1
|
Sharma D, Rai M, Singh A, Gaur R, Senguptaa D. Fullerene (C 60 & C 70)-Meso-Tris-4-Carboxyphenyl Porphyrin Dyads Inhibit Entry of Wild-Type and Drug-Resistant HIV-1 Clades B and C. J Med Virol 2025; 97:e70181. [PMID: 39868857 DOI: 10.1002/jmv.70181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025]
Abstract
The biological applications of noncationic porphyrin-fullerene (P-F) dyads as anti-HIV agents have been limited despite the established use of several cationic P-F dyads as anti-cancer photodynamic therapy (PDT) agents. This article explores the potential of amphiphilic non-cationic porphyrin-fullerene dyads as HIV-1 inhibitors under both PDT (light-treated) and non-PDT (dark) conditions. The amphiphilic P-F dyads, PB3C60 and PB3C70, demonstrated enhanced efficacy in inhibiting the entry and production of HIV-1 (subtypes B and C). Under light-harvested conditions, the dyads exhibited potent inhibitory effects (EC50 for PB3C60 and PB3C70 < 10 nM) and also maintained significant inhibition under non-PDT conditions (EC50 for PB3C60 = 1.43 μM and PB3C70 = 1.50 μM), while displaying notably reduced toxicity compared to their water-soluble porphyrin precursor. The P-F dyads exhibited substantial efficacy in neutralizing the T20-resistant strain 9491, both at the entry and postentry phases, thereby addressing the challenge of drug resistance.
Collapse
Affiliation(s)
| | - Madhu Rai
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Aradhana Singh
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Ritu Gaur
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | | |
Collapse
|
2
|
Sharma D, Singh A, Safi S, Gaur R, Sengupta D. Porphyrins with combinations of 4-carboxyphenyl and 4-hydroxyphenyl substituents in meso-positions as anti-HIV-1 agents. Sci Rep 2024; 14:10006. [PMID: 38693160 PMCID: PMC11063187 DOI: 10.1038/s41598-024-60728-w] [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: 02/06/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024] Open
Abstract
A series of 4-carboxyphenyl/4-hydroxyphenyl meso-substituted porphyrins were synthesized, purified, and characterized. The compounds exhibited anti-HIV-1 activities, in vitro, under both non-photodynamic (non-PDT) and photodynamic (PDT) conditions. Specifically, the porphyrins inhibited HIV-1 virus entry, with c-PB2(OH)2 and PB(OH)3 showing significant anti-HIV-1 activity. All of the porphyrins inhibited HIV-1 subtype B and C virus entry under PDT conditions. Our study demonstrated that the compounds bearing combinations of 4-carboxyphenyl/4-hydroxyphenyl moieties were not toxic even at higher concentrations, as compared to the reference porphyrins 5,10,15,20-tetra-(4-carboxyphenyl)porphyrin (TCPP) and 5,10,15,20-tetra-(4-hydroxyphenyl)porphyrin (THPP), under PDT conditions. This study underscores the promising potential of these compounds as HIV entry inhibitors in both non-PDT and PDT scenarios.
Collapse
Affiliation(s)
- Debdulal Sharma
- Department of Chemistry, Assam University, Silchar, Assam, 788011, India
| | - Aradhana Singh
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110068, India
| | - Sanaullah Safi
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110068, India
| | - Ritu Gaur
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, 110068, India.
| | - Devashish Sengupta
- Department of Chemistry, Assam University, Silchar, Assam, 788011, India.
| |
Collapse
|
3
|
Li F, Yu H, Qi A, Zhang T, Huo Y, Tu Q, Qi C, Wu H, Wang X, Zhou J, Hu L, Ouyang H, Pang D, Xie Z. Regulatory Non-Coding RNAs during Porcine Viral Infections: Potential Targets for Antiviral Therapy. Viruses 2024; 16:118. [PMID: 38257818 PMCID: PMC10818342 DOI: 10.3390/v16010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Pigs play important roles in agriculture and bio-medicine; however, porcine viral infections have caused huge losses to the pig industry and severely affected the animal welfare and social public safety. During viral infections, many non-coding RNAs are induced or repressed by viruses and regulate viral infection. Many viruses have, therefore, developed a number of mechanisms that use ncRNAs to evade the host immune system. Understanding how ncRNAs regulate host immunity during porcine viral infections is critical for the development of antiviral therapies. In this review, we provide a summary of the classification, production and function of ncRNAs involved in regulating porcine viral infections. Additionally, we outline pathways and modes of action by which ncRNAs regulate viral infections and highlight the therapeutic potential of artificial microRNA. Our hope is that this information will aid in the development of antiviral therapies based on ncRNAs for the pig industry.
Collapse
Affiliation(s)
- Feng Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Hao Yu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Aosi Qi
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Tianyi Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Yuran Huo
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Qiuse Tu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Chunyun Qi
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Heyong Wu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Xi Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Jian Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Lanxin Hu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
| | - Hongsheng Ouyang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
- Chongqing Research Institute, Jilin University, Chongqing 401120, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401120, China
| | - Daxin Pang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
- Chongqing Research Institute, Jilin University, Chongqing 401120, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401120, China
| | - Zicong Xie
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.L.); (H.Y.); (A.Q.); (T.Z.); (Y.H.); (Q.T.); (C.Q.); (H.W.); (X.W.); (J.Z.); (L.H.); (H.O.)
- Chongqing Research Institute, Jilin University, Chongqing 401120, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401120, China
| |
Collapse
|
4
|
Li L, Hu X, Li S, Li Y, Zhao S, Shen F, Wang C, Li Y, Wang T. Cobalt Protoporphyrin Blocks EqHV-8 Infection via IFN-α/β Production. Animals (Basel) 2023; 13:2690. [PMID: 37684954 PMCID: PMC10487175 DOI: 10.3390/ani13172690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Equid alphaherpesvirus type 8 (EqHV-8) is the causative agent of severe respiratory disease, abortions, and neurological syndromes in equines and has resulted in huge economic losses to the donkey industry. Currently, there exist no therapeutic molecules for controlling EqHV-8 infection. We evaluated the potential antiviral activity of cobalt protoporphyrin (CoPP) against EqHV-8 infection. Our results demonstrated that CoPP inhibited EqHV-8 infection in susceptible cells and mouse models. Furthermore, CoPP blocked the replication of EqHV-8 via HO-1 (heme oxygenase-1) mediated type I interferon (IFN) response. In conclusion, our data suggested that CoPP could serve as a novel potential molecule to develop an effective therapeutic strategy for EqHV-8 prevention and control.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yubao Li
- College of Agronomy, Liaocheng University, Liaocheng 252000, China
| | - Tongtong Wang
- College of Agronomy, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|
5
|
Nielsen A, Handel M, Stone J, Lee M. Misreport of burns as a result of 'coining', Gua sha; inherent harms from publication and ongoing citation of false facts. Integr Med Res 2023; 12:100953. [PMID: 37201160 PMCID: PMC10186471 DOI: 10.1016/j.imr.2023.100953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/02/2023] [Indexed: 05/20/2023] Open
Affiliation(s)
- Arya Nielsen
- Icahn School of Medicine at Mount Sinai, Department of Family Medicine & Community Health, New York, USA
- Corresponding author at: Icahn School of Medicine at Mount Sinai, Department of Family Medicine & Community Health, New York, USA
| | - Marsha Handel
- Icahn School of Medicine at Mount Sinai, Department of Family Medicine & Community Health, New York, USA
| | | | - Myeong Soo Lee
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
- Korean Convergence Medical Science, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
6
|
Holoubek J, Bednářová K, Haviernik J, Huvarová I, Dvořáková Z, Černý J, Outlá M, Salát J, Konkol'ová E, Boura E, Růžek D, Vorlíčková M, Eyer L, Renčiuk D. Guanine quadruplexes in the RNA genome of the tick-borne encephalitis virus: their role as a new antiviral target and in virus biology. Nucleic Acids Res 2022; 50:4574-4600. [PMID: 35420134 PMCID: PMC9071444 DOI: 10.1093/nar/gkac225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022] Open
Abstract
We have identified seven putative guanine quadruplexes (G4) in the RNA genome of tick-borne encephalitis virus (TBEV), a flavivirus causing thousands of human infections and numerous deaths every year. The formation of G4s was confirmed by biophysical methods on synthetic oligonucleotides derived from the predicted TBEV sequences. TBEV-5, located at the NS4b/NS5 boundary and conserved among all known flaviviruses, was tested along with its mutated variants for interactions with a panel of known G4 ligands, for the ability to affect RNA synthesis by the flaviviral RNA-dependent RNA polymerase (RdRp) and for effects on TBEV replication fitness in cells. G4-stabilizing TBEV-5 mutations strongly inhibited RdRp RNA synthesis and exhibited substantially reduced replication fitness, different plaque morphology and increased sensitivity to G4-binding ligands in cell-based systems. In contrast, strongly destabilizing TBEV-5 G4 mutations caused rapid reversion to the wild-type genotype. Our results suggest that there is a threshold of stability for G4 sequences in the TBEV genome, with any deviation resulting in either dramatic changes in viral phenotype or a rapid return to this optimal level of G4 stability. The data indicate that G4s are critical elements for efficient TBEV replication and are suitable targets to tackle TBEV infection.
Collapse
Affiliation(s)
- Jiří Holoubek
- Veterinary Research Institute, Emerging Viral Diseases, Brno CZ-62100, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-37005 Ceske Budejovice, Czech Republic
| | - Klára Bednářová
- Department of Biophysics of Nucleic Acids, Institute of Biophysics of the Czech Academy of Sciences, Brno CZ-61200, Czech Republic
| | - Jan Haviernik
- Veterinary Research Institute, Emerging Viral Diseases, Brno CZ-62100, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-37005 Ceske Budejovice, Czech Republic
| | - Ivana Huvarová
- Veterinary Research Institute, Emerging Viral Diseases, Brno CZ-62100, Czech Republic
| | - Zuzana Dvořáková
- Department of Biophysics of Nucleic Acids, Institute of Biophysics of the Czech Academy of Sciences, Brno CZ-61200, Czech Republic
| | - Jiří Černý
- Faculty of Tropical Agrisciences, Czech University of Life Sciences Prague, CZ-16500 Prague, Czech Republic
| | - Martina Outlá
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics of the Czech Academy of Sciences, Brno CZ-61200, Czech Republic.,National Centre for Biomolecular Research, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic
| | - Jiří Salát
- Veterinary Research Institute, Emerging Viral Diseases, Brno CZ-62100, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-37005 Ceske Budejovice, Czech Republic
| | - Eva Konkol'ová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy ofSciences, CZ-16000 Prague, Czech Republic
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry of the Czech Academy ofSciences, CZ-16000 Prague, Czech Republic
| | - Daniel Růžek
- Veterinary Research Institute, Emerging Viral Diseases, Brno CZ-62100, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-62500 Brno, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-37005 Ceske Budejovice, Czech Republic
| | - Michaela Vorlíčková
- Department of Biophysics of Nucleic Acids, Institute of Biophysics of the Czech Academy of Sciences, Brno CZ-61200, Czech Republic
| | - Luděk Eyer
- Veterinary Research Institute, Emerging Viral Diseases, Brno CZ-62100, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, CZ-37005 Ceske Budejovice, Czech Republic
| | - Daniel Renčiuk
- Department of Biophysics of Nucleic Acids, Institute of Biophysics of the Czech Academy of Sciences, Brno CZ-61200, Czech Republic
| |
Collapse
|
7
|
Dhawan S. Therapeutic Potential of Inducible Endogenous Cytoprotective Heme Oxygenase-1 in Mitigating SARS-CoV-2 Infection and Associated Inflammation. Antioxidants (Basel) 2022; 11:662. [PMID: 35453347 PMCID: PMC9028590 DOI: 10.3390/antiox11040662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
The inducible cytoprotective enzyme heme oxygenase-1 (HO-1) has gained significant recognition in recent years for mediating strong cellular resistance to a broad range of viral infections, regardless of the type of viruses, viral strains, or mutants. HO-1 is not a typical antiviral agent that targets any particular pathogen. It is a "viral tropism independent" endogenous host defense factor that upon induction provides general cellular protection against pathogens. By virtue of HO-1 being widely distributed intracellular enzyme in virtually every cell, this unique host factor presents a novel class of generic host defense system against a variety of viral infections. This Viewpoint proposes pharmacological evaluation of the HO-1-dependent cellular resistance for its potential in mitigating infections by deadly viruses, including the current severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), its variants, and mutants. HO-1-dependent cellular resistance against SARS-CoV-2 can complement current medical modalities for much effective control of the COVID-19 pandemic, especially with constantly emerging new viral variants and limited therapeutic options to treat SARS-CoV-2 infection and associated severe health consequences.
Collapse
Affiliation(s)
- Subhash Dhawan
- Retired Senior FDA Research & Regulatory Scientist, 9890 Washingtonian Blvd., #703, Gaithersburg, MD 20878, USA
| |
Collapse
|
8
|
A Journey into the Clinical Relevance of Heme Oxygenase 1 for Human Inflammatory Disease and Viral Clearance: Why Does It Matter on the COVID-19 Scene? Antioxidants (Basel) 2022; 11:antiox11020276. [PMID: 35204159 PMCID: PMC8868141 DOI: 10.3390/antiox11020276] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 01/27/2023] Open
Abstract
Heme oxygenase 1 (HO-1), the rate-limiting enzyme in heme degradation, is involved in the maintenance of cellular homeostasis, exerting a cytoprotective role by its antioxidative and anti-inflammatory functions. HO-1 and its end products, biliverdin, carbon monoxide and free iron (Fe2+), confer cytoprotection against inflammatory and oxidative injury. Additionally, HO-1 exerts antiviral properties against a diverse range of viral infections by interfering with replication or activating the interferon (IFN) pathway. Severe cases of coronavirus disease 2019 (COVID-19), an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), are characterized by systemic hyperinflammation, which, in some cases, leads to severe or fatal symptoms as a consequence of respiratory failure, lung and heart damage, kidney failure, and nervous system complications. This review summarizes the current research on the protective role of HO-1 in inflammatory diseases and against a wide range of viral infections, positioning HO-1 as an attractive target to ameliorate clinical manifestations during COVID-19.
Collapse
|
9
|
Bagyi J, Sripada V, Aidone AM, Lin HY, Ruder EH, Crawford DR. Dietary rational targeting of redox-regulated genes. Free Radic Biol Med 2021; 173:19-28. [PMID: 34274490 DOI: 10.1016/j.freeradbiomed.2021.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Nutrigenomics is the study of how food and associated nutrients affect gene expression. This field sits at the intersection of diet, the genome and health with the ultimate goal of exploiting its understanding to design a precision nutrition strategy for humans. We have studied diet and nutrigenomics in the context of something we call "dietary rational gene targeting." Here, healthy diet is used to alter disease-causing gene expression back toward the normal to treat various diseases and conditions while lowering treatment cost and toxicity. In this paper, we discuss the use of this strategy to modulate the expression of redox-associated genes to improve human health. Most human disorders are associated, at least to some extent, with oxidative stress and so treatments (including diet) that target redox-related genes have major potential clinical significance. Healthy dietary options here are wide-ranging and include whole foods and botanical-based beverages. In some cases, botanical supplements may also be useful gene modulators although their health benefits are less clear. Key redox gene targets for these dietary agents include antioxidant genes, related transcription factors, detoxification genes, and DNA repair genes. Other important considerations include bioavailability, the contribution of the microbiome, and advancing technologies. In this review, specific examples of redox associated genes and pathologies and their potential treatment with healthy diet are presented to illustrate our approach. This will also serve as a foundation for the design of future clinical studies to improve diet-related health.
Collapse
Affiliation(s)
- Joyce Bagyi
- Clinical Nutrition, Albany Medical Center, Albany, NY, 12208, USA
| | - Veda Sripada
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Andrea M Aidone
- Clinical Nutrition, Albany Medical Center, Albany, NY, 12208, USA
| | - H-Y Lin
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Wan Fang Hospital, Cancer Center, Taipei Medical University, Taipei, 11031, Taiwan
| | - Elizabeth H Ruder
- Wegmans School of Health and Nutrition, College of Health Science and Technology, Rochester Institute of Technology, Rochester, NY, 14620, USA
| | - Dana R Crawford
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA.
| |
Collapse
|
10
|
Huang H, Dabrazhynetskaya A, Pluznik J, Zheng J, Wu Y, Chizhikov V, Buehler PW, Yamada KM, Dhawan S. Hemin activation abrogates Mycoplasma hyorhinis replication in chronically infected prostate cancer cells via heme oxygenase-1 induction. FEBS Open Bio 2021; 11:2727-2739. [PMID: 34375508 PMCID: PMC8487054 DOI: 10.1002/2211-5463.13271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/16/2021] [Accepted: 08/09/2021] [Indexed: 11/20/2022] Open
Abstract
Mycoplasma hyorhinis (M. hyorhinis) lacks a cell wall and resists multiple antibiotics. We describe here the striking > 90% inhibitory effect of hemin, a natural inducer of the cytoprotective enzyme heme oxygenase‐1 (HO‐1), on M. hyorhinis replication in chronically infected LNCaP prostate cancer cells. The role of HO‐1 in interrupting M. hyorhinis replication was confirmed by HO‐1‐specific siRNA suppression of hemin‐induced HO‐1 protein expression, which increased intracellular M. hyorhinis DNA levels in LNCaP cells. Proteomic analysis and transmission electron microscopy of hemin‐treated cells confirmed the complete absence of M. hyorhinis proteins and intact microorganisms, respectively, strongly supporting these findings. Our study is the first to our knowledge suggesting therapeutic potential for activated HO‐1 in cellular innate responses against mycoplasma infection.
Collapse
Affiliation(s)
- Hanxia Huang
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring
| | - Alena Dabrazhynetskaya
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring
| | - Jacob Pluznik
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring
| | - Jiwen Zheng
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring
| | - Yong Wu
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring
| | - Vladimir Chizhikov
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring
| | - Paul W Buehler
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring.,Department of Pathology, Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore
| | - Kenneth M Yamada
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda
| | - Subhash Dhawan
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring.,Retired Senior FDA Research & Regulatory Scientist, 9890 Washingtonian Blvd., #703, Gaithersburg, 20878
| |
Collapse
|
11
|
Lu Z, Zhan F, Yang M, Li F, Shi F, Li Y, Zhang M, Zhao L, Zhang K, Li J, Lin L, Qin Z. The immune function of heme oxygenase-1 from grass carp (Ctenopharyngodon idellus) in response to bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2021; 112:168-178. [PMID: 32927052 DOI: 10.1016/j.fsi.2020.08.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/18/2020] [Accepted: 08/29/2020] [Indexed: 06/11/2023]
Abstract
Heme oxygenase (HO)-1, a rate-limiting enzyme in heme catabolism, results in the formation of equivalent amounts of biliverdin (BV), carbon monoxide (CO) and ferrous iron (Fe2+). Previous studies have revealed that HO-1 plays an important role in immune responses. However, the mechanism underlying the immune responses against bacterial infection of teleost HO-1 remains enigmatic. To decipher the mechanisms, we have cloned and characterized the HO-1 gene of grass carp (designated as GcHO-1) in this research. The results showed that the open reading frame (ORF) of GcHO-1 was 819 bp, which encoded a putative protein of 272 amino acids. The deduced amino acid sequence phylogenetically shared the highest identity with other teleosts, and contained two domains of heme-oxygenase and a single-pass transmembrane domain. The mRNA expressions of GcHO-1 in healthy grass carp have widely existed in examined tissues in the following order of spleen > head-kidney > middle head-kidney > intestines > liver > gills > heart > muscle > brain. Besides, the mRNA and protein transcription of GcHO-1 were both significantly up-regulated in the liver and head-kidney tissues after Staphylococcus aureus and Aeromonas hydrophila infection. In addition, overexpression of GcHO-1 in kidney cell line (CIK) cells of grass carp could reduce the expression of inflammatory cytokines (IL-1β, IL-8, TNFα, CCL1 and IL-6). Herein, we demonstrate that GcHO-1 plays an anti-inflammatory role in innate immunity. Our results shed new light on the mechanisms underlying the antibacterial immunity of teleost.
Collapse
Affiliation(s)
- Zhijie Lu
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Fanbin Zhan
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Minxuan Yang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Fenglin Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Fei Shi
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Yanan Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Menglan Zhang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Lijuan Zhao
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Kai Zhang
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China
| | - Jun Li
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China; School of Biological Sciences, Lake Superior State University, Sault Ste. Marie, MI, 49783, USA
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong Province, 510222, China.
| |
Collapse
|
12
|
You L, Chen J, Liu W, Xiang Q, Luo Z, Wang W, Xu W, Wu K, Zhang Q, Liu Y, Wu J. Enterovirus 71 induces neural cell apoptosis and autophagy through promoting ACOX1 downregulation and ROS generation. Virulence 2021; 11:537-553. [PMID: 32434419 PMCID: PMC7250321 DOI: 10.1080/21505594.2020.1766790] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Enterovirus 71 (EV71) infection causes hand, foot, and mouth disease (HFMD), and even fatal neurological complications. However, the mechanisms underlying EV71 neurological pathogeneses are largely unknown. This study reveals a distinct mechanism by which EV71 induces apoptosis and autophagy in neural cells. EV71 non-structure protein 3D (also known as RNA-dependent RNA polymerase, RdRp) interacts with the peroxisomal protein acyl-CoA oxidase 1 (ACOX1), and contributes to ACOX1 downregulation. Further studies demonstrate that EV71 reduces peroxisome numbers. Additionally, knockdown of ACOX1 or peroxin 19 (PEX19) induces apoptosis and autophagy in neural cells including human neuroblastoma (SK-N-SH) cells and human astrocytoma (U251) cells, and EV71 infection induces neural cell death through attenuating ACOX1 production. Moreover, EV71 infection and ACOX1 knockdown facilitate reactive oxygen species (ROS) production and attenuate the cytoprotective protein deglycase (DJ-1)/Nuclear factor erythroid 2-related factor 2 (NRF2)/Heme oxygenase 1 (HO-1) pathway (DJ-1/NRF2/HO-1), which collectively result in ROS accumulation in neural cells. In conclusion, EV71 downregulates ACOX1 protein expression, reduces peroxisome numbers, enhances ROS generation, and attenuates the DJ-1/NRF2/HO-1 pathway, thereby inducing apoptosis and autophagy in neural cells. These findings provide new insights into the mechanism underlying EV71-induced neural pathogenesis, and suggest potential treatments for EV71-associated diseases.
Collapse
Affiliation(s)
- Lei You
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Junbo Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Weiyong Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qi Xiang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhen Luo
- Guangzhou Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Wenbiao Wang
- Guangzhou Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Wei Xu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,Guangzhou Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,Guangzhou Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
13
|
Wang Y, Yuyin D, Fengyang C, Xukang Z, Jianliang L. Heme Oxygenase-1 suppresses duck Tembusu virus replication in vitro. Vet Microbiol 2020; 251:108885. [PMID: 33157356 DOI: 10.1016/j.vetmic.2020.108885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
Heme Oxygenase-1 (HO-1) is a ubiquitously expressed enzyme which plays important functions in antioxidant, anti-inflammatory and anti-apoptosis. Recent studies have demonstrated that HO-1 also has significant antiviral properties, inhibiting the replication of some kinds of viruses such as human immunodeficiency virus (HIV), hepatitis C virus (HCV), and dengue fever virus (DFV). In this study, we evaluated the role of HO-1 in Duck Tembusu virus (DTMUV) replication in vitro. The results showed that, the mRNA expression level of HO-1 was transient up-regulated and then significantly decreased in duck embryo fibroblast (DEF) infected with DTMUV. HO-1 induction by transfection of HO-1 over-expression plasmid or treatment with cobalt protoporphyrin (CoPP), a potent HO-1 inducer, could inhibit DTMUV replication effectively. In contrast, HO-1 siRNA knockdown in DEF increased DTMUV replication, implied that HO-1 was an important cellular factor against DTMUV replication. Furthermore, we found that ferric ion (Fe3+) but not biliverdin and carbon monoxide, products of heme degradation by HO-1, mediated the HO-1-induced anti-DTMUV effect. Overall, these finding revealed that a drug induced the HO-1 signal pathway was a promising strategy for treating DTMUV infection.
Collapse
Affiliation(s)
- Yixin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Du Yuyin
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Cao Fengyang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Zhang Xukang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Li Jianliang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China.
| |
Collapse
|
14
|
Batra N, De Souza C, Batra J, Raetz AG, Yu AM. The HMOX1 Pathway as a Promising Target for the Treatment and Prevention of SARS-CoV-2 of 2019 (COVID-19). Int J Mol Sci 2020; 21:E6412. [PMID: 32899231 PMCID: PMC7503392 DOI: 10.3390/ijms21176412] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease of 2019 (COVID-19) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a global pandemic with increasing incidence and mortality rates. Recent evidence based on the cytokine profiles of severe COVID-19 cases suggests an overstimulation of macrophages and monocytes associated with reduced T-cell abundance (lymphopenia) in patients infected with SARS-CoV-2. The SARS-CoV-2 open reading frame 3 a (ORF3a) protein was found to bind to the human HMOX1 protein at a high confidence through high-throughput screening experiments. The HMOX1 pathway can inhibit platelet aggregation, and can have anti-thrombotic and anti-inflammatory properties, amongst others, all of which are critical medical conditions observed in COVID-19 patients. Here, we review the potential of modulating the HMOX1-ORF3a nexus to regulate the innate immune response for therapeutic benefits in COVID-19 patients. We also review other potential treatment strategies and suggest novel synthetic and natural compounds that may have the potential for future development in clinic.
Collapse
Affiliation(s)
- Neelu Batra
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
| | - Cristabelle De Souza
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Jyoti Batra
- Gladstone Institute, San Francisco, CA 94158, USA;
| | - Alan G. Raetz
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA; (N.B.); (C.D.S.); (A.G.R.)
| |
Collapse
|
15
|
Hu K, Zhu Z, Mathahs MM, Tran H, Bommer J, Testa CA, Schmidt WN. Metalloprotoporphyrin Inhibition of HCV NS3-4A Protease: Structure-Activity Relationships. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:757-771. [PMID: 32158194 PMCID: PMC7048954 DOI: 10.2147/dddt.s201089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022]
Abstract
Background Antiviral actions of tetrapyrroles have been described in a number of systems. Our goal was to evaluate antagonism of the HCV NS3-4A protease by a variety of common porphyrins and characterize structure-activity relationships that may be useful for future drug design of HCV and related Flaviviruses. Methods Using fluorometric assays, common metalloprotoporphyrins (MPP) all inhibited NS3-4A protease with IC50 values in low micromolar ranges [CoPP (1.4 µM) < ZnPP = MnPP = SnPP < CuPP < FePP (6.5 µM) = protoporphyrin]. Results Lineweaver-Burk plots confirmed that MPP: NS3 inhibition was basically competitive. All tested MPPs inhibited HCV genotype 1A, 1B, 2A and 3A recombinant proteases with the same fidelity suggesting wide antagonistic capabilities. However, when the MPPs were tested in cellular incubations with HCV replicons only Zn, Fe and free-base protoporphyrin showed comparable EC50 and IC50 values suggesting that there may be critical differences in MPP uptake and intracellular availability. Meso, deutero, and isohematoporphyrin derivatives, with or without metal substitution, all showed less anti-protease and antiviral activities as compared to protoporphyrins, suggesting that the planar, vinyl side chains are important for protease active site binding. MPPs were also active against three common protease mutants (T54A, A156T, and V36M) with equivalent or better IC50 values as compared to wild type enzyme. Conclusion These findings document the versatility of MPPs as antiviral agents with an expanded sensitivity for HCV genotypes and resistance to some common viral mutations. The results also suggest that further study of MPP structure and function will be useful for the development of new antiviral agents.
Collapse
Affiliation(s)
- Katherine Hu
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA 52246, USA.,Department of Internal Medicine of the Roy G. And Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Zhaowen Zhu
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA 52246, USA.,Department of Internal Medicine of the Roy G. And Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Meleah M Mathahs
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA 52246, USA.,Department of Internal Medicine of the Roy G. And Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Huy Tran
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA 52246, USA.,Department of Internal Medicine of the Roy G. And Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jerry Bommer
- Frontier Scientific, Logan, UT 84321, USA.,Echelon Biosciences Inc, Salt Lake City, UT 84108, USA
| | | | - Warren N Schmidt
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA 52246, USA.,Department of Internal Medicine of the Roy G. And Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
16
|
Li C, Li L, Jin L, Yuan J. Heme Oxygenase-1 inhibits spring viremia of carp virus replication through carbon monoxide mediated cyclic GMP/Protein kinase G signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2018; 79:65-72. [PMID: 29753142 DOI: 10.1016/j.fsi.2018.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 06/08/2023]
Abstract
Spring viremia of carp virus (SVCV) is the etiological agent of spring viremia of carp (SVC) and causes mass mortality in common carp (Cyprinus carpio). Currently, no effective treatments or commercial vaccines against SVCV are available. Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Previous studies demonstrated that nuclear factor-erythroid 2 related factor 2 (Nrf2) functions as an important upstream regulator of HO-1 and exhibits robust activity against SVCV infection. In this study, we further examined the antiviral activity of HO-1 against SVCV infection. The elevated expression of HO-1 was induced upon cobalt protoporphyrin (CoPP) treatment in EPC cells without affecting cell viability and thus inhibited SVCV replication in a dose dependent manner. Knocking down of HO-1 rescued SVCV replication. Thereby, the antiviral activity of ROS/Nrf2/HO-1 axis was confirmed in EPC cells. Furthermore, HO-1 enzymatic products CO, but not biliverdin, markedly inhibited SVCV replication via the activation of cyclic GMP/protein kinase G signaling pathway. Collectively, these findings suggest potential drug or therapy that induced the Nrf2/HO-1/CO/cGMP/PKG signaling pathway as a promising strategy for treating SVC.
Collapse
Affiliation(s)
- Cong Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Lijuan Li
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China; Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan, 430070, People's Republic of China
| | - Ling Jin
- Department of Biomedical Science, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97330, United States
| | - Junfa Yuan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China; Hubei Engineering Research Center for Aquatic Animal Diseases Control and Prevention, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
17
|
Tseng CH, Lin CK, Chen YL, Tseng CK, Lee JY, Lee JC. Discovery of naphtho[1,2-d]oxazole derivatives as potential anti-HCV agents through inducing heme oxygenase-1 expression. Eur J Med Chem 2018; 143:970-982. [DOI: 10.1016/j.ejmech.2017.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/30/2017] [Accepted: 12/02/2017] [Indexed: 01/05/2023]
|
18
|
Chmielewski P, Strzelec B, Chmielowiec J, Chmielowiec K, Borysławski K. Association of serum bilirubin with longevity: Evidence from a retrospective longitudinal study and cross-sectional data. ANTHROPOLOGICAL REVIEW 2017. [DOI: 10.1515/anre-2017-0024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Abstract
Bilirubin is a potent antioxidant and an important anti-inflammatory factor. Therefore, there has been an increasing focus on serum bilirubin as a negative risk factor of cardiovascular mortality in men and an indicator of improved survival in both sexes, but the direct mechanisms of these links and the causes of sex differences are not well understood. Moreover, the evidence from longitudinal studies on effects of bilirubin on longevity is limited. In this study, we retrospectively analyzed two groups of older adults to explore age-dependent changes in serum bilirubin levels and their associations with long-term survival in both sexes. Longitudinal data from 142 individuals (68 men and 74 women) aged 45 to 70 years were compared with cross-sectional data from 225 individuals (113 men and 112 women). The latter group was divided into four categories of survival, i.e. 53, 63, 68, and 76+ based on data on lifespan. ANOVA, t-test, and regression analysis were run. The analysis of the longitudinal data showed an increase in serum total bilirubin levels in men (0.3038e0.093x, R2 = 0.667) and women (0.1838e0.0187x, R2 = 0.950), while the analysis of cross-sectional data revealed a U-shaped pattern of age-related changes in men (0.001x2 - 0.1263x + 4.4524, R2 = 0.999) but an inverted U-shaped pattern in women (0.0006x2 + 0.072x - 1.6924, R2 = 0.195). On balance, these results suggest that elevated but normal bilirubin levels might confer a survival advantage in older men but not women. Alternatively, the positive relationship between serum total bilirubin and lifespan was not causal but coincidental. Further studies are needed to elucidate the direct mechanisms of the association between serum bilirubin levels and longevity in elderly people of both sexes.
Collapse
Affiliation(s)
- Piotr Chmielewski
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Poland
| | - Bartłomiej Strzelec
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Poland
- Department and Clinic of Gastrointestinal and General Surgery, Wroclaw Medical University, Poland
| | - Jolanta Chmielowiec
- Faculty of Medicine and Health Sciences, The University of Zielona Gora , Poland
| | | | - Krzysztof Borysławski
- Department of Anthropology, Wroclaw University of Environmental and Life Sciences, Poland
| |
Collapse
|
19
|
Ibáñez FJ, Farías MA, Retamal-Díaz A, Espinoza JA, Kalergis AM, González PA. Pharmacological Induction of Heme Oxygenase-1 Impairs Nuclear Accumulation of Herpes Simplex Virus Capsids upon Infection. Front Microbiol 2017; 8:2108. [PMID: 29163402 PMCID: PMC5671570 DOI: 10.3389/fmicb.2017.02108] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/16/2017] [Indexed: 12/18/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible enzyme that is expressed in response to physical and chemical stresses, such as ultraviolet radiation, hyperthermia, hypoxia, reactive oxygen species (ROS), as well as cytokines, among others. Its activity can be positively modulated by cobalt protoporphyrin (CoPP) and negatively by tin protoporphirin (SnPP). Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. Importantly, numerous products of HO-1 are cytoprotective with anti-apoptotic, anti-oxidant, anti-inflammatory, and anti-cancer effects. The products of HO-1 also display antiviral properties against several viruses, such as the human immunodeficiency virus (HIV), influenza, hepatitis B, hepatitis C, and Ebola virus. Here, we sought to assess the effect of modulating HO-1 activity over herpes simplex virus type 2 (HSV-2) infection in epithelial cells and neurons. There are no vaccines against HSV-2 and treatment options are scarce in the immunosuppressed, in which drug-resistant variants emerge. By using HSV strains that encode structural and non-structural forms of the green fluorescent protein (GFP), we found that pharmacological induction of HO-1 activity with CoPP significantly decreases virus plaque formation and the expression of virus-encoded genes in epithelial cells as determined by flow cytometry and western blot assays. CoPP treatment did not affect virus binding to the cell surface or entry into the cytoplasm, but rather downstream events in the virus infection cycle. Furthermore, we observed that treating cells with a CO-releasing molecule (CORM-2) recapitulated some of the anti-HSV effects elicited by CoPP. Taken together, these findings indicate that HO-1 activity interferes with the replication cycle of HSV and that its antiviral effects can be recapitulated by CO.
Collapse
Affiliation(s)
- Francisco J Ibáñez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Janyra A Espinoza
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institut National de la Santé et de la Recherche Médicale U1064, Nantes, France
| | - Pablo A González
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
20
|
Liu F, Du Y, Feng WH. New perspective of host microRNAs in the control of PRRSV infection. Vet Microbiol 2017; 209:48-56. [DOI: 10.1016/j.vetmic.2017.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/22/2016] [Accepted: 01/03/2017] [Indexed: 02/09/2023]
|
21
|
Espinoza JA, González PA, Kalergis AM. Modulation of Antiviral Immunity by Heme Oxygenase-1. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:487-493. [PMID: 28082120 DOI: 10.1016/j.ajpath.2016.11.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/05/2016] [Accepted: 11/15/2016] [Indexed: 12/18/2022]
Abstract
Heme oxygenase-1 (HO-1) is a stress-inducible, anti-inflammatory, and cytoprotective enzyme expressed in most cell types in the organism. Under several stress stimuli, HO-1 expression and activity is up-regulated to catalyze the rate-limiting enzymatic step of heme degradation into carbon monoxide, free iron, and biliverdin. Besides its effects on cell metabolism, HO-1 is also capable of modulating host innate and adaptive immune responses in response to sepsis, transplantation, and autoimmunity, and preventing oxidative damage associated with inflammation. In addition, recent studies have reported that HO-1 can exert a significant antiviral activity against a wide variety of viruses, including HIV, hepatitis C virus, hepatitis B virus, enterovirus 71, influenza virus, respiratory syncytial virus, dengue virus, and Ebola virus, among others. Herein, we address the current understanding of the functional significance of HO-1 against a variety of viruses and its potential as a therapeutic strategy to prevent and control viral infections. Furthermore, we review the most important features of the immunoregulatory functions for this enzyme.
Collapse
Affiliation(s)
- Janyra A Espinoza
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Department of Molecular Genetics and Microbiology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM, Combined Research Unit 1064, Nantes University Hospital Nantes, Institute for Transplantation-Urology-Nephrology, Université de Nantes, Faculty of Medicine, Nantes, France; Department of Endocrinology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
22
|
Huang H, Falgout B, Takeda K, Yamada KM, Dhawan S. Nrf2-dependent induction of innate host defense via heme oxygenase-1 inhibits Zika virus replication. Virology 2017; 503:1-5. [PMID: 28068513 DOI: 10.1016/j.virol.2016.12.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/15/2016] [Accepted: 12/18/2016] [Indexed: 01/09/2023]
Abstract
We identified primary human monocyte-derived macrophages (MDM) as vulnerable target cells for Zika virus (ZIKV) infection. We demonstrate dramatic effects of hemin, the natural inducer of the heme catabolic enzyme heme oxygenase-1 (HO-1), in the reduction of ZIKV replication in vitro. Both LLC-MK2 monkey kidney cells and primary MDM exhibited hemin-induced HO-1 expression with major reductions of >90% in ZIKV replication, with little toxicity to infected cells. Silencing expression of HO-1 or its upstream regulatory gene, nuclear factor erythroid-related factor 2 (Nrf2), attenuated hemin-induced suppression of ZIKV infection, suggesting an important role for induction of these intracellular mediators in retarding ZIKV replication. The inverse correlation between hemin-induced HO-1 levels and ZIKV replication provides a potentially useful therapeutic modality based on stimulation of an innate cellular response against Zika virus infection.
Collapse
Affiliation(s)
- Hanxia Huang
- Food and Drug Administration, Silver Spring, MD, United States
| | - Barry Falgout
- Food and Drug Administration, Silver Spring, MD, United States
| | - Kazuyo Takeda
- Food and Drug Administration, Silver Spring, MD, United States
| | | | - Subhash Dhawan
- Food and Drug Administration, Silver Spring, MD, United States.
| |
Collapse
|
23
|
Xiao S, Du T, Wang X, Ni H, Yan Y, Li N, Zhang C, Zhang A, Gao J, Liu H, Pu F, Zhang G, Zhou EM. MiR-22 promotes porcine reproductive and respiratory syndrome virus replication by targeting the host factor HO-1. Vet Microbiol 2016; 192:226-230. [PMID: 27527787 DOI: 10.1016/j.vetmic.2016.07.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/29/2016] [Accepted: 07/31/2016] [Indexed: 11/30/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most economically important viruses affecting the swine industry worldwide. MicroRNAs (miRNAs) play vital roles in virus-host interactions by regulating the expression of viral or host gene at posttranscriptional level. Our previous research showed that PRRSV infection down-regulates the expression of heme oxygenase-1 (HO-1), a pivotal cytoprotective enzyme, and overexpression of HO-1 inhibits PRRSV replication. In this study, we demonstrate that host miRNA miR-22 can downregulate HO-1 expression by directly targeting its 3' untranslated region. Suppression of HO-1 expression by miR-22 facilitates PRRSV replication. This work suggests that PRRSV may utilize cellular miRNA to modify antiviral host factor expression, enabling viral replication, which not only provides new insights into virus-host interactions during PRRSV infection, but also suggests potential therapies for PRRSV infection.
Collapse
Affiliation(s)
- Shuqi Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Taofeng Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Xue Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Huaibao Ni
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Yunhuan Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Na Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Chong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Angke Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Jiming Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Hongliang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Fengxing Pu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| | - Gaiping Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China.
| | - En-Min Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, Yangling, Shaanxi 712100, China.
| |
Collapse
|
24
|
Assunção-Miranda I, Cruz-Oliveira C, Neris R, Figueiredo C, Pereira L, Rodrigues D, Araujo D, Da Poian A, Bozza M. Inactivation of Dengue and Yellow Fever viruses by heme, cobalt-protoporphyrin IX and tin-protoporphyrin IX. J Appl Microbiol 2016; 120:790-804. [DOI: 10.1111/jam.13038] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/16/2015] [Accepted: 12/22/2015] [Indexed: 12/25/2022]
Affiliation(s)
- I. Assunção-Miranda
- Instituto de Microbiologia Paulo de Góes; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - C. Cruz-Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - R.L.S. Neris
- Instituto de Microbiologia Paulo de Góes; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - C.M. Figueiredo
- Instituto de Microbiologia Paulo de Góes; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - L.P.S. Pereira
- Instituto de Bioquímica Médica Leopoldo de Meis; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - D. Rodrigues
- Instituto de Microbiologia Paulo de Góes; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - D.F.F. Araujo
- Instituto de Microbiologia Paulo de Góes; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - A.T. Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| | - M.T. Bozza
- Instituto de Microbiologia Paulo de Góes; Universidade Federal do Rio de Janeiro; Rio de Janeiro Brazil
| |
Collapse
|
25
|
Huang H, Zhou ZH, Adhikari R, Yamada KM, Dhawan S. Defective iron homeostasis in human immunodeficiency virus type-1 latency. CURRENT TRENDS IN IMMUNOLOGY 2016; 17:125-131. [PMID: 28824260 PMCID: PMC5562369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Highly active antiretroviral therapy has significantly improved the life of HIV-1-infected individuals, yet complete eradication of HIV-1 reservoirs (i.e., latently infected cells) remains a major challenge. We have previously shown that induction of the endogenous cytoprotective enzyme heme oxygenase-1 (HO-1) by its natural substrate hemin reduces susceptibility of T cells and macrophages to HIV-1 infection. In the present study, we demonstrate that hemin treatment stimulated virus production by latently infected ACH-2 cells, followed by cellular toxicity and death when stimulated with TNF-α or co-cultured with monocyte-derived macrophages (MDM). This cytotoxicity was associated with low levels of the iron-binding protein ferritin and the iron transporter ferroportin with lack of hemoglobin catabolic enzyme HO-1, resulting in substantial iron accumulation in the activated ACH-2 cells. Defective iron homeostasis in ACH-2 cells provides a model system for selective targeting of the latent HIV-1 reservoir by hemin-induced iron toxicity.
Collapse
Affiliation(s)
- Hanxia Huang
- Food and Drug Administration, National Institutes of Health, Bethesda, Maryland, USA
| | - Zhao-Hua Zhou
- Food and Drug Administration, National Institutes of Health, Bethesda, Maryland, USA
| | - Rewati Adhikari
- Food and Drug Administration, National Institutes of Health, Bethesda, Maryland, USA
| | - Kenneth M Yamada
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Subhash Dhawan
- Food and Drug Administration, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
26
|
Huang H, Konduru K, Solovena V, Zhou ZH, Kumari N, Takeda K, Nekhai S, Bavari S, Kaplan GG, Yamada KM, Dhawan S. Therapeutic potential of the heme oxygenase-1 inducer hemin against Ebola virus infection. CURRENT TRENDS IN IMMUNOLOGY 2016; 17:117-123. [PMID: 28133423 PMCID: PMC5267496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Promising drugs to treat Ebola virus (EBOV) infection are currently being developed, but so far none has shown efficacy in clinical trials. Drugs that can stimulate host innate defense responses may retard the progression of EBOV disease. We report here the dramatic effect of hemin, the natural inducer of the heme catabolic enzyme heme oxygenase-1 (HO-1), in the reduction of EBOV replication. Treatment of primary monocyte-derived macrophages (MDM), Vero E6 cells, HeLa cells, and human foreskin fibroblasts (HFF1) with hemin reduced EBOV infection by >90%, and showed minimal toxicity to infected cells. Inhibition of HO-1 enzymatic activity and silencing HO-1 expression prevented the hemin-mediated suppression of EBOV infection, suggesting an important role for induction of this intracellular mediator in restricting EBOV replication. The inverse correlation between hemin-induced HO-1 and EBOV replication provides a potentially useful therapeutic modality based on the stimulation of an innate cellular response against Ebola infection.
Collapse
Affiliation(s)
- Hanxia Huang
- Food and Drug Administration, Silver Spring, Maryland
| | | | - Veronica Solovena
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Zhao-Hua Zhou
- Food and Drug Administration, Silver Spring, Maryland
| | - Namita Kumari
- Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington DC
| | - Kazuyo Takeda
- Food and Drug Administration, Silver Spring, Maryland
| | - Sergei Nekhai
- Center for Sickle Cell Disease, Department of Medicine, Howard University, Washington DC
| | - Sina Bavari
- Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | | | - Kenneth M. Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
27
|
Corral-Jara KF, Trujillo-Ochoa JL, Realpe M, Panduro A, Roman S, Fierro NA. Rethinking the immune properties of bilirubin in viral hepatitis: from bench to bedside. Clin Transl Immunology 2015; 4:e54. [PMID: 26719800 PMCID: PMC4685441 DOI: 10.1038/cti.2015.37] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/04/2015] [Accepted: 11/15/2015] [Indexed: 02/07/2023] Open
Abstract
Communication between the immune system and metabolic components can be exemplified by the process of heme catabolism. The immunomodulatory functions of the enzymes, substrates and active products related to catabolism of the heme group have been extensively studied. Bilirubin (BR), the final breakdown product of heme, is primarily considered to be a toxic waste product but has recently been considered to be an immunomodulatory metabolite. Through mechanisms that include intracellular signaling and transcriptional control, BR affects those immune cell functions that regulate cell proliferation, differentiation and apoptosis. During the pathogenesis of viral hepatitis, the heme degradation pathway is disrupted, resulting in changes to normal BR concentrations. These alterations have been previously studied mainly as a consequence of the infection. However, little is known about the potential immunomodulatory role played by BR in the development of infectious hepatocellular diseases. Differences in BR levels in the context of viral hepatitis are likely to provide important insights into the metabolite-mediated mechanisms controlling the immune responses underlying both the long-term persistence of hepatitis C virus (HCV) infection and the resolution of hepatitis A virus (HAV) infection during the acute phase. In this review, the cross-talk between heme catabolism and immune function is described in detail. Special emphasis is given to discoveries that hold promise for identifying immunologic features of metabolic products in the resolution of viral diseases.
Collapse
Affiliation(s)
- Karla F Corral-Jara
- Unidad de Inmunovirología, Servicio de Biología Molecular en Medicina, Hospital Civil de Guadalajara 'Fray Antonio Alcalde' , Guadalajara, Mexico ; Departamento de Biología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, Mexico
| | - Jorge L Trujillo-Ochoa
- Unidad de Inmunovirología, Servicio de Biología Molecular en Medicina, Hospital Civil de Guadalajara 'Fray Antonio Alcalde' , Guadalajara, Mexico ; Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, Mexico
| | - Mauricio Realpe
- Departamento de Medicina Veterinaria, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara , Guadalajara, Mexico
| | - Arturo Panduro
- Servicio de Biología Molecular en Medicina, Hospital Civil of Guadalajara 'Fray Antonio Alcalde' , Guadalajara, Mexico ; Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, Mexico
| | - Sonia Roman
- Departamento de Biología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, Mexico ; Servicio de Biología Molecular en Medicina, Hospital Civil of Guadalajara 'Fray Antonio Alcalde' , Guadalajara, Mexico
| | - Nora A Fierro
- Unidad de Inmunovirología, Servicio de Biología Molecular en Medicina, Hospital Civil de Guadalajara 'Fray Antonio Alcalde' , Guadalajara, Mexico ; Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara , Guadalajara, Mexico
| |
Collapse
|
28
|
Heme Oxygenase-1 Suppresses Bovine Viral Diarrhoea Virus Replication in vitro. Sci Rep 2015; 5:15575. [PMID: 26510767 PMCID: PMC4625146 DOI: 10.1038/srep15575] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 09/28/2015] [Indexed: 02/07/2023] Open
Abstract
Viral cycle progression depends upon host-cell processes in infected cells, and this is true for bovine viral diarrhoea virus (BVDV), the causative agent of BVD that is a worldwide threat to the bovine industry. Heme oxygenase-1 (HO-1) is a ubiquitously expressed inducible isoform of the first and rate-limiting enzyme for heme degradation. Recent studies have demonstrated that HO-1 has significant antiviral properties, inhibiting the replication of viruses such as ebola virus, human immunodeficiency virus, hepatitis C virus, and porcine reproductive and respiratory syndrome virus. However, the function of HO-1 in BVDV infection is unclear. In the present study, the relationship between HO-1 and BVDV was investigated. In vitro analysis of HO-1 expression in BVDV-infected MDBK cells demonstrated that a decrease in HO-1 as BVDV replication increased. Increasing HO-1 expression through adenoviral-mediated overexpression or induction with cobalt protoporphyrin (CoPP, a potent HO-1 inducer), pre- and postinfection, effectively inhibited BVDV replication. In contrast, HO-1 siRNA knockdown in BVDV-infected cells increased BVDV replication. Therefore, the data were consistent with HO-1 acting as an anti-viral factor and these findings suggested that induction of HO-1 may be a useful prevention and treatment strategy against BVDV infection.
Collapse
|
29
|
Takeda K, Adhikari R, Yamada KM, Dhawan S. Hemin activation of innate cellular response blocks human immunodeficiency virus type-1-induced osteoclastogenesis. Biochem Biophys Res Commun 2015; 464:7-12. [PMID: 25998388 DOI: 10.1016/j.bbrc.2015.05.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/11/2015] [Indexed: 12/23/2022]
Abstract
The normal skeletal developmental and homeostatic process termed osteoclastogenesis is exacerbated in numerous pathological conditions and causes excess bone loss. In cancer and HIV-1-infected patients, this disruption of homeostasis results in osteopenia and eventual osteoporesis. Counteracting the factors responsible for these metabolic disorders remains a challenge for preventing or minimizing this co-morbidity associated with these diseases. In this report, we demonstrate that a hemin-induced host protection mechanism not only suppresses HIV-1 associated osteoclastogenesis, but it also exhibits anti-osteoclastogenic activity for non-infected cells. Since the mode of action of hemin is both physiological and pharmacological through induction of heme oxygenase-1 (HO-1), an endogenous host protective response to an FDA-licensed therapeutic used to treat another disease, our study suggests an approach to developing novel, safe and effective therapeutic strategies for treating bone disorders, because hemin administration in humans has previously met required FDA safety standards.
Collapse
Affiliation(s)
- Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Rewati Adhikari
- Division of Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA
| | - Kenneth M Yamada
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Subhash Dhawan
- Division of Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, USA.
| |
Collapse
|
30
|
Transcriptomic Analysis of mRNAs in Human Monocytic Cells Expressing the HIV-1 Nef Protein and Their Exosomes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:492395. [PMID: 25961023 PMCID: PMC4413250 DOI: 10.1155/2015/492395] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023]
Abstract
The Nef protein of human immunodeficiency virus (HIV) promotes viral replication and progression to AIDS. Besides its well-studied effects on intracellular signaling, Nef also functions through its secretion in exosomes, which are nanovesicles containing proteins, microRNAs, and mRNAs and are important for intercellular communication. Nef expression enhances exosome secretion and these exosomes can enter uninfected CD4 T cells leading to apoptotic death. We have recently reported the first miRNome analysis of exosomes secreted from Nef-expressing U937monocytic cells. Here we show genome-wide transcriptome analysis of Nef-expressing U937 cells and their exosomes. We identified four key mRNAs preferentially retained in Nef-expressing cells; these code for MECP2, HMOX1, AARSD1, and ATF2 and are important for chromatin modification and gene expression. Interestingly, their target miRNAs are exported out in exosomes. We also identified three key mRNAs selectively secreted in exosomes from Nef-expressing U937 cells and their corresponding miRNAs being preferentially retained in cells. These are AATK, SLC27A1, and CDKAL and are important in apoptosis and fatty acid transport. Thus, our study identifies selectively expressed mRNAs in Nef-expressing U937 cells and their exosomes and supports a new mode on intercellular regulation by the HIV-1 Nef protein.
Collapse
|
31
|
MicroRNA miR-24-3p promotes porcine reproductive and respiratory syndrome virus replication through suppression of heme oxygenase-1 expression. J Virol 2015; 89:4494-503. [PMID: 25653454 DOI: 10.1128/jvi.02810-14] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most economically important viruses affecting the swine industry worldwide. Our previous research showed that PRRSV downregulates the expression of heme oxygenase-1 (HO-1), a pivotal cytoprotective enzyme, postinfection and that overexpression of HO-1 inhibits PRRSV replication. MicroRNAs regulate gene expression at the posttranscriptional level and have recently been demonstrated to play vital roles in pathogen-host interactions. The present study sought to determine whether microRNAs modulate HO-1 expression and, by doing so, regulate PRRSV replication. Using bioinformatic prediction and experimental verification, we demonstrate that HO-1 expression is regulated by miR-24-3p. A direct interaction between miR-24-3p and HO-1 mRNA was confirmed using a number of approaches. Overexpression of miR-24-3p significantly decreased HO-1 mRNA and protein levels. PRRSV infection induced miR-24-3p expression to facilitate viral replication. The suppressive effect of HO-1 induction by protoporphyrin IX cobalt chloride (CoPP; a classical inducer of HO-1 expression) on PRRSV replication in MARC-145 cells and primary porcine alveolar macrophages could also be reversed by overexpression of miR-24-3p. Collectively, these results suggested that miR-24-3p promotes PRRSV replication through suppression of HO-1 expression, which not only provides new insights into virus-host interactions during PRRSV infection but also suggests potential new antiviral strategies against PRRSV infection. IMPORTANCE MicroRNAs (miRNAs) play vital roles in viral infections by regulating the expression of viral or host genes at the posttranscriptional level. Heme oxygenase-1 (HO-1), a pivotal cytoprotective enzyme, has antiviral activity for a number of viruses, such as Ebola virus, hepatitis C virus, human immunodeficiency virus, and our focus, PRRSV, which causes great economic losses each year in the swine industry worldwide. Here, we show that PRRSV infection induces host miRNA miR-24-3p expression and that miR-24-3p regulates HO-1 expression through both mRNA degradation and translation repression. Suppression of HO-1 expression by miR-24-3p facilitates PRRSV replication. This work lends credibility to the hypothesis that an arterivirus can manipulate cellular miRNAs to enhance virus replication by regulating antiviral responses following viral infection. Therefore, our findings provide new insights into the pathogenesis of PRRSV.
Collapse
|
32
|
Soota K, Maliakkal B. Ribavirin induced hemolysis: A novel mechanism of action against chronic hepatitis C virus infection. World J Gastroenterol 2014; 20:16184-16190. [PMID: 25473172 PMCID: PMC4239506 DOI: 10.3748/wjg.v20.i43.16184] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/11/2014] [Accepted: 07/25/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is not usually cleared by our immune system, leading to the development of chronic hepatitis C infection. Chronic HCV induces the production of various cytokines, predominantly by Kupffer cells (KCs), and creates a pro-inflammatory state in the liver. The chronic dysregulated production of interferon (IFN) and other cytokines by KCs also promotes innate immune tolerance. Ribavirin (RBV) monotherapy has been shown to decrease inflammation in liver of patients with chronic hepatitis C. Sustained virological response (SVR) is significantly higher when IFN is combined with RBV in chronic HCV (cHCV) infection. However, the mechanism of their synergy remains unclear. Previous theories have attempted to explain the anti-HCV effect based on direct action of RBV alone on the virus or on the immune system; however, these theories have serious shortcomings. We propose that hemolysis, which universally occurs with RBV therapy and which is considered a limiting side effect, is precisely the mechanism by which the anti-HCV effect is exerted. Passive hemolysis results in anti-inflammatory/antiviral actions within the liver that disrupt the innate immune tolerance, leading to the synergy of RBV with IFN-α. Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). These metabolites of heme possess anti-inflammatory and antioxidant properties. Thus, HMOX1 plays an extremely important anti-oxidant, anti-inflammatory and cytoprotective role, particularly in KCs and hepatocytes. HMOX1 has been noted to have anti-viral effects in hepatitis C infected cell lines. Additionally, it has been shown to enhance the response to IFN-α by restoring interferon-stimulated genes (ISGs). This mechanism can be clinically corroborated by the following observations that have been found in patients undergoing RBV/IFN combination therapy for cHCV: (1) SVR rates are higher in patients who develop anemia; (2) once anemia (due to hemolysis) occurs, the SVR rate does not depend on the treatment utilized to manage anemia; and (3) ribavirin analogs, such as taribavirin and levovirin, which increase intrahepatic ribavirin levels and which produce lesser hemolysis, are inferior to ribavirin for treating cHCV. This mechanism can also explain the observed RBV synergy with direct antiviral agents. This hypothesis is testable and may lead to newer and safer medications for treating cHCV infection.
Collapse
MESH Headings
- Anemia, Hemolytic/blood
- Anemia, Hemolytic/chemically induced
- Anemia, Hemolytic/immunology
- Animals
- Antiviral Agents/adverse effects
- Antiviral Agents/therapeutic use
- Drug Synergism
- Drug Therapy, Combination
- Heme Oxygenase-1/metabolism
- Hemolysis/drug effects
- Hepacivirus/drug effects
- Hepacivirus/immunology
- Hepatitis C, Chronic/blood
- Hepatitis C, Chronic/diagnosis
- Hepatitis C, Chronic/drug therapy
- Hepatitis C, Chronic/immunology
- Hepatitis C, Chronic/physiopathology
- Hepatitis C, Chronic/virology
- Host-Pathogen Interactions
- Humans
- Inflammation Mediators/metabolism
- Ribavirin/adverse effects
- Ribavirin/therapeutic use
- Treatment Outcome
Collapse
|
33
|
Bhaskar A, Munshi M, Khan SZ, Fatima S, Arya R, Jameel S, Singh A. Measuring glutathione redox potential of HIV-1-infected macrophages. J Biol Chem 2014; 290:1020-38. [PMID: 25406321 PMCID: PMC4294471 DOI: 10.1074/jbc.m114.588913] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Redox signaling plays a crucial role in the pathogenesis of human immunodeficiency virus type-1 (HIV-1). The majority of HIV redox research relies on measuring redox stress using invasive technologies, which are unreliable and do not provide information about the contributions of subcellular compartments. A major technological leap emerges from the development of genetically encoded redox-sensitive green fluorescent proteins (roGFPs), which provide sensitive and compartment-specific insights into redox homeostasis. Here, we exploited a roGFP-based specific bioprobe of glutathione redox potential (EGSH; Grx1-roGFP2) and measured subcellular changes in EGSH during various phases of HIV-1 infection using U1 monocytic cells (latently infected U937 cells with HIV-1). We show that although U937 and U1 cells demonstrate significantly reduced cytosolic and mitochondrial EGSH (approximately −310 mV), active viral replication induces substantial oxidative stress (EGSH more than −240 mV). Furthermore, exposure to a physiologically relevant oxidant, hydrogen peroxide (H2O2), induces significant deviations in subcellular EGSH between U937 and U1, which distinctly modulates susceptibility to apoptosis. Using Grx1-roGFP2, we demonstrate that a marginal increase of about ∼25 mV in EGSH is sufficient to switch HIV-1 from latency to reactivation, raising the possibility of purging HIV-1 by redox modulators without triggering detrimental changes in cellular physiology. Importantly, we show that bioactive lipids synthesized by clinical drug-resistant isolates of Mycobacterium tuberculosis reactivate HIV-1 through modulation of intracellular EGSH. Finally, the expression analysis of U1 and patient peripheral blood mononuclear cells demonstrated a major recalibration of cellular redox homeostatic pathways during persistence and active replication of HIV.
Collapse
Affiliation(s)
- Ashima Bhaskar
- From the Department of Microbiology and Cell Biology, Centre for Infectious Disease and Research, Indian Institute of Sciences, Bangalore 560012
| | - MohamedHusen Munshi
- From the Department of Microbiology and Cell Biology, Centre for Infectious Disease and Research, Indian Institute of Sciences, Bangalore 560012, the Department of Biotechnology, Jamia Millia Islamia, New Delhi 25, India
| | - Sohrab Zafar Khan
- the International Centre for Genetic Engineering and Biotechnology, New Delhi 110 67, and
| | - Sadaf Fatima
- the Department of Biotechnology, Jamia Millia Islamia, New Delhi 25, India
| | - Rahul Arya
- the International Centre for Genetic Engineering and Biotechnology, New Delhi 110 67, and
| | - Shahid Jameel
- the International Centre for Genetic Engineering and Biotechnology, New Delhi 110 67, and
| | - Amit Singh
- From the Department of Microbiology and Cell Biology, Centre for Infectious Disease and Research, Indian Institute of Sciences, Bangalore 560012,
| |
Collapse
|
34
|
Meseda CA, Srinivasan K, Wise J, Catalano J, Yamada KM, Dhawan S. Non-coding RNAs and heme oxygenase-1 in vaccinia virus infection. Biochem Biophys Res Commun 2014; 454:84-8. [PMID: 25450361 DOI: 10.1016/j.bbrc.2014.10.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 10/08/2014] [Indexed: 12/18/2022]
Abstract
Small nuclear RNAs (snRNAs) are <200 nucleotide non-coding uridylate-rich RNAs. Although the functions of many snRNAs remain undetermined, a population of snRNAs is produced during the early phase of infection of cells by vaccinia virus. In the present study, we demonstrate a direct correlation between expression of the cytoprotective enzyme heme oxygenase-1 (HO-1), suppression of selective snRNA expression, and inhibition of vaccinia virus infection of macrophages. Hemin induced HO-1 expression, completely reversed virus-induced host snRNA expression, and suppressed vaccinia virus infection. This involvement of specific virus-induced snRNAs and associated gene clusters suggests a novel HO-1-dependent host-defense pathway in poxvirus infection.
Collapse
Affiliation(s)
- Clement A Meseda
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, United States
| | - Kumar Srinivasan
- Division of Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, United States
| | | | - Jennifer Catalano
- Center for Tobacco Products, Food and Drug Administration, Bethesda, MD, United States
| | - Kenneth M Yamada
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Subhash Dhawan
- Division of Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, MD, United States.
| |
Collapse
|
35
|
Xiao S, Zhang A, Zhang C, Ni H, Gao J, Wang C, Zhao Q, Wang X, Wang X, Ma C, Liu H, Li N, Mu Y, Sun Y, Zhang G, Hiscox JA, Hsu WH, Zhou EM. Heme oxygenase-1 acts as an antiviral factor for porcine reproductive and respiratory syndrome virus infection and over-expression inhibits virus replication in vitro. Antiviral Res 2014; 110:60-9. [PMID: 25086213 DOI: 10.1016/j.antiviral.2014.07.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 07/19/2014] [Accepted: 07/22/2014] [Indexed: 02/06/2023]
Abstract
Virus replication depends upon host-cell processes in infected cells, and this is true for porcine reproductive and respiratory syndrome virus (PRRSV), the causative agent of PRRS that is a worldwide threat to the swine industry. Heme oxygenase-1 (HO-1) is a ubiquitously expressed inducible isoform of the first and rate-limiting enzyme for heme degradation. Our previous research suggested that HO-1 may play an important role in PRRSV infection. However, the function of HO-1 in PRRSV infection is unclear. In the present study, Marc-145, PK-15(CD163) cell lines and porcine alveolar macrophages (PAMs) were used to evaluate the effects of HO-1 induction and over-expression on the replication of two different PRRSV strains. Induction of HO-1 markedly decreased the replication of PRRSV strains in the different cells. Similarly, adenoviral-mediated over-expression of HO-1 also greatly decreased the replication of PRRSV. In contrast, ablation of HO-1 using small interfering RNA concomitantly increased PRRSV replication. Therefore, the data were consistent with HO-1 acting as an antiviral factor and these findings suggested that over-expression or induction of HO-1 may provide a potential therapeutic strategy against PRRSV infection.
Collapse
Affiliation(s)
- Shuqi Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Angke Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Chong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Huaibo Ni
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Jiming Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Chengbao Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Qin Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Xiangpeng Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Xue Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Chao Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Hongliang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Na Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Yang Mu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Yani Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Julian A Hiscox
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK
| | - Walter H Hsu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - En-Min Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Experimental Station of Veterinary Pharmacology and Veterinary Biotechnology, Ministry of Agriculture, China, No. 22 Xinong Road, Yangling, Shaanxi 712100, China.
| |
Collapse
|
36
|
Dimitrov JD, Planchais C, Scheel T, Ohayon D, Mesnage S, Berek C, Kaveri SV, Lacroix-Desmazes S. A cryptic polyreactive antibody recognizes distinct clades of HIV-1 glycoprotein 120 by an identical binding mechanism. J Biol Chem 2014; 289:17767-79. [PMID: 24802758 DOI: 10.1074/jbc.m114.556266] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Polyreactive antibodies play an important role for neutralization of human immunodeficiency virus (HIV). In addition to intrinsic polyreactive antibodies, the immune system of healthy individuals contains antibodies with cryptic polyreactivity. These antibodies acquire promiscuous antigen binding potential post-translationally, after exposure to various redox-active substances such as reactive oxygen species, iron ions, and heme. Here, we characterized the interaction of a prototypic human antibody that acquires binding potential to glycoprotein (gp) 120 after exposure to heme. The kinetic and thermodynamic analyses of interaction of the polyreactive antibody with distinct clades of gp120 demonstrated that the antigen-binding promiscuity of the antibody compensates for the molecular heterogeneity of the target antigen. Thus, the polyreactive antibody recognized divergent gp120 clades with similar values of the binding kinetics and quantitatively identical changes in the activation thermodynamic parameters. Moreover, this antibody utilized the same type of noncovalent forces for formation of complexes with gp120. In contrast, HIV-1-neutralizing antibodies isolated from HIV-1-infected individuals, F425 B4a1 and b12, demonstrated different binding behavior upon interaction with distinct variants of gp120. This study contributes to a better understanding of the physiological role and binding mechanism of antibodies with cryptic polyreactivity. Moreover, this study might be of relevance for understanding the basic aspects of HIV-1 interaction with human antibodies.
Collapse
Affiliation(s)
- Jordan D Dimitrov
- From the Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Unité Mixte de Recherche S 1138, 75006 Paris, France, the Université Paris Descartes, Unité Mixte de Recherche S 1138, Paris, France, INSERM U1138, 75006 Paris, France,
| | - Cyril Planchais
- From the Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Unité Mixte de Recherche S 1138, 75006 Paris, France, the Université Paris Descartes, Unité Mixte de Recherche S 1138, Paris, France, INSERM U1138, 75006 Paris, France
| | - Tobias Scheel
- the Deutsches Rheuma-Forschungszentrum, Institut der Leibniz-Gemeinschaft, 13092 Berlin, Germany, and
| | - Delphine Ohayon
- From the Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Unité Mixte de Recherche S 1138, 75006 Paris, France, the Université Paris Descartes, Unité Mixte de Recherche S 1138, Paris, France, INSERM U1138, 75006 Paris, France
| | - Stephane Mesnage
- the Krebs Institute, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, United Kingdom
| | - Claudia Berek
- the Deutsches Rheuma-Forschungszentrum, Institut der Leibniz-Gemeinschaft, 13092 Berlin, Germany, and
| | - Srinivas V Kaveri
- From the Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Unité Mixte de Recherche S 1138, 75006 Paris, France, the Université Paris Descartes, Unité Mixte de Recherche S 1138, Paris, France, INSERM U1138, 75006 Paris, France
| | - Sébastien Lacroix-Desmazes
- From the Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Unité Mixte de Recherche S 1138, 75006 Paris, France, the Université Paris Descartes, Unité Mixte de Recherche S 1138, Paris, France, INSERM U1138, 75006 Paris, France
| |
Collapse
|
37
|
Abstract
Reactive oxygen species (ROS) are deadly weapons used by phagocytes and other cell types, such as lung epithelial cells, against pathogens. ROS can kill pathogens directly by causing oxidative damage to biocompounds or indirectly by stimulating pathogen elimination by various nonoxidative mechanisms, including pattern recognition receptors signaling, autophagy, neutrophil extracellular trap formation, and T-lymphocyte responses. Thus, one should expect that the inhibition of ROS production promote infection. Increasing evidences support that in certain particular infections, antioxidants decrease and prooxidants increase pathogen burden. In this study, we review the classic infections that are controlled by ROS and the cases in which ROS appear as promoters of infection, challenging the paradigm. We discuss the possible mechanisms by which ROS could promote particular infections. These mechanisms are still not completely clear but include the metabolic effects of ROS on pathogen physiology, ROS-induced damage to the immune system, and ROS-induced activation of immune defense mechanisms that are subsequently hijacked by particular pathogens to act against more effective microbicidal mechanisms of the immune system. The effective use of antioxidants as therapeutic agents against certain infections is a realistic possibility that is beginning to be applied against viruses.
Collapse
Affiliation(s)
- Claudia N Paiva
- Departamento de Imunologia, Instituto de Microbiologia , CCS Bloco D, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | | |
Collapse
|
38
|
Zhu Z, Mathahs MM, Schmidt WN. Restoration of type I interferon expression by heme and related tetrapyrroles through inhibition of NS3/4A protease. J Infect Dis 2013; 208:1653-63. [PMID: 23901085 DOI: 10.1093/infdis/jit338] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Tetrapyrrole substrates and products of heme oxygenase are potent inhibitors of hepatitis C virus (HCV) replication. It is not clear whether this occurs through primary induction of type I interferon (IFN), inhibition of viral NS3/4A protease, or a combination of these mechanisms. We studied the antiviral actions of tetrapyrroles and their potential influence on type I IFN induction. METHODS The effects of tetrapyrrole on NS3/4A protease activity and type I IFN induction were assessed in HCV-permissive cells, replicons, or human embryonic kidney (HEK) 293 cells transfected with NS3/4A protease. Activation of innate immune signaling was determined after transfection of double-strand surrogate nucleic acid antigens or infection with defined sequence HCV cell culture (HCVcc) RNA. RESULTS Tetrapyrroles failed to directly induce IFN expression at concentrations that inhibited HCV replication and NS3/4A protease activity. However, they potently restored IFN induction after attenuation with NS3/4A protease, a process accompanied by preservation of the adapter protein, mitochondrial antiviral signaling protein, nuclear localization of IFN regulatory factor 3, and augmentation of IFN-stimulated gene products. CONCLUSIONS Tetrapyrroles do not directly induce IFN, but they dramatically restore type I IFN signaling pathway after attenuation with NS3/4A protease. They show immunomodulatory as well as antiprotease activity and may be useful for treatment of HCV infection.
Collapse
Affiliation(s)
- Zhaowen Zhu
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center
| | | | | |
Collapse
|
39
|
Kapitulnik J, Maines MD. The role of bile pigments in health and disease: effects on cell signaling, cytotoxicity, and cytoprotection. Front Pharmacol 2012; 3:136. [PMID: 22811668 PMCID: PMC3395811 DOI: 10.3389/fphar.2012.00136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 06/25/2012] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jaime Kapitulnik
- Laboratory of Drug Metabolism, Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem, Israel
| | | |
Collapse
|