1
|
Gao P, Ren J, Zhou Q, Chen P, Zhang A, Zhang Y, Zhou L, Ge X, Guo X, Han J, Yang H. Pseudorabies virus inhibits the unfolded protein response for viral replication during the late stages of infection. Vet Microbiol 2025; 301:110360. [PMID: 39756331 DOI: 10.1016/j.vetmic.2024.110360] [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: 09/18/2024] [Revised: 12/07/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
Pseudorabies virus (PRV) poses a significant threat to the global swine breeding industry and public health, but how the virus transverses the host defense systems for efficient viral replication and pathogenesis remains unclear. Here, we report that PRV could inhibit the unfolded protein response (UPR), a critical component of host innate immunity against viral infection, to promote virus replication during the late infection stages. PERK was shown phosphorylated and active in PRV-infected cells, but the subsequent events were suppressed post virus infection, such as eIF2α phosphorylation, ATF4 expression, and the formation of stress granules (SGs). In the meantime, although IRE1α was also active, its activated effector XBP1s was suppressed through downregulation of XBP1 mRNA levels and cleavage of XBP1s protein. Our findings also indicate that the Golgi apparatus, where ATF6 activation occur, was severely damaged in PRV-infected cells. Meanwhile, the downstream regulatory genes associated with the three UPR sensors, such as ERp60, CHOP, and EDEM1, remained silent in PRV-infected cells. Enhanced viral replication was observed post knockdown of UPR effectors ATF4 or XBP1, while stimulation with UPR activators inhibits virus replication. In conclusion, our findings address the critical question of how PRV regulates cellular UPR in favor of viral replication, and expand understanding of viruses mediated UPR suppression in general.
Collapse
Affiliation(s)
- Peng Gao
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Jianle Ren
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Qiongqiong Zhou
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Peng Chen
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Ailin Zhang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yongning Zhang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Lei Zhou
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xinna Ge
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xin Guo
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jun Han
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Hanchun Yang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
2
|
Reale A, Gatta A, Shaik AKB, Shallak M, Chiaravalli AM, Cerati M, Zaccaria M, La Rosa S, Calistri A, Accolla RS, Forlani G. An oncolytic HSV-1 vector induces a therapeutic adaptive immune response against glioblastoma. J Transl Med 2024; 22:862. [PMID: 39334370 PMCID: PMC11430576 DOI: 10.1186/s12967-024-05650-5] [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: 08/01/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most frequent and aggressive brain tumor in adults with the lowest survival rates five years post-diagnosis. Oncolytic viruses (OVs) selectively target and damage cancer cells, and for this reason they are being investigated as new therapeutic tools also against GBM. METHODS An oncolytic herpes simplex virus type 1 (oHSV-1) with deletions in the γ34.5 neurovirulence gene and the US12 gene, expressing enhanced green fluorescent protein (EGFP-oHSV-1) as reporter gene was generated and tested for its capacity to infect and kill the murine GL261 glioblastoma (GBM) cell line. Syngeneic mice were orthotopically injected with GL261cells. Seven days post-implantation, EGFP-oHSV-1 was administered intratumorally. Twenty-one days after parental tumor challenge in the opposite brain hemisphere, mice were sacrified and their brains were analysed by immunohistochemistry to assess tumor presence and cell infiltrate. RESULTS oHSV-1 replicates and induces cell death of GL261 cells in vitro. A single intracranial injection of EGFP-oHSV-1 in established GL261 tumors significantly prolongs survival in all treated mice compared to placebo treatment. Notably, 45% of treated mice became long-term survivors, and rejected GL261 cells upon rechallenge in the contralateral brain hemisphere, indicating an anamnestic antitumoral immune response. Post-mortem analysis revealed a profound modification of the tumor microenvironment with increased infiltration of CD4 + and CD8 + T lymphocytes, intertumoral vascular collapse and activation and redistribution of macrophage, microglia, and astroglia in the tumor area, with the formation of intense fibrotic tissue suggestive of complete rejection in long-term survivor mice. CONCLUSIONS EGFP-oHSV1 demonstrates potent antitumoral activity in an immunocompetent GBM model as a monotherapy, resulting from direct cell killing combined with the stimulation of a protective adaptive immune response. These results open the way to possible application of our strategy in clinical setting.
Collapse
Affiliation(s)
- Alberto Reale
- Department of Molecular Medicine, University of Padua, Padua, 35131, Italy
| | - Andrea Gatta
- Department of Medicine and Technological Innovation, University of Insubria, via O.Rossi 9, Varese, 21100, Italy
| | - Amruth Kaleem Basha Shaik
- Department of Medicine and Technological Innovation, University of Insubria, via O.Rossi 9, Varese, 21100, Italy
| | - Mariam Shallak
- Department of Medicine and Technological Innovation, University of Insubria, via O.Rossi 9, Varese, 21100, Italy
| | | | - Michele Cerati
- Unit of Pathology, ASST Sette-Laghi, Varese, 21100, Italy
| | - Martina Zaccaria
- Department of Medicine and Technological Innovation, University of Insubria, via O.Rossi 9, Varese, 21100, Italy
| | - Stefano La Rosa
- Department of Medicine and Technological Innovation, University of Insubria, via O.Rossi 9, Varese, 21100, Italy
- Unit of Pathology, ASST Sette-Laghi, Varese, 21100, Italy
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, Padua, 35131, Italy
| | - Roberto Sergio Accolla
- Department of Medicine and Technological Innovation, University of Insubria, via O.Rossi 9, Varese, 21100, Italy.
| | - Greta Forlani
- Department of Medicine and Technological Innovation, University of Insubria, via O.Rossi 9, Varese, 21100, Italy.
| |
Collapse
|
3
|
Zhou R, Wei K, Li X, Yan B, Li L. Mechanisms of ferroptosis and the relationship between ferroptosis and ER stress after JEV and HSV infection. Front Microbiol 2024; 15:1415417. [PMID: 39323885 PMCID: PMC11422203 DOI: 10.3389/fmicb.2024.1415417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/27/2024] [Indexed: 09/27/2024] Open
Abstract
Ferroptosis is a novel form of programmed cell death, which is different from apoptosis, pyroptosis and autophagy in morphology and biochemistry. Ferroptosis is characterized by condensed mitochondrial membrane densities, vanished of mitochondria crista and outer membrane rupture in morphology, and the accumulation of intracellular iron, lipid peroxidation (LPO), decrease of GSH and inhibition of GPX4 in biochemistry. Japanese encephalitis virus (JEV) and Herpes simplex virus (HSV) are both common neurotropic viruses that can cause neurological disorders, such as severe encephalitis. JEV and HSV have been demonstrated to be able to induce ferroptosis. This process is closely related to the inhibition of the GSH-GPX4 system, ACSL4 phosphorylation, and Nrf2 ubiquitination. In this review, we summarized the mechanisms by which JEV and HSV induced ferroptosis in the current study. In addition, we found a strong relationship between endoplasmic reticulum (ER) stress and ferroptosis, and we therefore speculated that sustained ER stress might be a prerequisite for ferroptosis in JEV and HSV-induced diseases.
Collapse
Affiliation(s)
- Rui Zhou
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Kexin Wei
- First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Xinyu Li
- First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Beibei Yan
- First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Lin Li
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
Chaumont L, Peruzzi M, Huetz F, Raffy C, Le Hir J, Minke J, Boudinot P, Collet B. Salmonid Double-stranded RNA-Dependent Protein Kinase Activates Apoptosis and Inhibits Protein Synthesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:700-717. [PMID: 39058317 DOI: 10.4049/jimmunol.2400076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024]
Abstract
dsRNA-dependent protein kinase R (PKR) is a key factor of innate immunity. It is involved in translation inhibition, apoptosis, and enhancement of the proinflammatory and IFN responses. However, how these antiviral functions are conserved during evolution remains largely unknown. Overexpression and knockout studies in a Chinook salmon (Oncorhynchus tshawytscha) cell line were conducted to assess the role of salmonid PKR in the antiviral response. Three distinct mRNA isoforms from a unique pkr gene, named pkr-fl (full length), pkr-ml (medium length) and pkr-sl (short length), were cloned and a pkr-/- clonal fish cell line was developed using CRISPR/Cas9 genome editing. PKR-FL includes an N-terminal dsRNA-binding domain and a C-terminal kinase domain, whereas PKR-ML and PKR-SL display a truncated or absent kinase domain, respectively. PKR-FL is induced during IFNA2 stimulation but not during viral hemorrhagic septicemia virus (VHSV) infection. Overexpression experiments showed that only PKR-FL possesses antiviral functions, including activation of apoptosis and inhibition of de novo protein synthesis. Knockout experiments confirmed that PKR is involved in apoptosis activation during the late stage of VHSV infection. Endogenous PKR also plays a critical role in translation inhibition upon poly(I:C) transfection after IFNA2 treatment. It is, however, not involved in translational arrest during VHSV infection. Extra- and intracellular titrations showed that endogenous PKR does not directly inhibit viral replication but apparently favors virion release into the supernatant, likely by triggering late apoptosis. Altogether, our data confirm that salmonid PKR has conserved molecular functions that VHSV appears to bypass with subversion strategies.
Collapse
Affiliation(s)
- Lise Chaumont
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Mathilde Peruzzi
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - François Huetz
- Unit of Antibodies in Therapy and Pathology, UMR 1222 INSERM, Institut Pasteur, Paris, France
| | | | | | | | - Pierre Boudinot
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Bertrand Collet
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| |
Collapse
|
5
|
Pérez-Martínez Z, Boga JA, Potes Y, Melón S, Coto-Montes A. Effect of Melatonin on Herpesvirus Type 1 Replication. Int J Mol Sci 2024; 25:4037. [PMID: 38612846 PMCID: PMC11012353 DOI: 10.3390/ijms25074037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Acute HSV-1 infection is associated with mild symptoms, such as fever and lesions of the mouth, face and skin. This phase is followed by a latency period before reactivation, which is associated with symptoms ranging from ulcers to encephalitis. Despite available anti-HSV-1 drugs, the development of new antiviral agents is sought due to the presence of resistant viruses. Melatonin, a molecule secreted by the pineal gland, has been shown to be an antioxidant, inducer of antioxidant enzymes, and regulator of various biological processes. Clinical trials have explored its therapeutic utility in conditions including infections. This study focuses on melatonin's role in HSV-1 replication and the underlying mechanisms. Melatonin was found to decrease the synthesis of HSV-1 proteins in infected Vero cells measured by immunofluorescence, indicating an inhibition of HSV-1 replication. Additionally, it regulates the activities of antioxidant enzymes and affects proteasome activity. Melatonin activates the unfolded protein response (UPR) and autophagy and suppresses apoptosis in HSV-1-infected cells. In summary, melatonin demonstrates an inhibitory role in HSV-1 replication by modulating various cellular responses, suggesting its potential utility in the treatment of viral infections.
Collapse
Affiliation(s)
- Zulema Pérez-Martínez
- Servicio de Microbiología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Z.P.-M.); (J.A.B.); (S.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Jose Antonio Boga
- Servicio de Microbiología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Z.P.-M.); (J.A.B.); (S.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
| | - Yaiza Potes
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Santiago Melón
- Servicio de Microbiología, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain; (Z.P.-M.); (J.A.B.); (S.M.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | - Ana Coto-Montes
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33006 Oviedo, Spain
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
6
|
Chaumont L, Collet B, Boudinot P. Double-stranded RNA-dependent protein kinase (PKR) in antiviral defence in fish and mammals. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104732. [PMID: 37172664 DOI: 10.1016/j.dci.2023.104732] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
The interferon-inducible double-stranded RNA-dependent protein kinase (PKR) is one of the key antiviral arms of the innate immune system. Upon binding of viral double stranded RNA, a viral Pattern Associated Molecular Pattern (PAMP), PKR gets activated and phosphorylates the eukaryotic translation initiation factor 2α (eIF2α) resulting in a protein shut-down that limits viral replication. Since its discovery in the mid-seventies, PKR has been shown to be involved in multiple important cellular processes including apoptosis, proinflammatory and innate immune responses. Viral subversion mechanisms of PKR underline its importance in the antiviral response of the host. PKR activation pathways and its mechanisms of action were previously identified and characterised mostly in mammalian models. However, fish Pkr and fish-specific paralogue Z-DNA-dependent protein kinase (Pkz) also play key role in antiviral defence. This review gives an update on the current knowledge on fish Pkr/Pkz, their conditions of activation and their implication in the immune responses to viruses, in comparison to their mammalian counterparts.
Collapse
Affiliation(s)
- Lise Chaumont
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, 78350, France
| | - Bertrand Collet
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, 78350, France
| | - Pierre Boudinot
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, 78350, France.
| |
Collapse
|
7
|
Zhang J, Wang J, Li M, Su X, Tian Y, Wang P, Zhou X, Jin G, Liu F. Oncolytic HSV-1 suppresses cell invasion through downregulating Sp1 in experimental glioblastoma. Cell Signal 2023; 103:110581. [PMID: 36572188 DOI: 10.1016/j.cellsig.2022.110581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Gliomas are highly aggressive intracranial tumors that are difficult to resect and have high lethality and recurrence rates. According to WHO grading criteria, glioblastoma with wild-type IDH1 has a poorer prognosis than WHO grade 4 IDH-mutant astrocytomas. To date, no effective therapeutic strategies have been developed to treat glioblastoma. Clinical trials have shown that herpes simplex virus (HSV)-1 is the safest and most efficacious oncolytic virus against glioblastoma, but the molecular antitumor mechanism of action of HSV-1 has not yet been determined. Deletion of the γ34.5 and ICP47 genes from a strain of HSV-1 yielded the oncolytic virus, oHSV-1, which reduced glioma cell viability, migration, and invasive capacity, as well as the growth of microvilli. Infected cell polypeptide 4 (ICP4) expressed by oHSV-1 was found to suppress the expression of the transcription factor Sp1, reducing the expression of host invasion-related genes. In vivo, oHSV-1 showed significant antitumor effects by suppressing the expression of Sp1 and invasion-associated genes, highly expressed in high-grade glioblastoma tissue specimens. These findings indicate that Sp1 may be a molecular marker predicting the antitumor effects of oHSV-1 in the treatment of glioma and that oHSV-1 suppresses host cell invasion through the ICP4-mediated downregulation of Sp1.
Collapse
Affiliation(s)
- Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Mingxin Li
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Xiaodong Su
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Yifu Tian
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Peiwen Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Xianzhe Zhou
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China.
| |
Collapse
|
8
|
Schlesinger M, McDonald C, Ahuja A, Alvarez Canete CA, Nuñez del Prado Z, Naipauer J, Lampidis T, Mesri EA. Glucose and mannose analogs inhibit KSHV replication by blocking N-glycosylation and inducing the unfolded protein response. J Med Virol 2023; 95:e28314. [PMID: 36380418 PMCID: PMC9839548 DOI: 10.1002/jmv.28314] [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: 07/26/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent for Kaposi's sarcoma (KS), an HIV/AIDS-associated malignancy. Effective treatments against KS remain to be developed. The sugar analog 2-deoxy- d-glucose (2-DG) is an anticancer agent that is well-tolerated and safe in patients and was recently demonstrated to be a potent antiviral, including KSHV and severe acute respiratory syndrome coronavirus 2. Because 2-DG inhibits glycolysis and N-glycosylation, identifying its molecular targets is challenging. Here we compare the antiviral effect of 2-DG with 2-fluoro-deoxy- d-glucose, a glycolysis inhibitor, and 2-deoxy-fluoro- d-mannose (2-DFM), a specific N-glycosylation inhibitor. At doses similar to those clinically achievable with 2-DG, the three drugs impair KSHV replication and virion production in iSLK.219 cells via downregulation of viral structural glycoprotein expression (K8.1 and gB), being 2-DFM the most potent KSHV inhibitor. Consistently with the higher potency of 2-DFM, we found that d-mannose rescues KSHV glycoprotein synthesis and virus production, indicating that inhibition of N-glycosylation is the main antiviral target using d-mannose competition experiments. Suppression of N-glycosylation by the sugar drugs triggers ER stress. It activates the host unfolded protein response (UPR), counteracting KSHV-induced inhibition of the protein kinase R-like endoplasmic reticulum kinase branch, particularly activating transcription factor 4 and C/EBP homologous protein expression. Finally, we demonstrate that sugar analogs induce autophagy (a prosurvival mechanism) and, thus, inhibit viral replication playing a protective role against KSHV-induced cell death, further supporting their direct antiviral effect and potential therapeutic use. Our work identifies inhibition of N-glycosylation leading to ER stress and UPR as an antienveloped virus target and sugar analogs such as 2-DG and the newly identified 2-DFM as antiviral drugs.
Collapse
Affiliation(s)
- Mariana Schlesinger
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Microbiology and ImmunologyMiami Center for AIDS ResearchMIAMIFloridaUSA
| | - Christian McDonald
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Microbiology and ImmunologyMiami Center for AIDS ResearchMIAMIFloridaUSA
| | - Anuj Ahuja
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Microbiology and ImmunologyMiami Center for AIDS ResearchMIAMIFloridaUSA
| | - Carolina Alejandra Alvarez Canete
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Microbiology and ImmunologyMiami Center for AIDS ResearchMIAMIFloridaUSA
| | - Zelmira Nuñez del Prado
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Microbiology and ImmunologyMiami Center for AIDS ResearchMIAMIFloridaUSA
| | - Julian Naipauer
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Microbiology and ImmunologyMiami Center for AIDS ResearchMIAMIFloridaUSA
- UM‐CFAR/SCCC Argentina Consortium for AIDS MalignanciesMiamiFloridaUSA
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE)CONICET‐Universidad de Buenos AiresBuenos AiresArgentina
| | - Theodore Lampidis
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Cell BiologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Enrique A. Mesri
- Tumor Biology ProgramSylvester Comprehensive Cancer CenterMiamiFloridaUSA
- Department of Microbiology and ImmunologyMiami Center for AIDS ResearchMIAMIFloridaUSA
- UM‐CFAR/SCCC Argentina Consortium for AIDS MalignanciesMiamiFloridaUSA
| |
Collapse
|
9
|
Endoplasmic Reticulum Stress in Hepatitis B Virus and Hepatitis C Virus Infection. Viruses 2022; 14:v14122630. [PMID: 36560634 PMCID: PMC9780809 DOI: 10.3390/v14122630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Endoplasmic reticulum (ER) stress, a type of cellular stress, always occurs when unfolded or misfolded proteins accumulating in the ER exceed the protein folding capacity. Because of the demand for rapid viral protein synthesis after viral infection, viral infections become a risk factor for ER stress. The hepatocyte is a cell with large and well-developed ER, and hepatitis virus infection is widespread in the population, indicating the interaction between hepatitis viruses and ER stress may have significance for managing liver diseases. In this paper, we review the process that is initiated by the hepatocyte through ER stress against HBV and HCV infection and explain how this information can be helpful in the treatment of HBV/HCV-related diseases.
Collapse
|
10
|
Henke W, Waisner H, Arachchige SP, Kalamvoki M, Stephens E. The envelope proteins from SARS-CoV-2 and SARS-CoV potently reduce the infectivity of human immunodeficiency virus type 1 (HIV-1). Retrovirology 2022; 19:25. [PMID: 36403071 PMCID: PMC9675205 DOI: 10.1186/s12977-022-00611-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Viroporins are virally encoded ion channels involved in virus assembly and release. Human immunodeficiency virus type 1 (HIV-1) and influenza A virus encode for viroporins. The human coronavirus SARS-CoV-2 encodes for at least two viroporins, a small 75 amino acid transmembrane protein known as the envelope (E) protein and a larger 275 amino acid protein known as Orf3a. Here, we compared the replication of HIV-1 in the presence of four different β-coronavirus E proteins. RESULTS We observed that the SARS-CoV-2 and SARS-CoV E proteins reduced the release of infectious HIV-1 yields by approximately 100-fold while MERS-CoV or HCoV-OC43 E proteins restricted HIV-1 infectivity to a lesser extent. Mechanistically, neither reverse transcription nor mRNA synthesis was involved in the restriction. We also show that all four E proteins caused phosphorylation of eIF2-α at similar levels and that lipidation of LC3-I could not account for the differences in restriction. However, the level of caspase 3 activity in transfected cells correlated with HIV-1 restriction in cells. Finally, we show that unlike the Vpu protein of HIV-1, the four E proteins did not significantly down-regulate bone marrow stromal cell antigen 2 (BST-2). CONCLUSIONS The results of this study indicate that while viroporins from homologous viruses can enhance virus release, we show that a viroporin from a heterologous virus can suppress HIV-1 protein synthesis and release of infectious virus.
Collapse
Affiliation(s)
- Wyatt Henke
- Department of Microbiology, Molecular Genetics and ImmunologyUniversity of Kansas Medical Center, 2000 Hixon Hall 3901 Rainbow Blvd, Kansas, KS 66160 USA
| | - Hope Waisner
- Department of Microbiology, Molecular Genetics and ImmunologyUniversity of Kansas Medical Center, 2000 Hixon Hall 3901 Rainbow Blvd, Kansas, KS 66160 USA
| | - Sachith Polpitiya Arachchige
- Department of Microbiology, Molecular Genetics and ImmunologyUniversity of Kansas Medical Center, 2000 Hixon Hall 3901 Rainbow Blvd, Kansas, KS 66160 USA
| | - Maria Kalamvoki
- Department of Microbiology, Molecular Genetics and ImmunologyUniversity of Kansas Medical Center, 2000 Hixon Hall 3901 Rainbow Blvd, Kansas, KS 66160 USA
| | - Edward Stephens
- Department of Microbiology, Molecular Genetics and ImmunologyUniversity of Kansas Medical Center, 2000 Hixon Hall 3901 Rainbow Blvd, Kansas, KS 66160 USA
| |
Collapse
|
11
|
Vaccinium bracteatum Thunb Extract Inhibits HSV-1 Infection by Regulating ER Stress and Apoptosis. Antioxidants (Basel) 2022; 11:antiox11091773. [PMID: 36139847 PMCID: PMC9495922 DOI: 10.3390/antiox11091773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/25/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022] Open
Abstract
Herpes simplex Type 1 (HSV-1) is a neurotropic virus that infects the peripheral and central nervous system. Usually, after primary infection in epithelial cells, HSV-1 migrates retrograde to the peripheral nervous system (PNS), where it establishes a latent infection. HSV-1 can remain latent in the nervous system, and its reactivation in the brain can rarely cause acute HSV-1 encephalitis, often a life-threatening condition, or asymptomatic reactivations that could lead to neuronal damage and ultimately neurodegenerative disorders. Acyclovir and related nucleoside analogs have been used as therapeutic agents for HSV-1 infection, but resistance to the drug can arise, and the protective effect of HSV-1 on brain cells is limited. Therefore, there is an urgent need for research into safe and effective new antiviral agents that can protect brain cells from the damage that is caused by HSV-1 infection. Vaccinium bracteatum Thunb. (VBT) is widely distributed in Korea and China, and has pharmacological actions such as anti-inflammatory, antioxidant, and antidiabetic activity. Studies on the antiviral effect of VBT on HSV-1 infection have not been reported so far. Therefore, we sought to determine the HSV-1 antiviral effect and molecular mechanism of VBT at the cellular level. We confirmed that VBT repressed the VP16 and IE genes in both Vero and SK-N-SH cells. We also found that the generation of HSV-1 virions was inhibited by VBT treatment. VBT inhibited the activities of the HSV-1-induced endoplasmic reticulum (ER) stressors PERK, ATF4, and CHOP. We confirmed that VBT inhibited the activity of apoptosis factors by regulating the expression of death receptor (DR) after HSV-1 infection. As HSV-1 is closely associated with brain diseases, the study of the antiviral drug effects and mechanism of VBT is meaningful. Further studies using animal models of infection will also be performed to determine the potential of VBT as an antiviral agent.
Collapse
|
12
|
Tognarelli EI, Retamal-Díaz A, Farías MA, Duarte LF, Palomino TF, Ibañez FJ, Riedel CA, Kalergis AM, Bueno SM, González PA. Pharmacological Inhibition of IRE-1 Alpha Activity in Herpes Simplex Virus Type 1 and Type 2-Infected Dendritic Cells Enhances T Cell Activation. Front Immunol 2022; 12:764861. [PMID: 35069537 PMCID: PMC8766714 DOI: 10.3389/fimmu.2021.764861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/06/2021] [Indexed: 12/25/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) infections are life-long and highly prevalent in the human population. These viruses persist in the host, eliciting either symptomatic or asymptomatic infections that may occur sporadically or in a recurrent manner through viral reactivations. Clinical manifestations due to symptomatic infection may be mild such as orofacial lesions, but may also translate into more severe diseases, such as ocular infections that may lead to blindness and life-threatening encephalitis. A key feature of herpes simplex viruses (HSVs) is that they have evolved molecular determinants that hamper numerous components of the host’s antiviral innate and adaptive immune system. Importantly, HSVs infect and negatively modulate the function of dendritic cells (DCs), by inhibiting their T cell-activating capacity and eliciting their apoptosis after infection. Previously, we reported that HSV-2 activates the splicing of the mRNA of XBP1, which is related to the activity of the unfolded protein response (UPR) factor Inositol-Requiring Enzyme 1 alpha (IRE-1α). Here, we sought to evaluate if the activation of the IRE-1α pathway in DCs upon HSV infection may be related to impaired DC function after infection with HSV-1 or HSV-2. Interestingly, the pharmacological inhibition of the endonuclease activity of IRE-1α in HSV-1- and HSV-2-infected DCs significantly reduced apoptosis in these cells and enhanced their capacity to migrate to lymph nodes and activate virus-specific CD4+ and CD8+ T cells. These findings suggest that the activation of the IRE-1α-dependent UPR pathway in HSV-infected DCs may play a significant role in the negative effects that these viruses exert over these cells and that the modulation of this signaling pathway may be relevant for enhancing the function of DCs upon infection with HSVs.
Collapse
Affiliation(s)
- Eduardo I Tognarelli
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Biotecnología, Facultad de Ciencias del Mar y de Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tomás F Palomino
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco J Ibañez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Biología Celular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
13
|
Wu Y, Zhang Z, Li Y, Li Y. The Regulation of Integrated Stress Response Signaling Pathway on Viral Infection and Viral Antagonism. Front Microbiol 2022; 12:814635. [PMID: 35222313 PMCID: PMC8874136 DOI: 10.3389/fmicb.2021.814635] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
The integrated stress response (ISR) is an adaptational signaling pathway induced in response to different stimuli, such as accumulation of unfolded and misfolded proteins, hypoxia, amino acid deprivation, viral infection, and ultraviolet light. It has been known that viral infection can activate the ISR, but the role of the ISR during viral infection is still unclear. In some cases, the ISR is a protective mechanism of host cells against viral infection, while viruses may hijack the ISR for facilitating their replication. This review highlighted recent advances on the induction of the ISR upon viral infection and the downstream responses, such as autophagy, apoptosis, formation of stress granules, and innate immunity response. We then discussed the molecular mechanism of the ISR regulating viral replication and how viruses antagonize this cellular stress response resulting from the ISR.
Collapse
Affiliation(s)
- Yongshu Wu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhidong Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
| | - Yanmin Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China
- *Correspondence: Yanmin Li,
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
14
|
Zhu T, Jiang X, Xin H, Zheng X, Xue X, Chen JL, Qi B. GADD34-mediated dephosphorylation of eIF2α facilitates pseudorabies virus replication by maintaining de novo protein synthesis. Vet Res 2021; 52:148. [PMID: 34930429 PMCID: PMC8686791 DOI: 10.1186/s13567-021-01018-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 11/10/2022] Open
Abstract
Viruses have evolved multiple strategies to manipulate their host's translational machinery for the synthesis of viral proteins. A common viral target is the alpha subunit of eukaryotic initiation factor 2 (eIF2α). In this study, we show that global protein synthesis was increased but the eIF2α phosphorylation level was markedly decreased in porcine kidney 15 (PK15) cells infected with pseudorabies virus (PRV), a swine herpesvirus. An increase in the eIF2α phosphorylation level by salubrinal treatment or transfection of constructs expressing wild-type eIF2α or an eIF2α phosphomimetic [eIF2α(S51D)] attenuated global protein synthesis and suppressed PRV replication. To explore the mechanism involved in the inhibition of eIF2α phosphorylation during PRV infection, we examined the phosphorylation status of protein kinase R-like endoplasmic reticulum kinase (PERK) and double-stranded RNA-dependent protein kinase R (PKR), two kinases that regulate eIF2α phosphorylation during infection with numerous viruses. We found that the level of neither phosphorylated (p)-PERK nor p-PKR was altered in PRV-infected cells or the lungs of infected mice. However, the expression of growth arrest and DNA damage-inducible protein 34 (GADD34), which promotes eIF2α dephosphorylation by recruiting protein phosphatase 1 (PP1), was significantly induced both in vivo and in vitro. Knockdown of GADD34 and inhibition of PP1 activity by okadaic acid treatment led to increased eIF2α phosphorylation but significantly suppressed global protein synthesis and inhibited PRV replication. Collectively, these results demonstrated that PRV induces GADD34 expression to promote eIF2α dephosphorylation, thereby maintaining de novo protein synthesis and facilitating viral replication.
Collapse
Affiliation(s)
- Ting Zhu
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China.
| | - Xueli Jiang
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hangkuo Xin
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaohui Zheng
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaonuan Xue
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Baomin Qi
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
15
|
Ming SL, Zhang S, Wang Q, Zeng L, Zhou LY, Wang MD, Ma YX, Han LQ, Zhong K, Zhu HS, Bai YL, Yang GY, Wang J, Chu BB. Inhibition of USP14 influences alphaherpesvirus proliferation by degrading viral VP16 protein via ER stress-triggered selective autophagy. Autophagy 2021; 18:1801-1821. [PMID: 34822318 DOI: 10.1080/15548627.2021.2002101] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Alphaherpesvirus infection results in severe health consequences in a wide range of hosts. USPs are the largest subfamily of deubiquitinating enzymes that play critical roles in immunity and other cellular functions. To investigate the role of USPs in alphaherpesvirus replication, we assessed 13 USP inhibitors for PRV replication. Our data showed that all the tested compounds inhibited PRV replication, with the USP14 inhibitor b-AP15 exhibiting the most dramatic effect. Ablation of USP14 also influenced PRV replication, whereas replenishment of USP14 in USP14 null cells restored viral replication. Although inhibition of USP14 induced the K63-linked ubiquitination of PRV VP16 protein, its degradation was not dependent on the proteasome. USP14 directly bound to ubiquitin chains on VP16 through its UBL domain during the early stage of viral infection. Moreover, USP14 inactivation stimulated EIF2AK3/PERK- and ERN1/IRE1-mediated signaling pathways, which were responsible for VP16 degradation through SQSTM1/p62-mediated selective macroautophagy/autophagy. Ectopic expression of non-ubiquitinated VP16 fully rescued PRV replication. Challenge of mice with b-AP15 activated ER stress and autophagy and inhibited PRV infection in vivo. Our results suggested that USP14 was a potential therapeutic target to treat alphaherpesvirus-induced infectious diseases.
Collapse
Affiliation(s)
- Sheng-Li Ming
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Shuang Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Qi Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Lu-Yu Zhou
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Meng-Di Wang
- The Education Department of Henan Province Henan University of Animal Husbandry and Economy, Zhengzhou, Henan Province, People's Republic of China
| | - Ying-Xian Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Li-Qiang Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Kai Zhong
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - He-Shui Zhu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Yi-Lin Bai
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi Province, People's Republic of China
| | - Guo-Yu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China.,International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China.,Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou, Henan Province, China.,Key Laboratory of Animal Growth and Development, Zhengzhou, Henan Province, People's Republic of China.,International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, Henan Province, People's Republic of China
| |
Collapse
|
16
|
Prasad V, Greber UF. The endoplasmic reticulum unfolded protein response - homeostasis, cell death and evolution in virus infections. FEMS Microbiol Rev 2021; 45:fuab016. [PMID: 33765123 PMCID: PMC8498563 DOI: 10.1093/femsre/fuab016] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Viruses elicit cell and organismic stress, and offset homeostasis. They trigger intrinsic, innate and adaptive immune responses, which limit infection. Viruses restore homeostasis by harnessing evolutionary conserved stress responses, such as the endoplasmic reticulum (ER) unfolded protein response (UPRER). The canonical UPRER restores homeostasis based on a cell-autonomous signalling network modulating transcriptional and translational output. The UPRER remedies cell damage, but upon severe and chronic stress leads to cell death. Signals from the UPRER flow along three branches with distinct stress sensors, the inositol requiring enzyme (Ire) 1, protein kinase R (PKR)-like ER kinase (PERK), and the activating transcription factor 6 (ATF6). This review shows how both enveloped and non-enveloped viruses use the UPRER to control cell stress and metabolic pathways, and thereby enhance infection and progeny formation, or undergo cell death. We highlight how the Ire1 axis bypasses apoptosis, boosts viral transcription and maintains dormant viral genomes during latency and persistence periods concurrent with long term survival of infected cells. These considerations open new options for oncolytic virus therapies against cancer cells where the UPRER is frequently upregulated. We conclude with a discussion of the evolutionary impact that viruses, in particular retroviruses, and anti-viral defense has on the UPRER.
Collapse
Affiliation(s)
- Vibhu Prasad
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
17
|
Shahryari A, Burtscher I, Nazari Z, Lickert H. Engineering Gene Therapy: Advances and Barriers. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alireza Shahryari
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- School of Medicine Department of Human Genetics Technical University of Munich Klinikum Rechts der Isar 81675 München Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
- Stem Cell Research Center Golestan University of Medical Sciences Gorgan 49341‐74515 Iran
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
| | - Zahra Nazari
- Department of Biology School of Basic Sciences Golestan University Gorgan 49361‐79142 Iran
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research Helmholtz Zentrum München 85764 Neuherberg Germany
- School of Medicine Department of Human Genetics Technical University of Munich Klinikum Rechts der Isar 81675 München Germany
- Institute of Stem Cell Research Helmholtz Zentrum München 85764 Neuherberg Germany
| |
Collapse
|
18
|
Zhao Y, Hu N, Jiang Q, Zhu L, Zhang M, Jiang J, Xiong M, Yang M, Yang J, Shen L, Zhang S, Niu L, Chen L, Chen D. Protective effects of sodium butyrate on rotavirus inducing endoplasmic reticulum stress-mediated apoptosis via PERK-eIF2α signaling pathway in IPEC-J2 cells. J Anim Sci Biotechnol 2021; 12:69. [PMID: 34112268 PMCID: PMC8194137 DOI: 10.1186/s40104-021-00592-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rotavirus (RV) is a major pathogen that causes severe gastroenteritis in infants and young animals. Endoplasmic reticulum (ER) stress and subsequent apoptosis play pivotal role in virus infection. However, the protective mechanisms of intestinal damage caused by RV are poorly defined, especially the molecular pathways related to enterocytes apoptosis. Thus, the aim of this study was to investigate the protective effect and mechanism of sodium butyrate (SB) on RV-induced apoptosis of IPEC-J2 cells. RESULTS The RV infection led to significant cell apoptosis, increased the expression levels of ER stress (ERS) markers, phosphorylated protein kinase-like ER kinase (PERK), eukaryotic initiation factor 2 alpha (eIF2α), caspase9, and caspase3. Blocking PERK pathway using specific inhibitor GSK subsequently reversed RV-induced cell apoptosis. The SB treatment significantly inhibited RV-induced ERS by decreasing the expression of glucose regulated protein 78 (GRP78), PERK, and eIF2α. In addition, SB treatment restrained the ERS-mediated apoptotic pathway, as indicated by downregulation of C/EBP homologous protein (CHOP) mRNA level, as well as decreased cleaved caspase9 and caspase3 protein levels. Furthermore, siRNA-induced GPR109a knockdown significantly suppressed the protective effect of SB on RV-induced cell apoptosis. CONCLUSIONS These results indicate that SB exerts protective effects against RV-induced cell apoptosis through inhibiting ERS mediated apoptosis by regulating PERK-eIF2α signaling pathway via GPR109a, which provide new ideas for the prevention and control of RV.
Collapse
Affiliation(s)
- Ye Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China.
| | - Ningming Hu
- Institute of Animal Nutrition, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China.,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China
| | - Qin Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China.
| | - Ming Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Jun Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Manyi Xiong
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Mingxian Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Jiandong Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Lili Niu
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Lei Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Huimin Road 211#, Chengdu, Sichuan Province, 611130, P. R. China. .,Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, P. R. China.
| |
Collapse
|
19
|
Dahal B, Lehman CW, Akhrymuk I, Bracci NR, Panny L, Barrera MD, Bhalla N, Jacobs JL, Dinman JD, Kehn-Hall K. PERK Is Critical for Alphavirus Nonstructural Protein Translation. Viruses 2021; 13:892. [PMID: 34065980 PMCID: PMC8151226 DOI: 10.3390/v13050892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) is an alphavirus that causes encephalitis. Previous work indicated that VEEV infection induced early growth response 1 (EGR1) expression, leading to cell death via the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) arm of the unfolded protein response (UPR) pathway. Loss of PERK prevented EGR1 induction and decreased VEEV-induced death. The results presented within show that loss of PERK in human primary astrocytes dramatically reduced VEEV and eastern equine encephalitis virus (EEEV) infectious titers by 4-5 log10. Loss of PERK also suppressed VEEV replication in primary human pericytes and human umbilical vein endothelial cells, but it had no impact on VEEV replication in transformed U87MG and 293T cells. A significant reduction in VEEV RNA levels was observed as early as 3 h post-infection, but viral entry assays indicated that the loss of PERK minimally impacted VEEV entry. In contrast, the loss of PERK resulted in a dramatic reduction in viral nonstructural protein translation and negative-strand viral RNA production. The loss of PERK also reduced the production of Rift Valley fever virus and Zika virus infectious titers. These data indicate that PERK is an essential factor for the translation of alphavirus nonstructural proteins and impacts multiple RNA viruses, making it an exciting target for antiviral development.
Collapse
Affiliation(s)
- Bibha Dahal
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
| | - Caitlin W. Lehman
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Ivan Akhrymuk
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Nicole R. Bracci
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Lauren Panny
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Michael D. Barrera
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
| | - Nishank Bhalla
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
| | | | - Jonathan D. Dinman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA 20110, USA; (B.D.); (C.W.L.); (I.A.); (N.R.B.); (L.P.); (M.D.B.); (N.B.)
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
20
|
Ghose J, Dona A, Murtadha M, Gunes EG, Caserta E, Yoo JY, Russell L, Jaime-Ramirez AC, Barwick BG, Gupta VA, Sanchez JF, Sborov DW, Rosen ST, Krishnan A, Boise LH, Kaur B, Hofmeister CC, Pichiorri F. Oncolytic herpes simplex virus infects myeloma cells in vitro and in vivo. MOLECULAR THERAPY-ONCOLYTICS 2021; 20:519-531. [PMID: 33738338 PMCID: PMC7940704 DOI: 10.1016/j.omto.2021.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/15/2021] [Indexed: 12/21/2022]
Abstract
Because most patients with multiple myeloma (MM) develop resistance to current regimens, novel approaches are needed. Genetically modified, replication-competent oncolytic viruses exhibit high tropism for tumor cells regardless of cancer stage and prior treatment. Receptors of oncolytic herpes simplex virus 1 (oHSV-1), NECTIN-1, and HVEM are expressed on MM cells, prompting us to investigate the use of oHSV-1 against MM. Using oHSV-1-expressing GFP, we found a dose-dependent increase in the GFP+ signal in MM cell lines and primary MM cells. Whereas NECTIN-1 expression is variable among MM cells, we discovered that HVEM is ubiquitously and highly expressed on all samples tested. Expression of HVEM was consistently higher on CD138+/CD38+ plasma cells than in non-plasma cells. HVEM blocking demonstrated the requirement of this receptor for infection. However, we observed that, although oHSV-1 could efficiently infect and kill all MM cell lines tested, no viral replication occurred. Instead, we identified that oHSV-1 induced MM cell apoptosis via caspase-3 cleavage. We further noted that oHSV-1 yielded a significant decrease in tumor volume in two mouse xenograft models. Therefore, oHSV-1 warrants exploration as a novel potentially effective treatment option in MM, and HVEM should be investigated as a possible therapeutic target.
Collapse
Affiliation(s)
- Jayeeta Ghose
- Department of Radiation Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ada Dona
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| | - Mariam Murtadha
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| | - Emine Gulsen Gunes
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| | - Enrico Caserta
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| | - Ji Young Yoo
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Luke Russell
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Benjamin G Barwick
- Department of Hematology & Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30307, USA
| | - Vikas A Gupta
- Department of Hematology & Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30307, USA
| | - James F Sanchez
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| | - Douglas W Sborov
- Division of Hematology & Hematologic Malignancies, Department of Internal Medicine, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Steven T Rosen
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| | - Lawrence H Boise
- Department of Hematology & Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30307, USA
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Craig C Hofmeister
- Department of Hematology & Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30307, USA
| | - Flavia Pichiorri
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Monrovia, CA 91016, USA
| |
Collapse
|
21
|
Su A, Wang H, Zheng D, Wu Z. TUDCA inhibits HSV-1 replication by the modulating unfolded protein response pathway. J Med Virol 2020; 92:3628-3637. [PMID: 32356915 DOI: 10.1002/jmv.25963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/29/2020] [Indexed: 01/12/2023]
Abstract
Tauroursodeoxycholic acid (TUDCA), an endogenous bile acid, was used to protect liver function through antiapoptosis or reducing endoplasmic reticulum stress (ER stress). Previous studies showed that ER stress was modulated by herpes simplex virus types 1 (HSV-1) infection to facilitate viral replication. Here, we investigated the effect of TUDCA on HSV-1 infection of HEC-1-A cells and showed that both replication and multiplication of the virus were inhibited by TUDCA in a dose dependent manner. Unfolded protein response was induced to deliver stress signals from ER to nucleus. We found that TUDCA alleviated activating transcription factor 6 branch inhibition, partially enhanced protein kinase RNA-like ER kinase pathway activation, and repressed inositol-requiring protein 1α arm activation significantly in infected cells. The findings of this study suggest that TUDCA inhibits HSV-1 replication through ER stress pathway, which may provide a potential therapeutic strategy for HSV-1 infection.
Collapse
Affiliation(s)
- Airong Su
- Clinical Molecular Diagnostic Laboratory, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- School of Life Sciences, Ningxia University, Yinchuan, China
| | - Huanru Wang
- School of Life Sciences, Ningxia University, Yinchuan, China
| | - Datong Zheng
- Clinical Molecular Diagnostic Laboratory, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Children's Health Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The Second Clinical School, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhiwei Wu
- School of Life Sciences, Ningxia University, Yinchuan, China
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
- State Key Lab of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
22
|
Zhu H, Zheng C. The Race between Host Antiviral Innate Immunity and the Immune Evasion Strategies of Herpes Simplex Virus 1. Microbiol Mol Biol Rev 2020; 84:e00099-20. [PMID: 32998978 PMCID: PMC7528619 DOI: 10.1128/mmbr.00099-20] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) is very successful in establishing acute and latent infections in humans by counteracting host antiviral innate immune responses. HSV-1 has evolved various strategies to evade host antiviral innate immunity and some cellular survival-associated pathways. Since there is still no vaccine available for HSV-1, a continuous update of information regarding the interaction between HSV-1 infection and the host antiviral innate immunity will provide novel insights to develop new therapeutic strategies for HSV-1 infection and its associated diseases. Here, we update recent studies about how HSV-1 evades the host antiviral innate immunity, specifically how HSV-1 proteins directly or indirectly target the adaptors in the antiviral innate immunity signaling pathways to downregulate the signal transduction. Additionally, some classical intracellular stress responses, which also play important roles in defense of viral invasion, will be discussed here. With a comprehensive review of evasion mechanisms of antiviral innate immunity by HSV-1, we will be able to develop potential new targets for therapies and a possible vaccine against HSV-1 infections.
Collapse
Affiliation(s)
- Huifang Zhu
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Neonatal/Pediatric Intensive Care Unit, Children's Medical Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
The Role of Molecular Chaperones in Virus Infection and Implications for Understanding and Treating COVID-19. J Clin Med 2020; 9:jcm9113518. [PMID: 33143379 PMCID: PMC7693988 DOI: 10.3390/jcm9113518] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/24/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
The COVID-19 pandemic made imperative the search for means to end it, which requires a knowledge of the mechanisms underpinning the multiplication and spread of its cause, the coronavirus SARS-CoV-2. Many viruses use members of the hosts’ chaperoning system to infect the target cells, replicate, and spread, and here we present illustrative examples. Unfortunately, the role of chaperones in the SARS-CoV-2 cycle is still poorly understood. In this review, we examine the interactions of various coronaviruses during their infectious cycle with chaperones in search of information useful for future research on SARS-CoV-2. We also call attention to the possible role of molecular mimicry in the development of autoimmunity and its widespread pathogenic impact in COVID-19 patients. Viral proteins share highly antigenic epitopes with human chaperones, eliciting anti-viral antibodies that crossreact with the chaperones. Both, the critical functions of chaperones in the infectious cycle of viruses and the possible role of these molecules in COVID-19 autoimmune phenomena, make clear that molecular chaperones are promising candidates for the development of antiviral strategies. These could consist of inhibiting-blocking those chaperones that are necessary for the infectious viral cycle, or those that act as autoantigens in the autoimmune reactions causing generalized destructive effects on human tissues.
Collapse
|
24
|
Wang S, Ma X, Wang H, He H. Induction of the Unfolded Protein Response during Bovine Alphaherpesvirus 1 Infection. Viruses 2020; 12:v12090974. [PMID: 32887282 PMCID: PMC7552016 DOI: 10.3390/v12090974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022] Open
Abstract
Bovine herpesvirus 1 (BoHV-1) is an alphaherpesvirus that causes great economic losses in the cattle industry. Herpesvirus infection generally induces endoplasmic reticulum (ER) stress, and the unfolded protein response (UPR) in infected cells. However, it is not clear whether ER stress and UPR can be induced by BoHV-1 infection. Here, we found that ER stress induced by BoHV-1 infection could activate all three UPR sensors (the activating transcription factor 6 (ATF6), the inositol-requiring enzyme 1 (IRE1), and the protein kinase RNA-like ER kinase (PERK)) in MDBK cells. During BoHV-1 infection, the ATF6 pathway of UPR did not affect viral replication. However, both knockdown and specific chemical inhibition of PERK attenuated the BoHV-1 proliferation, and chemical inhibition of PERK significantly reduced the viral replication at the post-entry step of the BoHV-1 life cycle. Furthermore, knockdown of IRE1 inhibits BoHV-1 replication, indicating that the IRE1 pathway may promote viral replication. Further study revealed that BoHV-1 replication was enhanced by IRE1 RNase activity inhibition at the stage of virus post-entry in MDBK cells. Furthermore, IRE1 kinase activity inhibition and RNase activity enhancement decrease BoHV1 replication via affecting the virus post-entry step. Our study revealed that BoHV-1 infection activated all three UPR signaling pathways in MDBK cells, and BoHV-1-induced PERK and IRE1 pathways may promote viral replication. This study provides a new perspective for the interactions of BoHV-1 and UPR, which is helpful to further elucidate the mechanism of BoHV-1 pathogenesis.
Collapse
Affiliation(s)
- Song Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250014, China; (S.W.); (X.M.)
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Xiaomei Ma
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250014, China; (S.W.); (X.M.)
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Hongmei Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250014, China; (S.W.); (X.M.)
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
- Correspondence: (H.W.); (H.H.)
| | - Hongbin He
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan 250014, China; (S.W.); (X.M.)
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
- Correspondence: (H.W.); (H.H.)
| |
Collapse
|
25
|
Gao Z, Xie W, Fan C, Cao Y. Non-ionizing radiofrequency field induces unfolded protein response (UPR) in endoplasmic reticulum of mouse neuronal cells. RADIATION MEDICINE AND PROTECTION 2020. [DOI: 10.1016/j.radmp.2020.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
26
|
Wan Q, Song D, Li H, He ML. Stress proteins: the biological functions in virus infection, present and challenges for target-based antiviral drug development. Signal Transduct Target Ther 2020; 5:125. [PMID: 32661235 PMCID: PMC7356129 DOI: 10.1038/s41392-020-00233-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/26/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
Stress proteins (SPs) including heat-shock proteins (HSPs), RNA chaperones, and ER associated stress proteins are molecular chaperones essential for cellular homeostasis. The major functions of HSPs include chaperoning misfolded or unfolded polypeptides, protecting cells from toxic stress, and presenting immune and inflammatory cytokines. Regarded as a double-edged sword, HSPs also cooperate with numerous viruses and cancer cells to promote their survival. RNA chaperones are a group of heterogeneous nuclear ribonucleoproteins (hnRNPs), which are essential factors for manipulating both the functions and metabolisms of pre-mRNAs/hnRNAs transcribed by RNA polymerase II. hnRNPs involve in a large number of cellular processes, including chromatin remodelling, transcription regulation, RNP assembly and stabilization, RNA export, virus replication, histone-like nucleoid structuring, and even intracellular immunity. Dysregulation of stress proteins is associated with many human diseases including human cancer, cardiovascular diseases, neurodegenerative diseases (e.g., Parkinson’s diseases, Alzheimer disease), stroke and infectious diseases. In this review, we summarized the biologic function of stress proteins, and current progress on their mechanisms related to virus reproduction and diseases caused by virus infections. As SPs also attract a great interest as potential antiviral targets (e.g., COVID-19), we also discuss the present progress and challenges in this area of HSP-based drug development, as well as with compounds already under clinical evaluation.
Collapse
Affiliation(s)
- Qianya Wan
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Dan Song
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Huangcan Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China. .,CityU Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
27
|
Multiple Herpes Simplex Virus-1 (HSV-1) Reactivations Induce Protein Oxidative Damage in Mouse Brain: Novel Mechanisms for Alzheimer's Disease Progression. Microorganisms 2020; 8:microorganisms8070972. [PMID: 32610629 PMCID: PMC7409037 DOI: 10.3390/microorganisms8070972] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 12/24/2022] Open
Abstract
Compelling evidence supports the role of oxidative stress in Alzheimer's disease (AD) pathophysiology. Interestingly, Herpes simplex virus-1 (HSV-1), a neurotropic virus that establishes a lifelong latent infection in the trigeminal ganglion followed by periodic reactivations, has been reportedly linked both to AD and to oxidative stress conditions. Herein, we analyzed, through biochemical and redox proteomic approaches, the mouse model of recurrent HSV-1 infection we previously set up, to investigate whether multiple virus reactivations induced oxidative stress in the mouse brain and affected protein function and related intracellular pathways. Following multiple HSV-1 reactivations, we found in mouse brains increased levels of oxidative stress hallmarks, including 4-hydroxynonenal (HNE), and 13 HNE-modified proteins whose levels were found significantly altered in the cortex of HSV-1-infected mice compared to controls. We focused on two proteins previously linked to AD pathogenesis, i.e., glucose-regulated protein 78 (GRP78) and collapsin response-mediated protein 2 (CRMP2), which are involved in the unfolded protein response (UPR) and in microtubule stabilization, respectively. We found that recurrent HSV-1 infection disables GRP78 function and activates the UPR, whereas it prevents CRMP2 function in mouse brains. Overall, these data suggest that repeated HSV-1 reactivation into the brain may contribute to neurodegeneration also through oxidative damage.
Collapse
|
28
|
Ren S, Ding C, Sun Y. Morphology Remodeling and Selective Autophagy of Intracellular Organelles during Viral Infections. Int J Mol Sci 2020; 21:ijms21103689. [PMID: 32456258 PMCID: PMC7279407 DOI: 10.3390/ijms21103689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/14/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
Viruses have evolved different strategies to hijack subcellular organelles during their life cycle to produce robust infectious progeny. Successful viral reproduction requires the precise assembly of progeny virions from viral genomes, structural proteins, and membrane components. Such spatial and temporal separation of assembly reactions depends on accurate coordination among intracellular compartmentalization in multiple organelles. Here, we overview the rearrangement and morphology remodeling of virus-triggered intracellular organelles. Focus is given to the quality control of intracellular organelles, the hijacking of the modified organelle membranes by viruses, morphology remodeling for viral replication, and degradation of intracellular organelles by virus-triggered selective autophagy. Understanding the functional reprogram and morphological remodeling in the virus-organelle interplay can provide new insights into the development of broad-spectrum antiviral strategies.
Collapse
Affiliation(s)
- Shanhui Ren
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai 200241, China;
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai 200241, China;
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (C.D.); (Y.S.); Tel.: +86-21-34293441 (C.D. & Y.S.); Fax: +86-21-54081818 (C.D. & Y.S.)
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute. Chinese Academy of Agricultural Science, Shanghai 200241, China;
- Correspondence: (C.D.); (Y.S.); Tel.: +86-21-34293441 (C.D. & Y.S.); Fax: +86-21-54081818 (C.D. & Y.S.)
| |
Collapse
|
29
|
Infectious Bronchitis Virus Regulates Cellular Stress Granule Signaling. Viruses 2020; 12:v12050536. [PMID: 32422883 PMCID: PMC7291021 DOI: 10.3390/v12050536] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
Viruses must hijack cellular translation machinery to express viral genes. In many cases, this is impeded by cellular stress responses. These stress responses result in the global inhibition of translation and the storage of stalled mRNAs, into RNA-protein aggregates called stress granules. This results in the translational silencing of the majority of mRNAs excluding those beneficial for the cell to resolve the specific stress. For example, the expression of antiviral factors is maintained during viral infection. Here we investigated stress granule regulation by Gammacoronavirus infectious bronchitis virus (IBV), which causes the economically important poultry disease, infectious bronchitis. Interestingly, we found that IBV is able to inhibit multiple cellular stress granule signaling pathways, whilst at the same time, IBV replication also results in the induction of seemingly canonical stress granules in a proportion of infected cells. Moreover, IBV infection uncouples translational repression and stress granule formation and both processes are independent of eIF2α phosphorylation. These results provide novel insights into how IBV modulates cellular translation and antiviral stress signaling.
Collapse
|
30
|
The UPR sensor IRE1α and the adenovirus E3-19K glycoprotein sustain persistent and lytic infections. Nat Commun 2020; 11:1997. [PMID: 32332742 PMCID: PMC7181865 DOI: 10.1038/s41467-020-15844-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Persistent viruses cause chronic disease, and threaten the lives of immunosuppressed individuals. Here, we elucidate a mechanism supporting the persistence of human adenovirus (AdV), a virus that can kill immunosuppressed patients. Cell biological analyses, genetics and chemical interference demonstrate that one of five AdV membrane proteins, the E3-19K glycoprotein specifically triggers the unfolded protein response (UPR) sensor IRE1α in the endoplasmic reticulum (ER), but not other UPR sensors, such as protein kinase R-like ER kinase (PERK) and activating transcription factor 6 (ATF6). The E3-19K lumenal domain activates the IRE1α nuclease, which initiates mRNA splicing of X-box binding protein-1 (XBP1). XBP1s binds to the viral E1A-enhancer/promoter sequence, and boosts E1A transcription, E3-19K levels and lytic infection. Inhibition of IRE1α nuclease interrupts the five components feedforward loop, E1A, E3-19K, IRE1α, XBP1s, E1A enhancer/promoter. This loop sustains persistent infection in the presence of the immune activator interferon, and lytic infection in the absence of interferon. Adenovirus (AdV) can cause persistent infections, but underlying mechanisms are poorly understood. Here, Prasad et al. show that the AdV glycoprotein E3-19K activates the unfolded protein response sensor IRE1α, and that this triggers a feedforward loop that sustains persistent infection in the presence of interferon.
Collapse
|
31
|
Yin Q, Strong MJ, Zhuang Y, Flemington EK, Kaminski N, de Andrade JA, Lasky JA. Assessment of viral RNA in idiopathic pulmonary fibrosis using RNA-seq. BMC Pulm Med 2020; 20:81. [PMID: 32245461 PMCID: PMC7119082 DOI: 10.1186/s12890-020-1114-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 03/13/2020] [Indexed: 11/23/2022] Open
Abstract
Background Numerous publications suggest an association between herpes virus infection and idiopathic pulmonary fibrosis (IPF). These reports have employed immunohistochemistry, in situ hybridization and/or PCR, which are susceptible to specificity artifacts. Methods We investigated the possible association between IPF and viral RNA expression using next-generation sequencing, which has the potential to provide a high degree of both sensitivity and specificity. We quantified viral RNA expression for 740 viruses in 28 IPF patient lung biopsy samples and 20 controls. Key RNA-seq results were confirmed using Real-time RT-PCR for select viruses (EBV, HCV, herpesvirus saimiri and HERV-K). Results We identified sporadic low-level evidence of viral infections in our lung tissue specimens, but did not find a statistical difference for expression of any virus, including EBV, herpesvirus saimiri and HERV-K, between IPF and control lungs. Conclusions To the best of our knowledge, this is the first publication that employs RNA-seq to assess whether viral infections are linked to the pathogenesis of IPF. Our results do not address the role of viral infection in acute exacerbations of IPF, however, this analysis patently did not support an association between herpes virus detection and IPF.
Collapse
Affiliation(s)
- Qinyan Yin
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Michael J Strong
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Yan Zhuang
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Erik K Flemington
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University, 300 Cedar Street, Ste S441D, New Haven, CT, 06519, USA
| | - Joao A de Andrade
- Division of Allergy, Pulmonary, Critical Care Medicine, Department of Medicine, Vanderbilt University, 1161 21st Avenue South, B1317 MCN, Nashville, TN, 37232-2650, USA
| | - Joseph A Lasky
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA.
| |
Collapse
|
32
|
Johnston BP, McCormick C. Herpesviruses and the Unfolded Protein Response. Viruses 2019; 12:E17. [PMID: 31877732 PMCID: PMC7019427 DOI: 10.3390/v12010017] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Herpesviruses usurp cellular stress responses to promote viral replication and avoid immune surveillance. The unfolded protein response (UPR) is a conserved stress response that is activated when the protein load in the ER exceeds folding capacity and misfolded proteins accumulate. The UPR aims to restore protein homeostasis through translational and transcriptional reprogramming; if homeostasis cannot be restored, the UPR switches from "helper" to "executioner", triggering apoptosis. It is thought that the burst of herpesvirus glycoprotein synthesis during lytic replication causes ER stress, and that these viruses may have evolved mechanisms to manage UPR signaling to create an optimal niche for replication. The past decade has seen considerable progress in understanding how herpesviruses reprogram the UPR. Here we provide an overview of the molecular events of UPR activation, signaling and transcriptional outputs, and highlight key evidence that herpesviruses hijack the UPR to aid infection.
Collapse
Affiliation(s)
- Benjamin P. Johnston
- Department of Microbiology & Immunology, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada;
- Beatrice Hunter Cancer Research Institute, 5850 College Street, Halifax, NS B3H 4R2, Canada
| | - Craig McCormick
- Department of Microbiology & Immunology, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada;
- Beatrice Hunter Cancer Research Institute, 5850 College Street, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
33
|
Johnston BP, Pringle ES, McCormick C. KSHV activates unfolded protein response sensors but suppresses downstream transcriptional responses to support lytic replication. PLoS Pathog 2019; 15:e1008185. [PMID: 31790507 PMCID: PMC6907875 DOI: 10.1371/journal.ppat.1008185] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 12/12/2019] [Accepted: 11/02/2019] [Indexed: 12/18/2022] Open
Abstract
Herpesviruses usurp host cell protein synthesis machinery to convert viral mRNAs into proteins, and the endoplasmic reticulum (ER) to ensure proper folding, post-translational modification and trafficking of secreted and transmembrane viral proteins. Overloading ER folding capacity activates the unfolded protein response (UPR), whereby sensor proteins ATF6, PERK and IRE1 initiate a stress-mitigating transcription program that accelerates catabolism of misfolded proteins while increasing ER folding capacity. Kaposi’s sarcoma-associated herpesvirus (KSHV) can be reactivated from latency by chemical induction of ER stress, which causes accumulation of the XBP1s transcription factor that transactivates the viral RTA lytic switch gene. The presence of XBP1s-responsive elements in the RTA promoter suggests that KSHV evolved a mechanism to respond to ER stress. Here, we report that ATF6, PERK and IRE1 were activated upon reactivation from latency and required for efficient KSHV lytic replication; genetic or pharmacologic inhibition of each UPR sensor diminished virion production. Despite UPR sensor activation during KSHV lytic replication, downstream UPR transcriptional responses were restricted; 1) ATF6 was cleaved to activate the ATF6(N) transcription factor but ATF6(N)-responsive genes were not transcribed; 2) PERK phosphorylated eIF2α but ATF4 did not accumulate; 3) IRE1 caused XBP1 mRNA splicing, but XBP1s protein did not accumulate and XBP1s-responsive genes were not transcribed. Ectopic expression of the KSHV host shutoff protein SOX did not affect UPR gene expression, suggesting that alternative viral mechanisms likely mediate UPR suppression during lytic replication. Complementation of XBP1s deficiency during KSHV lytic replication inhibited virion production in a dose-dependent manner in iSLK.219 cells but not in TREx-BCBL1-RTA cells. However, genetically distinct KSHV virions harvested from these two cell lines were equally susceptible to XBP1s restriction following infection of naïve iSLK cells. This suggests that cell-intrinsic properties of BCBL1 cells may circumvent the antiviral effect of ectopic XBP1s expression. Taken together, these findings indicate that while XBP1s plays an important role in reactivation from latency, it can inhibit virus replication at a later step, which the virus overcomes by preventing its synthesis. These findings suggest that KSHV hijacks UPR sensors to promote efficient viral replication while sustaining ER stress. Like all viruses, Kaposi’s sarcoma-associated herpesvirus (KSHV) uses cellular machinery to create viral proteins. Some of these proteins are folded and modified in the endoplasmic reticulum (ER) and traverse the cellular secretory apparatus. Exceeding ER protein folding capacity activates the unfolded protein response (UPR), which resolves ER stress by putting the brakes on protein synthesis and turning on stress-mitigating genes. We show that KSHV replication activates the three cellular proteins that sense ER stress, which are each required to support efficient viral replication. By contrast, KSHV blocks the UPR gene expression program downstream from each of these activated sensor proteins. The failure to resolve ER stress might normally be expected to put the virus at a disadvantage, but we demonstrate that reversal of this scenario is worse; when we supplement infected epithelial cells with the UPR transcription factor XBP1s to artificially stimulate the production of UPR-responsive gene products, virus replication is blocked at a late stage and very few viruses are released from infected cells. Taken together, these observations suggest that KSHV requires UPR sensor protein activation to replicate but has dramatically altered the outcome to prevent the synthesis of new UPR proteins and sustain stress in the ER compartment.
Collapse
Affiliation(s)
- Benjamin P. Johnston
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Eric S. Pringle
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Craig McCormick
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
- * E-mail:
| |
Collapse
|
34
|
Yang S, Zhu J, Zhou X, Wang H, Li X, Zhao A. Induction of the unfolded protein response (UPR) during pseudorabies virus infection. Vet Microbiol 2019; 239:108485. [DOI: 10.1016/j.vetmic.2019.108485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/25/2019] [Accepted: 10/25/2019] [Indexed: 01/17/2023]
|
35
|
Verwilst P, Kim K, Sunwoo K, Kim HR, Kang C, Kim JS. Revealing Protein Aggregates under Thapsigargin-Induced ER Stress Using an ER-Targeted Thioflavin. ACS Sens 2019; 4:2858-2863. [PMID: 31617349 DOI: 10.1021/acssensors.9b00568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Endoplasmic reticulum-thioflavin T (ER-ThT), a thioflavin T-based fluorescent chemosensor, was developed to detect protein aggregates in the endoplasmic reticulum (ER) and was applied to live cells under various forms of ER stress. Upon dithiothreitol (DTT)-induced reductive denaturation of lysozyme and albumin, the intensity was increased in a protein concentration-dependent way, following a nonfluorescent lag phase. ER-ThT detects protein aggregates rather than unfolded proteins in solution, and the protein aggregation can be visualized in the presence of lipid membranes or native proteins. Within live HeLa cells, ER-ThT is localized in the ER and its fluorescence was dramatically increased upon ER stress induction by DTT, Thapsigargin, or Brefeldin A. Moreover, in the presence of ER stress modulators (tauroursodeoxycholic acid, trimethylamine N-oxide, or 4-phenylbutyric acid), also known as chemical chaperones, the fluorescence under Thapsigargin treatment was suppressed to the level of the control group. Thus, ER-ThT is capable of detecting the accumulation of protein aggregates under ER stress in living cells and acts as an in vitro screening tool for ER stress modulators, putative prodrugs against ER-related proteopathy. Overall, the results strongly suggest that protein aggregation is intricately involved in the activation of the unfolded protein response following ER stress.
Collapse
Affiliation(s)
- Peter Verwilst
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Kyutae Kim
- School of East−West Medical Science, Kyung Hee University, Yongin 17104, Korea
| | - Kyoung Sunwoo
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Hye-Ri Kim
- School of East−West Medical Science, Kyung Hee University, Yongin 17104, Korea
| | - Chulhun Kang
- School of East−West Medical Science, Kyung Hee University, Yongin 17104, Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea
| |
Collapse
|
36
|
Gao P, Chai Y, Song J, Liu T, Chen P, Zhou L, Ge X, Guo X, Han J, Yang H. Reprogramming the unfolded protein response for replication by porcine reproductive and respiratory syndrome virus. PLoS Pathog 2019; 15:e1008169. [PMID: 31738790 PMCID: PMC6932825 DOI: 10.1371/journal.ppat.1008169] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 12/26/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
The unfolded protein response (UPR) in the endoplasmic reticulum (ER) constitutes a critical component of host innate immunity against microbial infections. In this report, we show that porcine reproductive and respiratory syndrome virus (PRRSV) utilizes the UPR machinery for its own benefit. We provide evidence that the virus targets the UPR central regulator GRP78 for proteasomal degradation via a mechanism that requires viral glycoprotein GP2a, while both IRE1-XBP1s and PERK-eIF2α-ATF4 signaling branches of the UPR are turned on at early stage of infection. The activated effector XBP1s was found to enter the nucleus, but ATF4 was unexpectedly diverted to cytoplasmic viral replication complexes by means of nonstructural proteins nsp2/3 to promote viral RNA synthesis. RNAi knockdown of either ATF4 or XBP1s dramatically attenuated virus titers, while overexpression caused increases. These observations reveal attractive host targets (e.g., ATF4 and XBP1s) for antiviral drugs and have implications in vaccine development. Porcine reproductive and respiratory syndrome virus (PRRSV) poses a major threat to the worldwide swine industry, but no effective vaccines or antiviral drugs are available. A better understanding of the pathogen-host interactions that support PRRSV replication is essential for understanding viral pathogenesis and the development of preventive measures. Here we report that PRRSV utilizes unconventional strategies to reprogram the unfolded protein response (UPR) of the host to its own advantage. The virus targets GRP78 for partial degradation to create a favorable environment for UPR induction and hijacks ATF4 into cytoplasmic replication complexes to promote viral RNA synthesis. The data also reveal potential targets (e.g., ATF4 and XBP1s) for antiviral drugs and have implications in vaccine development.
Collapse
Affiliation(s)
- Peng Gao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Yue Chai
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Jiangwei Song
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Teng Liu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Peng Chen
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Lei Zhou
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Xinna Ge
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Xin Guo
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| | - Jun Han
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
- * E-mail:
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, China Agricultural University College of Veterinary Medicine, Beijing, People’s Republic of China
| |
Collapse
|
37
|
Porcine parvovirus replication is suppressed by activation of the PERK signaling pathway and endoplasmic reticulum stress-mediated apoptosis. Virology 2019; 539:1-10. [PMID: 31605941 PMCID: PMC7127029 DOI: 10.1016/j.virol.2019.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/27/2019] [Accepted: 09/24/2019] [Indexed: 12/16/2022]
Abstract
Endoplasmic reticulum (ER) stress is associated with numerous mammalian diseases, especially viral diseases. Porcine parvovirus (PPV) is the causative agent of reproductive failure in swine. Here, we observed that the PPV infection of porcine kidney 15 and porcine testis cells resulted in the activation of ER stress sensors mediated by protein kinase R-like ER kinase (PERK), but not inositol-requiring enzyme 1 and activating transcription factor 6 (ATF6). ER stress activation obviously blocked PPV replication. Depletion of proteins, such as PERK, eukaryotic initiation factor 2, and ATF4, by small interfering RNA significantly enhanced PPV replication. Moreover, the pro-apoptotic factor C/EBP homologous protein was identified a key factor in the inhibition of PPV replication. These data demonstrate that PPV infection activates ER stress through the PERK signaling pathway and that ER stress inhibits further PPV replication by promoting apoptosis.
Collapse
|
38
|
Mammalian orthoreovirus Infection is Enhanced in Cells Pre-Treated with Sodium Arsenite. Viruses 2019; 11:v11060563. [PMID: 31216693 PMCID: PMC6631071 DOI: 10.3390/v11060563] [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: 05/21/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/19/2022] Open
Abstract
Following reovirus infection, cells activate stress responses that repress canonical translation as a mechanism to limit progeny virion production. Work by others suggests that these stress responses, which are part of the integrated stress response (ISR), may benefit rather than repress reovirus replication. Here, we report that compared to untreated cells, treating cells with sodium arsenite (SA) to activate the ISR prior to infection enhanced viral protein expression, percent infectivity, and viral titer. SA-mediated enhancement was not strain-specific, but was cell-type specific. While SA pre-treatment of cells offered the greatest enhancement, treatment within the first 4 h of infection increased the percent of cells infected. SA activates the heme-regulated eIF2α (HRI) kinase, which phosphorylates eukaryotic translation initiation factor 2 alpha (eIF2α) to induce stress granule (SG) formation. Heat shock (HS), another activator of HRI, also induced eIF2α phosphorylation and SGs in cells. However, HS had no effect on percent infectivity or viral yield but did enhance viral protein expression. These data suggest that SA pre-treatment perturbs the cell in a way that is beneficial for reovirus and that this enhancement is independent of SG induction. Understanding how to manipulate the cellular stress responses during infection to enhance replication could help to maximize the oncolytic potential of reovirus.
Collapse
|
39
|
Grimaldi G, Catara G, Palazzo L, Corteggio A, Valente C, Corda D. PARPs and PAR as novel pharmacological targets for the treatment of stress granule-associated disorders. Biochem Pharmacol 2019; 167:64-75. [PMID: 31102582 DOI: 10.1016/j.bcp.2019.05.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022]
Abstract
Among the post-translational modifications, ADP-ribosylation has been for long time the least integrated in the scheme of the structural protein modifications affecting physiological functions. In spite of the original findings on bacterial-dependent ADP-ribosylation catalysed by toxins such as cholera and pertussis toxin, only with the discovery of the poly-ADP-ribosyl polymerase (PARP) family the field has finally expanded and the role of ADP-ribosylation has been recognised in both physiological and pathological processes, including cancer, infectious and neurodegenerative diseases. This is now a rapidly expanding field of investigation, centred on the role of the different PARPs and their substrates in various diseases, and on the potential of PARP inhibitors as novel pharmacological tools to be employed in relevant pathological context. In this review we analyse the role that members of the PARP family and poly-ADP-ribose (PAR; the product of PARP1 and PARP5a activity) play in the processes following the exposure of cells to different stresses. The cell response that arises following conditions such as heat, osmotic, oxidative stresses or viral infection relies on the formation of stress granules, which are transient cytoplasmic membrane-less structures, that include untranslated mRNA, specific proteins and PAR, this last one serving as the "collector" of all components (that bind to it in a non-covalent manner). The resulting phenotypes are cells in which translation, intracellular transport or pro-apoptotic pathways are reversibly inhibited, for the time the given stress holds. Interestingly, the formation of defective stress granules has been detected in diverse pathological conditions including neurological disorders and cancer. Analysing the molecular details of stress granule formation under these conditions offers a novel view on the pathogenesis of these diseases and, as a consequence, the possibility of identifying novel drug targets for their treatment.
Collapse
Affiliation(s)
- Giovanna Grimaldi
- Institute of Protein Biochemistry, National Research Council of Italy, Via Pietro Castellino 111, Naples 80131, Italy.
| | - Giuliana Catara
- Institute of Protein Biochemistry, National Research Council of Italy, Via Pietro Castellino 111, Naples 80131, Italy
| | - Luca Palazzo
- Institute of Protein Biochemistry, National Research Council of Italy, Via Pietro Castellino 111, Naples 80131, Italy
| | - Annunziata Corteggio
- Institute of Protein Biochemistry, National Research Council of Italy, Via Pietro Castellino 111, Naples 80131, Italy
| | - Carmen Valente
- Institute of Protein Biochemistry, National Research Council of Italy, Via Pietro Castellino 111, Naples 80131, Italy
| | - Daniela Corda
- Institute of Protein Biochemistry, National Research Council of Italy, Via Pietro Castellino 111, Naples 80131, Italy.
| |
Collapse
|
40
|
Porcine circovirus type 2 ORF5 protein induces endoplasmic reticulum stress and unfolded protein response in porcine alveolar macrophages. Arch Virol 2019; 164:1323-1334. [PMID: 30877450 DOI: 10.1007/s00705-019-04185-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/19/2019] [Indexed: 01/12/2023]
Abstract
Porcine circovirus type 2 (PCV2) is the essential infectious agent causing porcine circovirus-associated disease (PCVD) in pigs and one of the important viruses that severely jeopardize the swine husbandry industry. PCV2 elicits the unfolded protein response (UPR) via activation of the PERK pathway, and its capsid protein (Cap) has also been found to induce UPR with subsequent activation of apoptosis. The open reading frame 5 (ORF5) protein is a recently discovered non-structural protein, and its function in PCV2 pathogenesis remains unknown. The aim of this study was to determine whether the PCV2 ORF5 protein could induce endoplasmic reticulum stress (ERS) and UPR in porcine alveolar macrophages (PAMs). pEGFP-tagged ORF5 protein was transiently overexpressed in PAMs. Transmission electron microscopy (TEM) was employed to examine changes in ER morphology, and quantitative real-time PCR and western blotting analysis were used to measure UPR-related cell signaling alterations. We found that the ORF5 protein triggers swelling and degranulation of the ER and upregulates the expression of ERS markers. Further experiments demonstrated that the PCV2 ORF5 protein induces ERS and UPR via the PERK (RNA-activated protein kinase-like endoplasmic reticulum kinase), ATF6 (activating transcription factor 6) and IRE1 (inositol requiring enzyme 1) signaling pathways. Together with previous studies, we provide new information on the ERS-UPR induced by the PCV2 ORF5 protein.
Collapse
|
41
|
Zhao Z, Zhu X, Wu N, Qin X, Huang C, Wu G, Zhang Q, Zhang Z. Species-specific inhibition of capripoxvirus replication by host antiviral protein kinase R. Ann N Y Acad Sci 2018; 1438:3-17. [PMID: 30381842 PMCID: PMC7379242 DOI: 10.1111/nyas.13976] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/17/2018] [Accepted: 09/12/2018] [Indexed: 12/29/2022]
Abstract
The role of interferon (IFN)‐induced protein kinase R (PKR) in capripoxvirus (CaPV)‐infected cells remains unknown. In this study, we show that CaPV infection triggered PKR and eukaryotic translation initiation factor 2 alpha (eIF2α) protein phosphorylation in a dose‐dependent manner, and that this leads to decreased CaPV replication. Overexpression of PKR compromised viral gene expression and inhibited sheeppox virus (SPPV) replication. Downregulation of PKR with siRNAs significantly decreased eIF2α phosphorylation and reduced the mRNA level of IFN‐β, which increased virus replication. In luciferase assays, species‐different CaPVs K3L proteins inhibited sheep PKR (sPKR): goatpox virus K3L strongly inhibited sPKR and goat PKR (gPKR), but SPPV K3L only partially inhibited gPKR. These results are the first to show that SPPV infection induces phosphorylation of eIF2α through PKR activation, which then results in restriction of CaPV replication. Furthermore, our data show that CaPV K3L inhibits PKR in a species‐specific manner. The results presented are consistent with the hypothesis that different levels of PKR inhibition by K3L orthologs from various viruses could potentially contribute to the host range function of K3L.
Collapse
Affiliation(s)
- Zhixun Zhao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xueliang Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Na Wu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiaodong Qin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Caiyun Huang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Guohua Wu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Qiang Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhidong Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
42
|
Neerukonda SN, Katneni UK, Bott M, Golovan SP, Parcells MS. Induction of the unfolded protein response (UPR) during Marek's disease virus (MDV) infection. Virology 2018; 522:1-12. [PMID: 29979959 DOI: 10.1016/j.virol.2018.06.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/14/2018] [Accepted: 06/27/2018] [Indexed: 12/22/2022]
Abstract
Marek's disease (MD) is a pathology of chickens associated with paralysis, immune suppression, and the rapid formation of T-cell lymphomas. MD is caused by the herpesvirus, Marek's disease virus (MDV). We examined endoplasmic reticulum (ER) stress and the activation of unfolded protein response (UPR) pathways during MDV infection of cells in culture and lymphocytes in vivo. MDV strains activate the UPR as measured by increased mRNA expression of GRP78/BiP with concomitant XBP1 splicing and induction of its target gene, EDEM1. Cell culture replication of virulent, but not vaccine MDVs, activated the UPR at late in infection. Pathotype-associated UPR activation was induced to a greater level by a vv + MDV. Discrete UPR activation was observed during MDV in vivo infection, with the level of UPR modulation being affected by the MDV oncoprotein Meq. Finally, ATF6 was found to be activated in vv + MDV-induced primary lymphomas, suggesting a possible role in tumor progression.
Collapse
Affiliation(s)
- Sabari Nath Neerukonda
- Department of Animal and Food Sciences, University of Delaware, 052 Townsend Hall, 531 South College Ave, Newark, DE 19716, United States.
| | - Upendra K Katneni
- Department of Animal and Food Sciences, University of Delaware, 052 Townsend Hall, 531 South College Ave, Newark, DE 19716, United States.
| | - Matthew Bott
- Department of Animal and Food Sciences, University of Delaware, 052 Townsend Hall, 531 South College Ave, Newark, DE 19716, United States.
| | | | - Mark S Parcells
- Department of Animal and Food Sciences, University of Delaware, 052 Townsend Hall, 531 South College Ave, Newark, DE 19716, United States.
| |
Collapse
|
43
|
Sato E, Hiromatsu K, Murata K, Imafuku S. Loss of ATP2A2 Allows Herpes Simplex Virus 1 Infection of a Human Epidermis Model by Disrupting Innate Immunity and Barrier Function. J Invest Dermatol 2018; 138:2540-2549. [PMID: 29870688 DOI: 10.1016/j.jid.2018.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 04/16/2018] [Accepted: 05/17/2018] [Indexed: 12/24/2022]
Abstract
Destruction of epidermal barrier function associated with atopic dermatitis or Darier's disease often causes severe secondary skin infections. Patients with skin barrier disorders often repeatedly acquire Kaposi varicelliform eruption, which is caused by herpes simplex virus, but the underlying mechanisms and effective preventive methods have yet to be found. Viral infection through an impaired epidermal barrier can be prevented by enhancing innate immunity and/or inhibiting viral entry. In this study, we established a three-dimensional skin barrier dysfunction model by silencing ATP2A2, which is mutated in some Darier's disease patients. We confirmed the loss of desmosomes and presence of histopathological clefts in the suprabasal layer. Herpes simplex virus 1 applied to the stratum corneum infected the deep epidermis. An innate immune reaction was assessed by evaluating the expression of IFNB1 and related genes. Pretreatment with polyinosinic-polycytidylic acid alone or plus the antimicrobial peptide, LL37 enhanced IFN-β production and suppressed viral replication. Furthermore, topical application of a white petrolatum ointment containing heparin, which binds viral glycoproteins related to virus entry, strongly inhibited viral replication, probably by inhibiting invasion. Our human barrier-dysfunctional model will have future application for identifying the mechanism of Kaposi varicelliform eruption onset, preventive methods, and therapies.
Collapse
Affiliation(s)
- Emi Sato
- Department of Microbiology and Immunology, Fukuoka University Faculty of Medicine, Fukuoka, Japan; Department of Dermatology, Fukuoka University Hospital, Fukuoka, Japan.
| | - Kenji Hiromatsu
- Department of Microbiology and Immunology, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Kunihiko Murata
- The Center for Electron Microscopy, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Shinichi Imafuku
- Department of Dermatology, Fukuoka University Faculty of Medicine, Fukuoka, Japan; Department of Dermatology, Fukuoka University Hospital, Fukuoka, Japan
| |
Collapse
|
44
|
Bommareddy PK, Peters C, Saha D, Rabkin SD, Kaufman HL. Oncolytic Herpes Simplex Viruses as a Paradigm for the Treatment of Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050254] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Praveen K. Bommareddy
- Department of Surgery, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Cole Peters
- Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
- Program in Virology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Dipongkor Saha
- Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Samuel D. Rabkin
- Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Howard L. Kaufman
- Department of Surgery, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
| |
Collapse
|
45
|
He W, Xu H, Gou H, Yuan J, Liao J, Chen Y, Fan S, Xie B, Deng S, Zhang Y, Chen J, Zhao M. CSFV Infection Up-Regulates the Unfolded Protein Response to Promote Its Replication. Front Microbiol 2017; 8:2129. [PMID: 29163417 PMCID: PMC5673830 DOI: 10.3389/fmicb.2017.02129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/18/2017] [Indexed: 01/07/2023] Open
Abstract
Classical swine fever (CSF) is an OIE-listed, highly contagious animal disease caused by classical swine fever virus (CSFV). The endoplasmic reticulum (ER) is an organelle in which the replication of many RNA viruses takes place. During viral infection, a series of events elicited in cells can destroy the ER homeostasis that cause ER stress and induce an unfolded protein response (UPR). In this study, we demonstrate that ER stress was induced during CSFV infection as several UPR-responsive elements such as XBP1(s), GRP78 and CHOP were up-regulated. Specifically, CSFV transiently activated IRE1 pathway at the initial stage of infection but rapidly switched off, likely due to the reduction in cytoplasm Ca2+ after viral incubation. Additionally, our data show that the ER stress induced by CSFV can promote CSFV production, which the IRE1 pathway play an important role in it. Evidence of ER stress in vivo was also confirmed by the marked elevation of GRP78 in CSFV-infected pig PBMC and tissues. Collectively, these data indicate that the ER stress was induced upon CSFV infection and that the activation of the IRE1 pathway benefits CSFV replication.
Collapse
Affiliation(s)
- Wencheng He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hailuan Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongchao Gou
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jin Yuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiedan Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuming Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Baoming Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shaofeng Deng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yangyi Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
46
|
Li J, He J, Fu Y, Hu X, Sun LQ, Huang Y, Fan X. Hepatitis B virus X protein inhibits apoptosis by modulating endoplasmic reticulum stress response. Oncotarget 2017; 8:96027-96034. [PMID: 29221184 PMCID: PMC5707078 DOI: 10.18632/oncotarget.21630] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/26/2017] [Indexed: 12/11/2022] Open
Abstract
Chronic Hepatitis B virus (HBV) infection is a major risk of hepatocellular carcinoma (HCC) worldwide. Hepatitis B virus X protein (HBx) is encoded by one of the four open reading frames of HBV, and is well known as an important coactivator for HBV replication and HBV-associated hepatocellular carcinogenesis. However, its role in keeping cells from apoptosis to promote HCC proliferation remains controversial. Here, we used HBx expressing HCC cells as a model, to investigate the mechanism of HBx-mediated cellular response to endoplasmic reticulum (ER) stress. We found that HBx protein was localized in ER lumen and interacted with GRP78 directly. This interaction resulted in suppression of eIF2α phosphorylation, inhibited expression of ATF4/CHOP/Bcl-2, and reduced cleavage of poly ADP-ribose polymerase (PARP) and level of γH2AX, thus preventing HCC cells from cell death and negatively regulating DNA repair. This study reveals a novel mechanism of the HBx-mediated oncogenesis and provides a basis for potential HBx-targeted therapeutic intervention of HCC.
Collapse
Affiliation(s)
- Jia Li
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Emergency, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiang He
- Center for Molecular Medicine, Center for Molecular Medicine, Xiangya Hospital, Collaborative Innovation Center for Cancer Medicine, Central South University, Changsha 410078, China.,Key Laboratory of Molecular Radiation Oncology, Changsha 410008, China
| | - Yongming Fu
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xingwang Hu
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lun-Quan Sun
- Center for Molecular Medicine, Center for Molecular Medicine, Xiangya Hospital, Collaborative Innovation Center for Cancer Medicine, Central South University, Changsha 410078, China.,Key Laboratory of Molecular Radiation Oncology, Changsha 410008, China
| | - Yan Huang
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xuegong Fan
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
47
|
Escherichia coli O157:H7 suppresses host autophagy and promotes epithelial adhesion via Tir-mediated and cAMP-independent activation of protein kinase A. Cell Death Discov 2017; 3:17055. [PMID: 28975041 PMCID: PMC5624281 DOI: 10.1038/cddiscovery.2017.55] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 07/07/2017] [Indexed: 11/18/2022] Open
Abstract
Autophagy is a pivotal innate immune response that not only degrades cytosolic components, but also serves as one of the critical antimicrobial mechanisms eliminating intracellular pathogens. However, its role in host defense against extracellular pathogens is largely unknown. Here we showed that E. coli O157:H7 altered autophagy to evade host defense and facilitate adhesion. Enhancing host cell autophagy with tumor necrosis factor (TNF), host starvation or rapamycin reduced the adherence of E. coli O157:H7 to HT-29 cells. As a key regulator of autophagy, protein kinase A (PKA) was activated by E. coli O157:H7 infection. PKA inhibition by H89 abrogated E. coli O157:H7 inhibition of autophagy and prevented bacterial epithelial adhesion. Thus, PKA had a mediatory role in blocking autophagy and E. coli O157:H7 epithelial adhesion. Furthermore, deletion of translocated intimin receptor (tir) prevented PKA activation, whereas ectopic tir expression in a Δtir mutant strain restored its ability to activate PKA and inhibited autophagy in host cells. This indicated that Tir and PKA played pivotal roles in manipulating host autophagy during infection. Consistent with autophagy inhibition, E. coli O157:H7 infection inhibited endoplasmic reticulum (ER) stress in HT-29 cells, which was reversed by TNF, starvation, or H89 treatment. Additionally, E. coli O157:H7-induced PKA activation suppressed extracellular signal-regulated kinase 1/2 (ERK1/2) activation and enhanced phosphatidylinositol 3-kinase/Akt (PI3K/Akt) signaling, thereby repressing autophagic signaling. Conversely, PKA inhibition prevented downregulation of ERK1/2 signaling due to E. coli O157:H7 infection. In summary, E. coli O157:H7 inhibited host autophagy via Tir-mediated PKA activation that favored bacterial persistence on intestinal epithelial cell surfaces.
Collapse
|
48
|
Opposite Roles of RNase and Kinase Activities of Inositol-Requiring Enzyme 1 (IRE1) on HSV-1 Replication. Viruses 2017; 9:v9090235. [PMID: 28832521 PMCID: PMC5618002 DOI: 10.3390/v9090235] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 08/12/2017] [Accepted: 08/16/2017] [Indexed: 12/30/2022] Open
Abstract
In response to the endoplasmic reticulum (ER) stress induced by herpes simplex virus type 1 (HSV-1) infection, host cells activate the unfolded protein response (UPR) to reduce the protein-folding burden in the ER. The regulation of UPR upon HSV-1 infection is complex, and the downstream effectors can be detrimental to viral replication. Therefore, HSV-1 copes with the UPR to create a beneficial environment for its replication. UPR has three branches, including protein kinase RNA (PKR)-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1), and activated transcription factor 6 (ATF6). IRE1α is the most conserved branch of UPR which has both RNase and kinase activities. Previous studies have shown that IRE1α RNase activity was inactivated during HSV-1 infection. However, the effect of the two activities of IRE1α on HSV-1 replication remains unknown. Results in this study showed that IRE1α expression was up-regulated during HSV-1 infection. We found that in HEC-1-A cells, increasing RNase activity, or inhibiting kinase activity of IRE1α led to viral suppression, indicating that the kinase activity of IRE1α was beneficial, while the RNase activity was detrimental to viral replication. Further evidence showed that the kinase activity of IRE1α leads to the activation of the JNK (c-Jun N-terminal kinases) pathway, which enhances viral replication. Taken together, our evidence suggests that IRE1α is involved in HSV-1 replication, and its RNase and kinase activities play differential roles during viral infection.
Collapse
|
49
|
Abstract
Efficient viral gene expression is threatened by cellular stress response programmes that rapidly reprioritize the translation machinery in response to varied environmental assaults, including virus infection. This results in inhibition of bulk synthesis of housekeeping proteins and causes the aggregation of messenger ribonucleoprotein complexes into cytoplasmic foci that are known as stress granules, which can entrap viral mRNAs. There is accumulating evidence for the antiviral nature of stress granules, which is supported by the discovery of many viral factors that interfere with stress granule formation and/or function. This Review focuses on recent advances in our understanding of the role of translation inhibition and stress granules in antiviral immune responses.
Collapse
|
50
|
Herpes Simplex Virus 1 UL41 Protein Suppresses the IRE1/XBP1 Signal Pathway of the Unfolded Protein Response via Its RNase Activity. J Virol 2017; 91:JVI.02056-16. [PMID: 27928013 DOI: 10.1128/jvi.02056-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/30/2016] [Indexed: 12/18/2022] Open
Abstract
During viral infection, accumulation of viral proteins can cause stress in the endoplasmic reticulum (ER) and trigger the unfolded protein response (UPR) to restore ER homeostasis. The inositol-requiring enzyme 1 (IRE1)-dependent pathway is the most conserved of the three UPR signal pathways. Upon activation, IRE1 splices out an intron from the unspliced inactive form of X box binding protein 1 [XBP1(u)] mRNA and produces a transcriptionally potent spliced form [XBP1(s)]. Previous studies have reported that the IRE1/XBP1 pathway is inhibited upon herpes simplex virus 1 (HSV-1) infection; however, the underlying molecular mechanism is still elusive. Here, we uncovered a role of the HSV-1 UL41 protein in inhibiting the IRE1/XBP1 signal pathway. Ectopic expression of UL41 decreased the expression of XBP1 and blocked XBP1 splicing activation induced by the ER stress inducer thapsigargin. Wild-type (WT) HSV-1, but not the UL41-null mutant HSV-1 (R2621), decreased XBP1 mRNA induced by thapsigargin. Nevertheless, infection with both WT HSV-1 and R2621 without drug pretreatment could reduce the mRNA and protein levels of XBP1(s), and additional mechanisms might contribute to this inhibition of XBP1(s) during R2621 infection. Taking these findings together, our results reveal XBP1 as a novel target of UL41 and provide insights into the mechanism by which HSV-1 modulates the IRE1/XBP1 pathway. IMPORTANCE During viral infection, viruses hijack the host translation apparatus to produce large amounts of viral proteins, which leads to ER stress. To restore ER homeostasis, cells initiate the UPR to alleviate the effects of ER stress. The IRE1/XBP1 pathway is the most conserved UPR branch, and it activates ER-associated protein degradation (ERAD) to reduce the ER load. The IRE1/XBP1 branch is repressed during HSV-1 infection, but little is known about the underlying molecular mechanism. Our results show for the first time that UL41 suppresses the IRE1/XBP1 signal pathway by reducing the accumulation of XBP1 mRNA, and characterization of the underlying molecular mechanism provides new insight into the modulation of UPR by HSV-1.
Collapse
|