockout (R-). Here we investigated the mechanisms whereby GRP78 is upregulated in the R+ cells. Our studies revealed that suppression of PI3K/AKT/mTOR downstream of IGF-1R signaling resulted in concurrent decrease in GRP78 and the transcription factor ATF4. Through knock-down and overexpression studies, we established ATF4 as the essential downstream nodal of the PI3K/AKT/mTOR signaling pathway critical for GRP78 transcriptional upregulation mediated by IGF-1R.
Collapse
Affiliation(s)
- Dat P Ha
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
38
|
Lu G, Luo H, Zhu X. Targeting the GRP78 Pathway for Cancer Therapy. Front Med (Lausanne) 2020; 7:351. [PMID: 32850882 PMCID: PMC7409388 DOI: 10.3389/fmed.2020.00351] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/11/2020] [Indexed: 12/24/2022] Open
Abstract
The 78-kDa glucose-regulated protein (GRP78) plays an important part in maintaining protein stability, regulating protein folding, and inducing apoptosis autophagy, which is considered as a powerful protein. Meanwhile, it also plays a role in ensuring the normal function of organs. In recent years, more and more researches have been carried out on the targeted therapy of GRP78, mainly focusing on its relevant role in tumor and its role as a major modulator and modulator of subordinate pathways. The ability of GRP78 to respond to endoplasmic reticulum stress (ERS) determines whether tumor cells survive and whether the changes in expression level of GRP78 regulated by endoplasmic reticulum (ER) caused by various factors will directly or indirectly affect cell proliferation, apoptosis, and injury, or reduce the body's defense ability, or have protective effects on various organs.
Collapse
Affiliation(s)
- Guanhua Lu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Hui Luo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China.,The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory, Zhanjiang, China.,The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
39
|
AR-12 Exhibits Direct and Host-Targeted Antibacterial Activity toward Mycobacterium abscessus. Antimicrob Agents Chemother 2020; 64:AAC.00236-20. [PMID: 32482678 PMCID: PMC7526805 DOI: 10.1128/aac.00236-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
Therapeutic options for Mycobacterium abscessus infections are extremely limited. New or repurposed drugs are needed. The anti-M. abscessus activity of AR-12 (OSU-03012), reported to express broad-spectrum antimicrobial effects, was investigated in vitro and in vivo Antimicrobial susceptibility testing was performed on 194 clinical isolates. Minimum bactericidal concentration and time-kill kinetics assays were conducted to distinguish the bactericidal versus bacteriostatic activity of AR-12. Synergy between AR-12 and five clinically important antibiotics was determined using a checkerboard synergy assay. The activity of AR-12 against intracellular M. abscessus residing within macrophage was also evaluated. Finally, the potency of AR-12 in vivo was determined in a neutropenic mouse model that mimics pulmonary M. abscessus infection. AR-12 exhibited high anti-M. abscessus activity in vitro, with an MIC50 of 4 mg/liter (8.7 μM) and an MIC90 of 8 mg/liter (17.4 μM) for both subsp. abscessus and subsp. massiliense AR-12 and amikacin exhibited comparable bactericidal activity against extracellular M. abscessus in culture. AR-12, however, exhibited significantly greater intracellular antibacterial activity than amikacin and caused a significant reduction in the bacterial load in the lungs of neutropenic mice infected with M. abscessus No antagonism between AR-12 and clarithromycin, amikacin, imipenem, cefoxitin, or tigecycline was evident. In conclusion, AR-12 is active against M. abscessus in vitro and in vivo and does not antagonize the most frequently used anti-M. abscessus drugs. As such, AR-12 is a potential candidate to include in novel strategies to treat M. abscessus infections.
Collapse
|
40
|
Glucose-Regulated Protein 78 Interacts with Zika Virus Envelope Protein and Contributes to a Productive Infection. Viruses 2020; 12:v12050524. [PMID: 32397571 PMCID: PMC7290722 DOI: 10.3390/v12050524] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 12/23/2022] Open
Abstract
Zika virus (ZIKV; Flaviviridae) is a mosquito-borne flavivirus shown to cause fetal abnormalities collectively known as congenital Zika syndrome and Guillain-Barré syndrome in recent outbreaks. Currently, there is no specific treatment or vaccine available, and more effort is needed to identify cellular factors in the viral life cycle. Here, we investigated interactors of ZIKV envelope (E) protein by combining protein pull-down with mass spectrometry. We found that E interacts with the endoplasmic reticulum (ER) resident chaperone, glucose regulated protein 78 (GRP78). Although other flaviviruses are known to co-opt ER resident proteins, including GRP78, to enhance viral infectivity, the role ER proteins play during the ZIKV life cycle is yet to be elucidated. We showed that GRP78 levels increased during ZIKV infection and localised to sites coincident with ZIKV E staining. Depletion of GRP78 using specific siRNAs significantly reduced reporter-virus luciferase readings, viral protein synthesis, and viral titres. Additionally, GRP78 depletion reduced the ability of ZIKV to disrupt host cell translation and altered the localisation of viral replication factories, though there was no effect on viral RNA synthesis. In summary, we showed GRP78 is a vital host-factor during ZIKV infection, which may be involved in the coordination of viral replication factories.
Collapse
|
41
|
Molecular Chaperones in Cancer Stem Cells: Determinants of Stemness and Potential Targets for Antitumor Therapy. Cells 2020; 9:cells9040892. [PMID: 32268506 PMCID: PMC7226806 DOI: 10.3390/cells9040892] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) are a great challenge in the fight against cancer because these self-renewing tumorigenic cell fractions are thought to be responsible for metastasis dissemination and cases of tumor recurrence. In comparison with non-stem cancer cells, CSCs are known to be more resistant to chemotherapy, radiotherapy, and immunotherapy. Elucidation of mechanisms and factors that promote the emergence and existence of CSCs and their high resistance to cytotoxic treatments would help to develop effective CSC-targeting therapeutics. The present review is dedicated to the implication of molecular chaperones (protein regulators of polypeptide chain folding) in both the formation/maintenance of the CSC phenotype and cytoprotective machinery allowing CSCs to survive after drug or radiation exposure and evade immune attack. The major cellular chaperones, namely heat shock proteins (HSP90, HSP70, HSP40, HSP27), glucose-regulated proteins (GRP94, GRP78, GRP75), tumor necrosis factor receptor-associated protein 1 (TRAP1), peptidyl-prolyl isomerases, protein disulfide isomerases, calreticulin, and also a transcription heat shock factor 1 (HSF1) initiating HSP gene expression are here considered as determinants of the cancer cell stemness and potential targets for a therapeutic attack on CSCs. Various approaches and agents are discussed that may be used for inhibiting the chaperone-dependent development/manifestations of cancer cell stemness.
Collapse
|
42
|
Identification and Characterization of Novel Compounds with Broad-Spectrum Antiviral Activity against Influenza A and B Viruses. J Virol 2020; 94:JVI.02149-19. [PMID: 31941776 DOI: 10.1128/jvi.02149-19] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 12/23/2022] Open
Abstract
Influenza A (IAV) and influenza B (IBV) viruses are highly contagious pathogens that cause fatal respiratory disease every year, with high economic impact. In addition, IAV can cause pandemic infections with great consequences when new viruses are introduced into humans. In this study, we evaluated 10 previously described compounds with antiviral activity against mammarenaviruses for their ability to inhibit IAV infection using our recently described bireporter influenza A/Puerto Rico/8/34 (PR8) H1N1 (BIRFLU). Among the 10 tested compounds, eight (antimycin A [AmA], brequinar [BRQ], 6-azauridine, azaribine, pyrazofurin [PF], AVN-944, mycophenolate mofetil [MMF], and mycophenolic acid [MPA]), but not obatoclax or Osu-03012, showed potent anti-influenza virus activity under posttreatment conditions [median 50% effective concentration (EC50) = 3.80 nM to 1.73 μM; selective index SI for 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay, >28.90 to 13,157.89]. AmA, 6-azauridine, azaribine, and PF also showed potent inhibitory effect in pretreatment (EC50 = 0.14 μM to 0.55 μM; SI-MTT = 70.12 to >357.14) or cotreatment (EC50 = 34.69 nM to 7.52 μM; SI-MTT = 5.24 to > 1,441.33) settings. All of the compounds tested inhibited viral genome replication and gene transcription, and none of them affected host cellular RNA polymerase II activities. The antiviral activity of the eight identified compounds against BIRFLU was further confirmed with seasonal IAVs (A/California/04/2009 H1N1 and A/Wyoming/3/2003 H3N2) and an IBV (B/Brisbane/60/2008, Victoria lineage), demonstrating their broad-spectrum prophylactic and therapeutic activity against currently circulating influenza viruses in humans. Together, our results identified a new set of antiviral compounds for the potential treatment of influenza viral infections.IMPORTANCE Influenza viruses are highly contagious pathogens and are a major threat to human health. Vaccination remains the most effective tool to protect humans against influenza infection. However, vaccination does not always guarantee complete protection against drifted or, more noticeably, shifted influenza viruses. Although U.S. Food and Drug Administration (FDA) drugs are approved for the treatment of influenza infections, influenza viruses resistant to current FDA antivirals have been reported and continue to emerge. Therefore, there is an urgent need to find novel antivirals for the treatment of influenza viral infections in humans, a search that could be expedited by repurposing currently approved drugs. In this study, we assessed the influenza antiviral activity of 10 compounds previously shown to inhibit mammarenavirus infection. Among them, eight drugs showed antiviral activities, providing a new battery of drugs that could be used for the treatment of influenza infections.
Collapse
|
43
|
Regulation of Molecular Chaperone GRP78 by Hepatitis B Virus: Control of Viral Replication and Cell Survival. Mol Cell Biol 2020; 40:MCB.00475-19. [PMID: 31712392 DOI: 10.1128/mcb.00475-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/01/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic hepatitis B (CHB) remains a global health problem, carrying a high risk for progression into cirrhosis and liver failure. Molecular chaperones are involved in diverse pathophysiological processes including viral infection. However, the role of molecular chaperones in hepatitis B virus (HBV) infection and its underlying mechanisms remain unclear. Here, we identified GRP78 as one of the molecular chaperones most strongly induced by HBV in human hepatocytes. Gain- and loss-of-function analyses demonstrated that GRP78 exerted an inhibitory effect on HBV transcription and replication. Further study showed that GRP78 was involved in the activation of AKT/mTOR signaling in hepatocytes, which contributed to GRP78-mediated inhibition of HBV. Of note, HBV-upregulated GRP78 was found to play a crucial role in maintaining the survival of hepatocytes via facilitating a mild endoplasmic reticulum (ER) stress. Together, our findings suggest that HBV may sacrifice part of its replication for establishing a persistent infection through induction of GRP78, a master ER stress regulator. Targeting GRP78 may help develop to design novel therapeutic strategies against chronic HBV infection and the associated hepatocellular carcinoma.
Collapse
|
44
|
Integrative multi-omic analysis identifies new drivers and pathways in molecularly distinct subtypes of ALS. Sci Rep 2019; 9:9968. [PMID: 31292500 PMCID: PMC6620285 DOI: 10.1038/s41598-019-46355-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable and fatal neurodegenerative disease. Increasing the chances of success for future clinical strategies requires more in-depth knowledge of the molecular basis underlying disease heterogeneity. We recently laid the foundation for a molecular taxonomy of ALS by whole-genome expression profiling of motor cortex from sporadic ALS (SALS) patients. Here, we analyzed copy number variants (CNVs) occurring in the same patients, by using a customized exon-centered comparative genomic hybridization array (aCGH) covering a large panel of ALS-related genes. A large number of novel and known disease-associated CNVs were detected in SALS samples, including several subgroup-specific loci, suggestive of a great divergence of two subgroups at the molecular level. Integrative analysis of copy number profiles with their associated transcriptomic data revealed subtype-specific genomic perturbations and candidate driver genes positively correlated with transcriptional signatures, suggesting a strong interaction between genomic and transcriptomic events in ALS pathogenesis. The functional analysis confirmed our previous pathway-based characterization of SALS subtypes and identified 24 potential candidates for genomic-based patient stratification. To our knowledge, this is the first comprehensive "omics" analysis of molecular events characterizing SALS pathology, providing a road map to facilitate genome-guided personalized diagnosis and treatments for this devastating disease.
Collapse
|
45
|
Krishnamurthy M, Lemmon MM, Falcinelli EM, Sandy RA, Dootz JN, Mott TM, Rajamani S, Schaecher KE, Duplantier AJ, Panchal RG. Enhancing the antibacterial activity of polymyxins using a nonantibiotic drug. Infect Drug Resist 2019; 12:1393-1405. [PMID: 31239720 PMCID: PMC6555264 DOI: 10.2147/idr.s196874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/06/2019] [Indexed: 11/24/2022] Open
Abstract
Purpose: The rapid emergence of multidrug-resistant (MDR) bacteria and the lack of new therapies to eliminate them poses a major threat to global health. With the alarming rise in antimicrobial resistance (AMR), focus has now shifted to the use of the polymyxin class of antibiotics as the last line of defense for treatment of Gram-negative infections. Unfortunately, the growing resistance of bacteria against polymyxins is threatening the treatment of MDR infections, necessitating the need for novel strategies. The objective of this study was to determine if combination of polymyxin (polymyxin B or colistin) with a nonantibiotic small molecule AR-12, a celecoxib derivative that is devoid of cyclooxygenase 2 (COX-2) inhibitory activities, can be an effective strategy against polymyxin-resistant MDR bacteria. Methods: Growth inhibition studies, time-kill assays and permeability assays were conducted to investigate the effect of AR-12 on the antibacterial activity of polymyxins. Results: Growth studies were performed on a panel of polymyxin-resistant MDR strains using the combination of AR-12 with either colistin or polymyxin B. The combination treatment had no effect on strains that have inherent polymyxin resistance; however, AR-12 was effective in lowering the minimal inhibitory concentration (MIC) of polymyxins by 4–60-fold in several strains that had acquired polymyxin resistance. Time-kill assays using the combination of AR-12 and colistin with select MDR strains suggest rapid killing and bactericidal activity, while the permeability assays using fluorescently labeled dansylated polymyxin and 1-N-phenylnaphthylamine (NPN) in these MDR strains suggest that AR-12 can potentiate the antibacterial activity of polymyxins by possibly altering the bacterial outer membrane via modification of lipopolysaccharide and thereby improving the uptake of polymyxins. Conclusion: Our studies indicate that the combination of AR-12 and polymyxin is effective in targeting select Gram-negative bacteria that have acquired polymyxin resistance. Further understanding of the mechanism of action of AR-12 will provide new avenues for developing narrow-spectrum antibacterials to target select Gram-negative MDR bacteria. Importantly, our studies show that the use of nonantibiotic small molecules in combination with polymyxins is an attractive strategy to counter the growing resistance of bacteria to polymyxins.
Collapse
Affiliation(s)
- Malathy Krishnamurthy
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Margaret M Lemmon
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Evan M Falcinelli
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Reuel A Sandy
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Jennifer N Dootz
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Tiffany M Mott
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Sathish Rajamani
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA.,General Dynamics Information Technology, Frederick, MD, USA
| | - Kurt E Schaecher
- Department of Pathology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Allen J Duplantier
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA.,Cherokee Nation Assurance, Frederick, MD, USA
| | - Rekha G Panchal
- Department of Target Discovery and Experimental Microbiology, Division of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| |
Collapse
|
46
|
Almanza A, Carlesso A, Chintha C, Creedican S, Doultsinos D, Leuzzi B, Luís A, McCarthy N, Montibeller L, More S, Papaioannou A, Püschel F, Sassano ML, Skoko J, Agostinis P, de Belleroche J, Eriksson LA, Fulda S, Gorman AM, Healy S, Kozlov A, Muñoz‐Pinedo C, Rehm M, Chevet E, Samali A. Endoplasmic reticulum stress signalling - from basic mechanisms to clinical applications. FEBS J 2019; 286:241-278. [PMID: 30027602 PMCID: PMC7379631 DOI: 10.1111/febs.14608] [Citation(s) in RCA: 600] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/24/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER) is a membranous intracellular organelle and the first compartment of the secretory pathway. As such, the ER contributes to the production and folding of approximately one-third of cellular proteins, and is thus inextricably linked to the maintenance of cellular homeostasis and the fine balance between health and disease. Specific ER stress signalling pathways, collectively known as the unfolded protein response (UPR), are required for maintaining ER homeostasis. The UPR is triggered when ER protein folding capacity is overwhelmed by cellular demand and the UPR initially aims to restore ER homeostasis and normal cellular functions. However, if this fails, then the UPR triggers cell death. In this review, we provide a UPR signalling-centric view of ER functions, from the ER's discovery to the latest advancements in the understanding of ER and UPR biology. Our review provides a synthesis of intracellular ER signalling revolving around proteostasis and the UPR, its impact on other organelles and cellular behaviour, its multifaceted and dynamic response to stress and its role in physiology, before finally exploring the potential exploitation of this knowledge to tackle unresolved biological questions and address unmet biomedical needs. Thus, we provide an integrated and global view of existing literature on ER signalling pathways and their use for therapeutic purposes.
Collapse
Affiliation(s)
- Aitor Almanza
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | - Antonio Carlesso
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGöteborgSweden
| | - Chetan Chintha
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | | | - Dimitrios Doultsinos
- INSERM U1242University of RennesFrance
- Centre de Lutte Contre le Cancer Eugène MarquisRennesFrance
| | - Brian Leuzzi
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | - Andreia Luís
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CentreViennaAustria
| | - Nicole McCarthy
- Institute for Experimental Cancer Research in PaediatricsGoethe‐UniversityFrankfurtGermany
| | - Luigi Montibeller
- Neurogenetics GroupDivision of Brain SciencesFaculty of MedicineImperial College LondonUK
| | - Sanket More
- Department Cellular and Molecular MedicineLaboratory of Cell Death and TherapyKU LeuvenBelgium
| | - Alexandra Papaioannou
- INSERM U1242University of RennesFrance
- Centre de Lutte Contre le Cancer Eugène MarquisRennesFrance
| | - Franziska Püschel
- Cell Death Regulation GroupOncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Maria Livia Sassano
- Department Cellular and Molecular MedicineLaboratory of Cell Death and TherapyKU LeuvenBelgium
| | - Josip Skoko
- Institute of Cell Biology and ImmunologyUniversity of StuttgartGermany
| | - Patrizia Agostinis
- Department Cellular and Molecular MedicineLaboratory of Cell Death and TherapyKU LeuvenBelgium
| | - Jackie de Belleroche
- Neurogenetics GroupDivision of Brain SciencesFaculty of MedicineImperial College LondonUK
| | - Leif A. Eriksson
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGöteborgSweden
| | - Simone Fulda
- Institute for Experimental Cancer Research in PaediatricsGoethe‐UniversityFrankfurtGermany
| | | | - Sandra Healy
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | - Andrey Kozlov
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CentreViennaAustria
| | - Cristina Muñoz‐Pinedo
- Cell Death Regulation GroupOncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Markus Rehm
- Institute of Cell Biology and ImmunologyUniversity of StuttgartGermany
| | - Eric Chevet
- INSERM U1242University of RennesFrance
- Centre de Lutte Contre le Cancer Eugène MarquisRennesFrance
| | - Afshin Samali
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| |
Collapse
|
47
|
Chaperoning the Mononegavirales: Current Knowledge and Future Directions. Viruses 2018; 10:v10120699. [PMID: 30544818 PMCID: PMC6315898 DOI: 10.3390/v10120699] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 01/02/2023] Open
Abstract
The order Mononegavirales harbors numerous viruses of significant relevance to human health, including both established and emerging infections. Currently, vaccines are only available for a small subset of these viruses, and antiviral therapies remain limited. Being obligate cellular parasites, viruses must utilize the cellular machinery for their replication and spread. Therefore, targeting cellular pathways used by viruses can provide novel therapeutic approaches. One of the key challenges confronted by both hosts and viruses alike is the successful folding and maturation of proteins. In cells, this task is faced by cellular molecular chaperones, a group of conserved and abundant proteins that oversee protein folding and help maintain protein homeostasis. In this review, we summarize the current knowledge of how the Mononegavirales interact with cellular chaperones, highlight key gaps in our knowledge, and discuss the potential of chaperone inhibitors as antivirals.
Collapse
|
48
|
McGrath EP, Logue SE, Mnich K, Deegan S, Jäger R, Gorman AM, Samali A. The Unfolded Protein Response in Breast Cancer. Cancers (Basel) 2018; 10:cancers10100344. [PMID: 30248920 PMCID: PMC6211039 DOI: 10.3390/cancers10100344] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/12/2018] [Accepted: 09/18/2018] [Indexed: 01/18/2023] Open
Abstract
In 2018, in the US alone, it is estimated that 268,670 people will be diagnosed with breast cancer, and that 41,400 will die from it. Since breast cancers often become resistant to therapies, and certain breast cancers lack therapeutic targets, new approaches are urgently required. A cell-stress response pathway, the unfolded protein response (UPR), has emerged as a promising target for the development of novel breast cancer treatments. This pathway is activated in response to a disturbance in endoplasmic reticulum (ER) homeostasis but has diverse physiological and disease-specific functions. In breast cancer, UPR signalling promotes a malignant phenotype and can confer tumours with resistance to widely used therapies. Here, we review several roles for UPR signalling in breast cancer, highlighting UPR-mediated therapy resistance and the potential for targeting the UPR alone or in combination with existing therapies.
Collapse
Affiliation(s)
- Eoghan P McGrath
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Susan E Logue
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Katarzyna Mnich
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Shane Deegan
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Richard Jäger
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, 53359 Rheinbach, Germany.
| | - Adrienne M Gorman
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| | - Afshin Samali
- Apoptosis Research Centre, National University of Ireland (NUI), Galway, University Road, Galway, H91 TK33 Galway, Ireland.
- School of Natural Sciences, NUI Galway, University Road, H91 TK33 Galway, Ireland.
| |
Collapse
|
49
|
Songprakhon P, Limjindaporn T, Perng GC, Puttikhunt C, Thaingtamtanha T, Dechtawewat T, Saitornuang S, Uthaipibull C, Thongsima S, Yenchitsomanus PT, Malasit P, Noisakran S. Human glucose-regulated protein 78 modulates intracellular production and secretion of nonstructural protein 1 of dengue virus. J Gen Virol 2018; 99:1391-1406. [PMID: 30102148 DOI: 10.1099/jgv.0.001134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Virus-host interactions play important roles in virus infection and host cellular response. Several viruses, including dengue virus (DENV), usurp host chaperones to support their amplification and survival in the host cell. We investigated the interaction of nonstructural protein 1 (NS1) of DENV with three endoplasmic reticulum-resident chaperones (i.e. GRP78, calnexin and calreticulin) to delineate their functional roles and potential binding sites for protein complex formation. GRP78 protein showed prominent association with DENV NS1 in virus-infected Huh7 cells as evidenced by co-localization and co-immunoprecipitation assays. Further studies on the functional interaction of GRP78 protein were performed by using siRNA-mediated gene knockdown in a DENV replicon transfection system. GRP78 knockdown significantly decreased intracellular NS1 production and delayed NS1 secretion but had no effect on viral RNA replication. Dissecting the important domain of GRP78 required for DENV NS1 interaction showed co-immunoprecipitation of DENV NS1 with a full-length and substrate-binding domain (SBD), but not an ATPase domain, of GRP78, confirming their interaction through SBD binding. Molecular dynamics simulations of DENV NS1 and human GRP78 complex revealed their potential binding sites through hydrogen and hydrophobic bonding. The majority of GRP78-binding sites were located in a β-roll domain and connector subdomains on the DENV NS1 structure involved in hydrophobic surface formation. Taken together, our findings demonstrated the roles of human GRP78 in facilitating the intracellular production and secretion of DENV NS1 as well as predicted potential binding sites between the DENV NS1 and GRP78 complex, which could have implications in the future development of target-based antiviral drugs.
Collapse
Affiliation(s)
- Pucharee Songprakhon
- 1Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Thawornchai Limjindaporn
- 2Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Guey Chuen Perng
- 3Department of Microbiology and Immunology, College of Medicine, and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 70101, Taiwan, ROC
| | - Chunya Puttikhunt
- 4Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 10700, Thailand.,5Division of Dengue Hemorrhagic Fever Research, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | | | - Thanyaporn Dechtawewat
- 1Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sawanan Saitornuang
- 4Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 10700, Thailand
| | - Chairat Uthaipibull
- 7Protein-Ligand Engineering and Molecular Biology Laboratory, Medical Molecular Biology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Sissades Thongsima
- 8Biostatistics and Informatic Laboratory, Genome Institute, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Pa-Thai Yenchitsomanus
- 1Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Prida Malasit
- 4Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 10700, Thailand.,5Division of Dengue Hemorrhagic Fever Research, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sansanee Noisakran
- 5Division of Dengue Hemorrhagic Fever Research, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.,4Medical Biotechnology Research Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Bangkok 10700, Thailand
| |
Collapse
|
50
|
Yuan Z, Chen M, Wang J, Li Z, Geng X, Sun J. Identification of Litopenaeus vannamei BiP as a novel cellular attachment protein for white spot syndrome virus by using a biotinylation based affinity chromatography method. FISH & SHELLFISH IMMUNOLOGY 2018; 79:130-139. [PMID: 29738871 DOI: 10.1016/j.fsi.2018.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/09/2018] [Accepted: 05/02/2018] [Indexed: 06/08/2023]
Abstract
White spot syndrome virus (WSSV) is a dangerous threat to shrimp farming that also attacks a wide range of crustaceans. Knowledge of the surface protein-protein interactions between the pathogen and host is very crucial to unraveling the molecular pathogenesis mechanisms of WSSV. In this study, LvBiP (Litopenaeus vannamei immunoglobulin heavy-chain-binding protein) was identified as a novel WSSV binding protein of L. vannamei by a biotinylation based affinity chromatography method. By using pull-down and ELISA assays, the binding of recombinant LvBiP to WSSV was proved to be specific and ATP- dependent. The interaction was also confirmed by the result of co-immunoprecipitation assay. Immunofluorescence studies revealed the co-localization of LvBiP with WSSV on the cell surface of shrimp haemocytes. Additionally, LvBiP is likely to play an important role in WSSV infection. Treatment of gill cellular membrane proteins (CMPs) with purified rLvBiP and antibody that specifically recognizes LvBiP, led to a significant reduction in the binding of WSSV to gill CMPs. In the in vivo neutralization assay, rLvBiP and anti-LvBiP polyclonal antibody partially blocked the infection of WSSV. Taken together, the results indicate that LvBiP, a molecular chaperon of the HSP70 family, is a novel host factor involved at the step of attachment of the WSSV to the host cells and a potential candidate of therapeutic target.
Collapse
Affiliation(s)
- Zengzhi Yuan
- Tianjin Key Laboratory of Animal and Plant Resistance, Tianjin, 300387, PR China; College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin, 300387, PR China
| | - Meng Chen
- College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin, 300387, PR China
| | - Jingting Wang
- College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin, 300387, PR China
| | - Zhuoyu Li
- College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin, 300387, PR China
| | - Xuyun Geng
- Tianjin Center for Control and Prevention of Aquatic Animal Infectious Disease, 442 South Jiefang Road, Hexi District, Tianjin 300221, China
| | - Jinsheng Sun
- Tianjin Key Laboratory of Animal and Plant Resistance, Tianjin, 300387, PR China; College of Life Sciences, Tianjin Normal University, 393 West Binshui Road, Xiqing District, Tianjin, 300387, PR China.
| |
Collapse
|