1
|
Mocarski ES. Programmed Necrosis in Host Defense. Curr Top Microbiol Immunol 2023; 442:1-40. [PMID: 37563336 DOI: 10.1007/82_2023_264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Host control over infectious disease relies on the ability of cells in multicellular organisms to detect and defend against pathogens to prevent disease. Evolution affords mammals with a wide variety of independent immune mechanisms to control or eliminate invading infectious agents. Many pathogens acquire functions to deflect these immune mechanisms and promote infection. Following successful invasion of a host, cell autonomous signaling pathways drive the production of inflammatory cytokines, deployment of restriction factors and induction of cell death. Combined, these innate immune mechanisms attract dendritic cells, neutrophils and macrophages as well as innate lymphoid cells such as natural killer cells that all help control infection. Eventually, the development of adaptive pathogen-specific immunity clears infection and provides immune memory of the encounter. For obligate intracellular pathogens such as viruses, diverse cell death pathways make a pivotal contribution to early control by eliminating host cells before progeny are produced. Pro-apoptotic caspase-8 activity (along with caspase-10 in humans) executes extrinsic apoptosis, a nonlytic form of cell death triggered by TNF family death receptors (DRs). Over the past two decades, alternate extrinsic apoptosis and necroptosis outcomes have been described. Programmed necrosis, or necroptosis, occurs when receptor interacting protein kinase 3 (RIPK3) activates mixed lineage kinase-like (MLKL), causing cell leakage. Thus, activation of DRs, toll-like receptors (TLRs) or pathogen sensor Z-nucleic acid binding protein 1 (ZBP1) initiates apoptosis as well as necroptosis if not blocked by virus-encoded inhibitors. Mammalian cell death pathways are blocked by herpesvirus- and poxvirus-encoded cell death suppressors. Growing evidence has revealed the importance of Z-nucleic acid sensor, ZBP1, in the cell autonomous recognition of both DNA and RNA virus infection. This volume will explore the detente between viruses and cells to manage death machinery and avoid elimination to support dissemination within the host animal.
Collapse
Affiliation(s)
- Edward S Mocarski
- Robert W. Woodruff Professor Emeritus, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Professor Emeritus, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
2
|
Exploring the multifaceted roles of heat shock protein B8 (HSPB8) in diseases. Eur J Cell Biol 2018; 97:216-229. [PMID: 29555102 DOI: 10.1016/j.ejcb.2018.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/09/2018] [Accepted: 03/12/2018] [Indexed: 02/06/2023] Open
Abstract
HSPB8 is a member of ubiquitous small heat shock protein (sHSP) family, whose expression is induced in response to a wide variety of unfavorable physiological and environmental conditions. Investigation of HSPB8 structure indicated that HSPB8 belongs to the group of so-called intrinsically disordered proteins and possesses a highly flexible structure. Unlike most other sHSPs, HSPB8 tends to form small-molecular-mass oligomers and exhibits substrate-dependent chaperone activity. In cooperation with BAG3, the chaperone activity of HSPB8 was reported to be involved in the delivery of misfolded proteins to the autophagy machinery. Through this way, HSPB8 interferes with pathological processes leading to neurodegenerative diseases. Accordingly, published studies have identified genetic links between mutations of HSPB8 and some kind of neuromuscular diseases, further supporting its important role in neurodegenerative disorders. In addition to their anti-aggregation properties, HSPB8 is indicated to interact with a wide range of client proteins, modulating their maturations and activities, and therefore, regulates a large repertoire of cellular functions, including apoptosis, proliferation, inflammation and etc. As a result, HSPB8 has key roles in cancer biology, autoimmune diseases, cardiac diseases and cerebral vascular diseases.
Collapse
|
3
|
Fountzilas C, Patel S, Mahalingam D. Review: Oncolytic virotherapy, updates and future directions. Oncotarget 2017; 8:102617-102639. [PMID: 29254276 PMCID: PMC5731986 DOI: 10.18632/oncotarget.18309] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022] Open
Abstract
Oncolytic viruses (OVs) are viral strains that can infect and kill malignant cells while spare their normal counterparts. OVs can access cells through binding to receptors on their surface or through fusion with the plasma membrane and establish a lytic cycle in tumors, while leaving normal tissue essentially unharmed. Multiple viruses have been investigated in humans for the past century. IMLYGIC™ (T-VEC/Talimogene Laherparepvec), a genetically engineered Herpes Simplex Virus, is the first OV approved for use in the United States and the European Union for patients with locally advanced or non-resectable melanoma. Although OVs have a favorable toxicity profile and are impressively active anticancer agents in vitro and in vivo the majority of OVs have limited clinical efficacy as a single agent. While a virus-induced antitumor immune response can enhance oncolysis, when OVs are used systemically, the antiviral immune response can prevent the virus reaching the tumor tissue and having a therapeutic effect. Intratumoral administration can provide direct access to tumor tissue and be beneficial in reducing side effects. Immune checkpoint stimulation in tumor tissue has been noted after OV therapy and can be a natural response to viral-induced oncolysis. Also for immune checkpoint inhibition to be effective in treating cancer, an immune response to tumor neoantigens and an inflamed tumor microenvironment are required, both of which treatment with an OV may provide. Therefore, direct and indirect mechanisms of tumor killing provide rationale for clinical trials investigating the combination of OVs other forms of cancer therapy, including immune checkpoint inhibition.
Collapse
Affiliation(s)
- Christos Fountzilas
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sukeshi Patel
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | |
Collapse
|
4
|
H11/HspB8 and Its Herpes Simplex Virus Type 2 Homologue ICP10PK Share Functions That Regulate Cell Life/Death Decisions and Human Disease. Autoimmune Dis 2012; 2012:395329. [PMID: 23056924 PMCID: PMC3463903 DOI: 10.1155/2012/395329] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 12/24/2022] Open
Abstract
Small heat shock proteins (sHsp) also known as HspB are a large family of widely expressed proteins that contain a 90 residues domain known as α-crystallin. Here, we focus on the family member H11/HspB8 and its herpes simplex virus type 2 (HSV-2) homologue ICP10PK, and discuss the possible impact of this relationship on human disease. H11/HspB8 and ICP10PK are atypical protein kinases. They share multi-functional activity that encompasses signaling, unfolded protein response (UPR) and the regulation of life cycle potential. In melanocytes H11/HspB8 causes growth arrest. It is silenced in a high proportion of melanoma prostate cancer, Ewing's sarcoma and hematologic malignancies through aberrant DNA methylation. Its restored expression induces cell death and inhibits tumor growth in xenograft models, identifying H11/HspB8 as a tumor suppressor. This function involves the activation of multiple and distinct death pathways, all of which initiate with H11/HspB8-mediated phosphorylation of transforming growth factor β-activated kinase 1 (TAK1). Both ICP10PK and H11/HspB8 were implicated in inflammatory processes that involve dendritic cells activation through Toll-like receptor-dependent pathways and may contribute to the onset of autoimmunity. The potential evolutionary relationship of H11/HspB8 to ICP10PK, its impact on human disorders and the development of therapeutic strategies are discussed.
Collapse
|
5
|
Oncolytic viruses in the treatment of bladder cancer. Adv Urol 2012; 2012:404581. [PMID: 22899907 PMCID: PMC3414001 DOI: 10.1155/2012/404581] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/05/2012] [Indexed: 01/22/2023] Open
Abstract
Bladder carcinoma is the second most common malignancy of the urinary tract. Up to 85% of patients with bladder cancer are diagnosed with a tumor that is limited to the bladder mucosa (Ta, T1, and CIS). These stages are commonly termed as non-muscle-invasive bladder cancer (NMIBC). Although the treatment of NMIBC has greatly improved in recent years, there is a need for additional therapies when patients fail bacillus Calmette-Guérin (BCG) and chemotherapeutic agents. We propose that bladder cancer may be an ideal target for oncolytic viruses engineered to selectively replicate in and lyse tumor cells leaving normal cells unharmed. In support of this hypothesis, here we review current treatment strategies for bladder cancer and their shortcomings, as well as recent advancements in oncolytic viral therapy demonstrating encouraging safety profiles and antitumor activity.
Collapse
|
6
|
Mymrikov EV, Seit-Nebi AS, Gusev NB. Large potentials of small heat shock proteins. Physiol Rev 2011; 91:1123-59. [PMID: 22013208 DOI: 10.1152/physrev.00023.2010] [Citation(s) in RCA: 316] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Modern classification of the family of human small heat shock proteins (the so-called HSPB) is presented, and the structure and properties of three members of this family are analyzed in detail. Ubiquitously expressed HSPB1 (HSP27) is involved in the control of protein folding and, when mutated, plays a significant role in the development of certain neurodegenerative disorders. HSPB1 directly or indirectly participates in the regulation of apoptosis, protects the cell against oxidative stress, and is involved in the regulation of the cytoskeleton. HSPB6 (HSP20) also possesses chaperone-like activity, is involved in regulation of smooth muscle contraction, has pronounced cardioprotective activity, and seems to participate in insulin-dependent regulation of muscle metabolism. HSPB8 (HSP22) prevents accumulation of aggregated proteins in the cell and participates in the regulation of proteolysis of unfolded proteins. HSPB8 also seems to be directly or indirectly involved in regulation of apoptosis and carcinogenesis, contributes to cardiac cell hypertrophy and survival and, when mutated, might be involved in development of neurodegenerative diseases. All small heat shock proteins play important "housekeeping" roles and regulate many vital processes; therefore, they are considered as attractive therapeutic targets.
Collapse
Affiliation(s)
- Evgeny V Mymrikov
- Department of Biochemistry, School of Biology, Moscow State University, Moscow, Russian Federation
| | | | | |
Collapse
|
7
|
The ribonucleotide reductase R1 subunits of herpes simplex virus types 1 and 2 protect cells against TNFα- and FasL-induced apoptosis by interacting with caspase-8. Apoptosis 2011; 16:256-71. [PMID: 21107701 DOI: 10.1007/s10495-010-0560-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We previously reported that HSV-2 R1, the R1 subunit (ICP10; UL39) of herpes simplex virus type-2 ribonucleotide reductase, protects cells against apoptosis induced by the death receptor (DR) ligands tumor necrosis factor-alpha- (TNFα) and Fas ligand (FasL) by interrupting DR-mediated signaling at, or upstream of, caspase-8 activation. Further investigation of the molecular mechanism underlying HSV-2 R1 protection showed that extracellular-regulated kinase 1/2 (ERK1/2), phosphatidylinositol 3-kinase (PI3-K)/Akt, NF-κB and JNK survival pathways do not play a major role in this antiapoptotic function. Interaction studies revealed that HSV-2 R1 interacted constitutively with caspase-8. The HSV-2 R1 deletion mutant R1(1-834)-GFP and Epstein-Barr virus (EBV) R1, which did not protect against apoptosis induced by DR ligands, did not interact with caspase-8, indicating that interaction is required for protection. HSV-2 R1 impaired caspase-8 activation induced by caspase-8 over-expression, suggesting that interaction between the two proteins prevents caspase-8 dimerization/activation. HSV-2 R1 bound to caspase-8 directly through its prodomain but did not interact with either its caspase domain or Fas-associated death domain protein (FADD). Interaction between HSV-2 R1 and caspase-8 disrupted FADD-caspase-8 binding. We further demonstrated that individually expressed HSV-1 R1 (ICP6) shares, with HSV-2 R1, the ability to bind caspase-8 and to protect cells against DR-induced apoptosis. Finally, as the long-lived Fas protein remained stable during the early period of infection, experiments with the HSV-1 UL39 deletion mutant ICP6∆ showed that HSV-1 R1 could be essential for the protection of HSV-1-infected cells against FasL.
Collapse
|
8
|
Fu X, Tao L, Zhang X. A short polypeptide from the herpes simplex virus type 2 ICP10 gene can induce antigen aggregation and autophagosomal degradation for enhanced immune presentation. Hum Gene Ther 2011; 21:1687-96. [PMID: 20583863 DOI: 10.1089/hum.2010.080] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
It has been reported that certain polypeptides derived from aggregation-prone cellular proteins can turn soluble green fluorescent protein (GFP) into aggregates. Here we report our finding that a short peptide derived from a viral gene, ICP10 of herpes simplex virus (HSV)-2, also possesses such a property. A sequence as short as 13 amino acids from the middle region of the gene can convert GFP into an aggregation-prone toxic protein once it is fused to the C terminus. Moreover, this short peptide can direct a surrogate tumor antigen into the autophagosome/lysosome degradation pathway, drastically increasing both MHC class I and class II antigen presentation. The simultaneous induction of both arms of the T cell immune response to the tumor antigen effectively protects the immunized animals from tumor challenge. Designated VIPA (i.e., viral inducer of protein aggregation), this unique viral sequence may represent an attractive candidate as a molecular adjuvant for cancer immunotherapy and for other immunologically preventable diseases.
Collapse
Affiliation(s)
- Xinping Fu
- Department of Biology and Biochemistry, University of Houston, TX 77204, USA
| | | | | |
Collapse
|
9
|
Abstract
Phosphorylation represents one the most abundant and important posttranslational modifications of proteins, including viral proteins. Virus-encoded serine/threonine protein kinases appear to be a feature that is unique to large DNA viruses. Although the importance of these kinases for virus replication in cell culture is variable, they invariably play important roles in virus virulence. The current review provides an overview of the different viral serine/threonine protein kinases of several large DNA viruses and discusses their function, importance, and potential as antiviral drug targets.
Collapse
|
10
|
Lembo D, Brune W. Tinkering with a viral ribonucleotide reductase. Trends Biochem Sci 2008; 34:25-32. [PMID: 18990579 DOI: 10.1016/j.tibs.2008.09.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 09/16/2008] [Accepted: 09/16/2008] [Indexed: 10/21/2022]
Abstract
Ribonucleotide reductase (RNR), a crucial enzyme for nucleotide anabolism, is encoded by all living organisms and by large DNA viruses such as the herpesviruses. Surprisingly, the beta-herpesvirus subfamily RNR R1 subunit homologues are catalytically inactive and their function remained enigmatic for many years. Recent work sheds light on the function of M45, the murine cytomegalovirus R1 homologue; during viral evolution, M45 apparently lost its original RNR activity but gained the ability, via inhibiting RIP1, a cellular adaptor protein, to block cellular signaling pathways involved in innate immunity and inflammation. The discovery of this novel mechanism of viral immune subversion provides further support to the concept of evolutionary tinkering.
Collapse
Affiliation(s)
- David Lembo
- Department of Clinical and Biological Sciences, University of Turin, S Luigi Gonzaga Hospital, Orbassano, Turin, Italy.
| | | |
Collapse
|
11
|
Buchkovich NJ, Yu Y, Zampieri CA, Alwine JC. The TORrid affairs of viruses: effects of mammalian DNA viruses on the PI3K-Akt-mTOR signalling pathway. Nat Rev Microbiol 2008; 6:266-75. [PMID: 18311165 DOI: 10.1038/nrmicro1855] [Citation(s) in RCA: 194] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The successful replication of mammalian DNA viruses requires that they gain control of key cellular signalling pathways that affect broad aspects of cellular macromolecular synthesis, metabolism, growth and survival. The phosphatidylinositol 3'-kinase-Akt-mammalian target of rapamycin (PI3K-Akt-mTOR) pathway is one such pathway. Mammalian DNA viruses have evolved various mechanisms to activate this pathway to obtain the benefits of Akt activation, including the maintenance of translation through the activation of mTOR. In addition, viruses must overcome the inhibition of this pathway that results from the activation of cellular stress responses during viral infection. This Review will discuss the range of mechanisms that mammalian DNA viruses use to activate this pathway, as well as the multiple mechanisms these viruses have evolved to circumvent inhibitory stress signalling.
Collapse
Affiliation(s)
- Nicholas J Buchkovich
- Department of Cancer Biology and Abramson Family Cancer Research Institute, University of Pennsylvania, 314 Biomedical Research Building, 421 Curie Blvd, Philadelphia, 19104-6142 Pennsylvania, USA
| | | | | | | |
Collapse
|
12
|
Shemetov AA, Seit-Nebi AS, Gusev NB. Structure, properties, and functions of the human small heat-shock protein HSP22 (HspB8, H11, E2IG1): A critical review. J Neurosci Res 2008; 86:264-9. [PMID: 17722063 DOI: 10.1002/jnr.21441] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The recently described human HSP22 belongs to the superfamily of small heat-shock proteins containing a conservative alpha-crystallin domain. HSP22 seems to be involved in regulation of cell proliferation, cardiac hypertrophy, apoptosis, and carcinogenesis, and expression of point mutants of HSP22 correlates with development of different neuromuscular diseases. Therefore, an investigation of the structure and properties of HSP22 is desirable for understanding its multiple functions. HSP22 seems to belong to the group of so-called intrinsically disordered proteins and possesses a highly flexible structure. HSP22 tends to form small-molecular-mass oligomers and interacts with biological membranes and many different proteins, among them glycolytic enzymes and different protein kinases. HSP22 possesses chaperonelike activity and prevents aggregation of denatured proteins both in vitro and in vivo. Depending on the cell type and its expression, HSP22 might have either pro- or anti-apoptotic effects. Chaperonelike activity seems to be important for antiapoptotic effects, whereas interaction with and regulation of certain protein kinases might be important for the proapoptotic effects of HSP22. Expression of K141N or K141E mutants of HSP22 correlates with development of distal hereditary motor neuropathy and/or Charcot-Marie-Tooth disease. These mutations destabilize the structure of HSP22, affect its interaction with other small heat-shock proteins, and decrease its chaperonelike activity. HSP22 decreases or prevents aggregation of Huntingtin fragments and amyloid-beta peptide 1-40 of the Dutch type. Thus, HSP22 seems to play an important role in the nervous system, and further investigations are needed to understand the molecular mechanisms of its functioning.
Collapse
Affiliation(s)
- Anton A Shemetov
- Department of Biochemistry, School of Biology, Moscow State University, Moscow, Russia
| | | | | |
Collapse
|
13
|
Chabaud S, Sasseville AMJ, Elahi SM, Caron A, Dufour F, Massie B, Langelier Y. The ribonucleotide reductase domain of the R1 subunit of herpes simplex virus type 2 ribonucleotide reductase is essential for R1 antiapoptotic function. J Gen Virol 2007; 88:384-394. [PMID: 17251554 DOI: 10.1099/vir.0.82383-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The R1 subunit (ICP10) of herpes simplex virus type 2 (HSV-2) ribonucleotide reductase (RR), which in addition to its C-terminal reductase domain possesses a unique N-terminal domain of about 400 aa, protects cells against apoptosis. As the NH2domain on its own is not antiapoptotic, it has been postulated that both domains of R1 or part(s) of them could be necessary for this function. Here, N- and C-terminal deletions were introduced in HSV-2 R1 to map the domain(s) involved in its antiapoptotic potential. The results showed that, whereas most of the NH2domain including part of the recently described putativeα-crystallin domain is dispensable for antiapoptotic activity, it is the integrity of the structured RR domain that is required for protection. As theα-crystallin domain appears to play an important role in protein folding and oligomerization, the N-terminal boundary of the antiapoptotic domain could not be defined precisely. In addition, this study provided evidence that overexpression of HSV-2 R2 at levels up to 30-fold more than HSV-2 R1 did not decrease protection from tumour necrosis factor alpha, indicating that the R1 surface where R2 binds is not involved in antiapoptotic activity. Importantly, this result suggests that the co-expression of both RR subunits during the lytic cycle should not affect protection from this cytokine.
Collapse
Affiliation(s)
- Stéphane Chabaud
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, QC H2L 4M1, Canada
| | - A Marie-Josée Sasseville
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, QC H2L 4M1, Canada
| | - Seyyed Mehdy Elahi
- Institut de Recherche en Biotechnologie, 6100 ave Royalmount, Montréal, QC H4P 2R2, Canada
| | - Antoine Caron
- Institut de Recherche en Biotechnologie, 6100 ave Royalmount, Montréal, QC H4P 2R2, Canada
| | - Florent Dufour
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, QC H2L 4M1, Canada
| | - Bernard Massie
- INRS-IAF, Université du Québec, Laval, QC H7N 4Z3, Canada
- Département de Microbiologie et Immunologie, Université de Montréal, QC, Canada
- Institut de Recherche en Biotechnologie, 6100 ave Royalmount, Montréal, QC H4P 2R2, Canada
| | - Yves Langelier
- Département de Médecine, Université de Montréal, QC, Canada
- Département de Microbiologie et Immunologie, Université de Montréal, QC, Canada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, QC H2L 4M1, Canada
| |
Collapse
|
14
|
Brandt CR. The role of viral and host genes in corneal infection with herpes simplex virus type 1. Exp Eye Res 2005; 80:607-21. [PMID: 15862167 DOI: 10.1016/j.exer.2004.09.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Accepted: 09/28/2004] [Indexed: 11/26/2022]
Abstract
Herpes simplex virus infection of the eye is the leading cause of blindness due to infection in the US despite the availability of several antiviral drugs. Studies with animal models have shown that three factors, innate host resistance, the host adaptive immune response, and the strain of virus interact to determine whether an infection is asymptomatic or proceeds to the development of blinding keratitis (HSK). Of these, the role of adaptive immunity has received the most attention. This work has clearly shown that stromal keratitis is an immunopathological disease, most likely due to the induction of a delayed type hypersensitivity response. Substantially less is known about the role of specific host genes in resistance to HSK. The fact that different strains of virus display different disease phenotypes indicates that viral 'virulence' genes are critical. Of the 80 plus HSV genes, few have been formally tested for their role in HSV keratitis. Most studies of virulence genes to date have focused on a single gene or protein and large changes in disease phenotypes are usually measured. Large changes in the ability to cause disease are likely to reduce the fitness of the virus, thus such studies, although useful, do not mimic the natural situation. Viral gene products are known to interact with each other, and with host proteins and these interactions are critical in determining the outcome of infection. In reality, the 'constellation' of genes encoded by each particular strain is critical, and how this constellation of genes works together and with host proteins determines the outcome of an infection. The goal of this review is to discuss the current state of knowledge regarding the role of host and viral genes in HSV keratitis. The roles of specific genes that have been shown to influence keratitis are discussed. Recent data showing that different viral genes cooperate to influence disease severity and confirming that the constellation of genes within a particular strain determines the disease phenotype are also discussed, as are the methods used to test the role of viral genes in virulence. It will become apparent that there is a paucity of information regarding the function of many viral genes in keratitis. Improving our knowledge of the role of viral genes is critical for devising more effective treatments for this disease.
Collapse
Affiliation(s)
- Curtis R Brandt
- Department of Ophthalmology and Visual Sciences, University of Wisconsin Medical School, 6630 MSC, 1300 University Avenue, Madison, WI 53706, USA.
| |
Collapse
|
15
|
Kim MV, Seit-Nebi AS, Gusev NB. The problem of protein kinase activity of small heat shock protein Hsp22 (H11 or HspB8). Biochem Biophys Res Commun 2004; 325:649-52. [PMID: 15541337 DOI: 10.1016/j.bbrc.2004.10.074] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2004] [Indexed: 11/23/2022]
Abstract
The recently described protein denoted H11, Hsp22 or HspB8 seems to participate in regulation of proliferation, apoptosis, and cardiac hypertrophy. Mutation of Hsp22 causes distal motor neuropathy. Multitude action of Hsp22 is supposed to be due to its protein kinase and/or chaperone-like activities. There are many indirect evidences indicating that Hsp22 possesses intrinsic protein kinase activity. However, low homology to protein kinases, low extent of autophosphorylation, lack of significant protein kinase activity with commonly used substrates, and lack of information on stoichiometry, kinetics, and substrate specificity make the existence of intrinsic protein kinase activity of Hsp22 questionable. It is supposed that protein kinase activity ascribed to Hsp22 is due to contaminating protein kinases. Hsp22 is highly homologous to small heat shock proteins and effectively prevents aggregation of denatured protein both in vitro and in vivo. Therefore, it is supposed that chaperone-like activity is of great importance for Hsp22 functioning.
Collapse
Affiliation(s)
- Maria V Kim
- Department of Biochemistry, School of Biology, Moscow State University, Moscow 119992, Russia
| | | | | |
Collapse
|
16
|
Patrone M, Percivalle E, Secchi M, Fiorina L, Pedrali-Noy G, Zoppé M, Baldanti F, Hahn G, Koszinowski UH, Milanesi G, Gallina A. The human cytomegalovirus UL45 gene product is a late, virion-associated protein and influences virus growth at low multiplicities of infection. J Gen Virol 2003; 84:3359-3370. [PMID: 14645917 DOI: 10.1099/vir.0.19452-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human cytomegalovirus (HCMV) encodes a protein related to the large (R1) subunit of ribonucleotide reductase (RR), but does not encode the corresponding small (R2) subunit. The R1 homologue, UL45, lacks many catalytic residues, and its impact on deoxyribonucleotide (dNTP) production remains unknown. Here, UL45 is shown to accumulate at late stages of infection and to be a virion tegument protein. To study UL45 function in its genome context, UL45 was disrupted by transposon insertion. The UL45-knockout (UL45-KO) mutant exhibited a growth defect in fibroblasts at a low m.o.i. and also a cell-to-cell spread defect. This did not result from a reduced dNTP supply because dNTP pools were unchanged in resting cells infected with the mutant virus. Irrespective of UL45 expression, all cellular RR subunits - S-phase RR subunits, and the p53-dependent p53R2 - were induced by infection. p53R2 was targeted to the infected cell nucleus, suggesting that HCMV diverts a mechanism normally activated by DNA damage response. Cells infected with the UL45-KO mutant were moderately sensitized to Fas-induced apoptosis relative to those infected with the parental virus. Together with the report on the UL45-KO endotheliotropic HCMV mutant (Hahn et al., J Virol 76, 9551-9555, 2002), these data suggest that UL45 does not share the prominent antiapototic role attributed to the mouse cytomegalovirus homologue M45 (Brune et al., Science 291, 303-305, 2001).
Collapse
Affiliation(s)
- Marco Patrone
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Elena Percivalle
- Servizio di Virologia, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Massimiliano Secchi
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Loretta Fiorina
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Guido Pedrali-Noy
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Monica Zoppé
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Fausto Baldanti
- Servizio di Virologia, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gabriele Hahn
- Max von Pettenkofer-Institut, Lehrstuhl für Virologie, LMU-München, München, Germany
| | - Ulrich H Koszinowski
- Max von Pettenkofer-Institut, Lehrstuhl für Virologie, LMU-München, München, Germany
| | - Gabriele Milanesi
- Department of Medicine and Surgery, San Paolo Hospital, University of Milano, via A. di Rudinı ` 8, I-20142 Milano, Italy
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Andrea Gallina
- Department of Medicine and Surgery, San Paolo Hospital, University of Milano, via A. di Rudinı ` 8, I-20142 Milano, Italy
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, I-27100 Pavia, Italy
| |
Collapse
|
17
|
Chabaud S, Lambert H, Sasseville AMJ, Lavoie H, Guilbault C, Massie B, Landry J, Langelier Y. The R1 subunit of herpes simplex virus ribonucleotide reductase has chaperone-like activity similar to Hsp27. FEBS Lett 2003; 545:213-8. [PMID: 12804778 DOI: 10.1016/s0014-5793(03)00547-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
HSV-2 R1, the R1 subunit of herpes simplex virus (HSV) ribonucleotide reductase, protects cells against apoptosis. Here, we report the presence in HSV-2 R1 of a stretch exhibiting similarity to the alpha-crystallin domain of the small heat shock proteins, a domain known to be important for oligomerization and cytoprotective activities of these proteins. Also, the HSV-2 R1 protein, which forms multimeric structures in the absence of nucleotide, displayed chaperone ability as good as Hsp27 in a thermal denaturation assay using citrate synthase. In contrast, mammalian R1, which does not contain an alpha-crystallin domain, has neither chaperone nor anti-apoptotic activity. Thus, we propose that the chaperone activity of HSV-2 R1 could play an important role in viral pathogenesis.
Collapse
Affiliation(s)
- Stéphane Chabaud
- Centre de recherche, Centre hospitalier, Université de Montréal, Montreal, QC, Canada H2L 4M1
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Langelier Y, Bergeron S, Chabaud S, Lippens J, Guilbault C, Sasseville AMJ, Denis S, Mosser DD, Massie B. The R1 subunit of herpes simplex virus ribonucleotide reductase protects cells against apoptosis at, or upstream of, caspase-8 activation. J Gen Virol 2002; 83:2779-2789. [PMID: 12388814 DOI: 10.1099/0022-1317-83-11-2779] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The R1 subunit of herpes simplex virus (HSV) ribonucleotide reductase, which in addition to its C-terminal reductase domain possesses a unique N-terminal domain of about 400 amino acids, is thought to have an additional, as yet unknown, function. Here, we report that the full-length HSV-2 R1 has an anti-apoptotic function able to protect cells against death triggered by expression of R1(Delta2-357), an HSV-2 R1 subunit with its first 357 amino acids deleted. We further substantiate the R1 anti-apoptotic activity by showing that its accumulation at low level could completely block apoptosis induced by TNF-receptor family triggering. Activation of caspase-8 induced either by TNF or by Fas ligand expression was prevented by the R1 protein. As HSV R1 did not inhibit cell death mediated by several agents acting via the mitochondrial pathway (Bax overexpression, etoposide, staurosporine and menadione), it is proposed that it functions to interrupt specifically death receptor-mediated signalling at, or upstream of, caspase-8 activation. The N-terminal domain on its own did not exhibit anti-apoptotic activity, suggesting that both domains of R1 or part(s) of them are necessary for this new function. Evidence for the importance of HSV R1 in protecting HSV-infected cells against cytokine-induced apoptosis was obtained with the HSV-1 R1 deletion mutants ICP6Delta and hrR3. These results show that, in addition to its ribonucleotide reductase function, which is essential for virus reactivation, HSV R1 could contribute to virus propagation by preventing apoptosis induced by the immune system.
Collapse
Affiliation(s)
- Yves Langelier
- Département de microbiologie et immunologie de l'Université de Montréal, Montréal, Québec, Canada2
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, Québec, CanadaH2L 4M11
| | - Stéphane Bergeron
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, Québec, CanadaH2L 4M11
| | - Stéphane Chabaud
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, Québec, CanadaH2L 4M11
| | - Julie Lippens
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, Québec, CanadaH2L 4M11
| | - Claire Guilbault
- Institut de recherche en biotechnologie, 6100 ave Royalmount, Montréal, CanadaH4P 2R23
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, Québec, CanadaH2L 4M11
| | - A Marie-Josée Sasseville
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, Québec, CanadaH2L 4M11
| | - Stéphan Denis
- Centre de recherche du Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montréal, Québec, CanadaH2L 4M11
| | - Dick D Mosser
- Institut de recherche en biotechnologie, 6100 ave Royalmount, Montréal, CanadaH4P 2R23
| | - Bernard Massie
- INRS-IAF Université du Québec, Laval, Québec, CanadaH7N 4Z34
- Institut de recherche en biotechnologie, 6100 ave Royalmount, Montréal, CanadaH4P 2R23
- Département de microbiologie et immunologie de l'Université de Montréal, Montréal, Québec, Canada2
| |
Collapse
|
19
|
Shibaki T, Suzutani T, Yoshida I, Ogasawara M, Azuma M. Participation of type I interferon in the decreased virulence of the UL13 gene-deleted mutant of herpes simplex virus type 1. J Interferon Cytokine Res 2001; 21:279-85. [PMID: 11429158 DOI: 10.1089/107999001300177466] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We isolated a UL13 gene-deleted mutant of the herpes simplex virus type 1 (HSV-1) strain VR3 (VRDelta13) and its revertant virus (VRDelta13R). This deletion mutant still had virus host shutoff (vhs) activity, although a previous report had suggested the possibility of a functional relation between the UL13 product, that is protein kinase (PK), and vhs activity. We compared the in vivo growth of these viruses in BALB/c mice. VRDelta13 was cleared in the early period of intraperitoneal infection. VRDelta13 had a higher sensitivity to the mouse type I interferon (IFN) and showed a higher level of IFN induction in the study period of infection than did VR3 and VRDelta13R. These results suggest that a nonspecific antiviral response (i.e., the IFN system) may contribute to this rapid inhibition of viral replication in vivo.
Collapse
Affiliation(s)
- T Shibaki
- Department of Microbiology and First Department of Surgery, Asahikawa Medical College, Asahikawa, 078-8510, Japan
| | | | | | | | | |
Collapse
|
20
|
Smith CC, Yu YX, Kulka M, Aurelian L. A novel human gene similar to the protein kinase (PK) coding domain of the large subunit of herpes simplex virus type 2 ribonucleotide reductase (ICP10) codes for a serine-threonine PK and is expressed in melanoma cells. J Biol Chem 2000; 275:25690-9. [PMID: 10833516 DOI: 10.1074/jbc.m002140200] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The large subunit of herpes simplex virus type 2 ribonucleotide reductase (ICP10) is a multifunctional protein that contains a serine-threonine protein kinase (PK) activity (Nelson, J. W., Zhu, J. , Smith, C. C., Kulka, M., and Aurelian, L. (1996) J. Biol. Chem. 271, 17021-17027). Phylogenetic analyses indicated that ICP10 PK belongs to a distinct subfamily of growth factor receptor serine-threonine PKs that are characterized by their ability to function with a limited number of conserved catalytic motifs (Hunter, J. C. R., Smith, C. C., and Aurelian, L. (1995) Int. J. Onc. 7, 515-522). Here, we report the isolation and characterization of a novel gene, designated H11, that contains an open reading frame of 588 nucleotides, which encodes a protein similar to ICP10 PK. The H11 protein has Mn(2+)-dependent serine-threonine-specific PK activity as determined with a GST-H11 fusion protein and by immununocomplex PK/immunoblotting assays of 293 cells transfected with a H11 eukaryotic expression vector. PK activity is ablated by mutation of Lys(113) within the presumtive catalytic motif II (invariant Lys). 293 cells stably transfected with H11 acquire anchorage-independent growth. Endogenous H11 RNA and the H11 phosphoprotein are expressed in melanoma cell lines and primary melanoma tissues at levels higher than in normal melanocytes and in benign nevi. Melanoma cell proliferation is inhibited by treatment with antisense oligonucleotides that inhibit H11 translation, suggesting that H11 expression is associated with cell growth.
Collapse
Affiliation(s)
- C C Smith
- Virology/Immunology Laboratories, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
21
|
Chen MR, Chang SJ, Huang H, Chen JY. A protein kinase activity associated with Epstein-Barr virus BGLF4 phosphorylates the viral early antigen EA-D in vitro. J Virol 2000; 74:3093-104. [PMID: 10708424 PMCID: PMC111808 DOI: 10.1128/jvi.74.7.3093-3104.2000] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Epstein-Barr virus (EBV) open reading frame BGLF4 was identified as a potential Ser/Thr protein kinase gene through the recognition of amino acid sequence motifs characteristic of conserved regions within the catalytic domains of protein kinases. In order to investigate this potential kinase activity, BGLF4 was expressed in Escherichia coli and the purified protein was used to generate a specific antiserum. Recombinant vaccinia virus vTF7-3, which expresses the T7 RNA polymerase, was used to infect 293 and 293T cells after transient transfection with a plasmid containing BGLF4 under the control of the T7 promoter. Autophosphorylation of the BGLF4 protein was demonstrated using the specific antiserum in an immune complex kinase assay. In addition, EBNA-1-tagged BGLF4 and EBNA-1 monoclonal antibody 5C11 were used to demonstrate the specificity of the kinase activity and to locate BGLF4 in the cytoplasm of transfected cells. Manganese ions were found to be essential for autophosphorylation of BGLF4, and magnesium can stimulate the activity. BGLF4 can utilize GTP, in addition to ATP, as a phosphate donor in this assay. BGLF4 can phosphorylate histone and casein in vitro. Among the potential viral protein substrates we examined, the EBV early antigen (EA-D, BMRF1), a DNA polymerase accessory factor and an important transactivator during lytic infection, was found to be phosphorylated by BGLF4 in vitro. Amino acids 1 to 26 of BGLF4, but not the predicted conserved catalytic domain, were found to be essential for autophosphorylation of BGLF4.
Collapse
Affiliation(s)
- M R Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | | | | | | |
Collapse
|
22
|
Raya A, Revert F, Navarro S, Saus J. Characterization of a novel type of serine/threonine kinase that specifically phosphorylates the human goodpasture antigen. J Biol Chem 1999; 274:12642-9. [PMID: 10212244 DOI: 10.1074/jbc.274.18.12642] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Goodpasture disease is an autoimmune disorder that occurs naturally only in humans. Also exclusive to humans is the phosphorylation process that targets the unique N-terminal region of the Goodpasture antigen. Here we report the molecular cloning of GPBP (Goodpasture antigen-binding protein), a previously unknown 624-residue polypeptide. Although the predicted sequence does not meet the conventional structural requirements for a protein kinase, its recombinant counterpart specifically binds to and phosphorylates the exclusive N-terminal region of the human Goodpasture antigen in vitro. This novel kinase is widely expressed in human tissues but shows preferential expression in the histological structures that are targets of common autoimmune responses. The work presented in this report highlights a novel gene to be explored in human autoimmunity.
Collapse
Affiliation(s)
- A Raya
- Fundación Valenciana de Investigaciones Biomédicas, Instituto de Investigaciones Citológicas, 46010 Valencia, Spain
| | | | | | | |
Collapse
|
23
|
Abstract
Protein kinase CK2 is a pleiotropic, ubiquitous and constitutively active protein kinase that can use both ATP and GTP as phosphoryl donors with specificity for serine/threonine residues in the vicinity of acidic amino acids. Recent results show that the enzyme is involved in transcription, signaling, proliferation and in various steps of development. The tetrameric holoenzyme (alpha2beta2) consists of two catalytic alpha-subunits and two regulatory beta-subunits. The structure of the catalytic subunit with the fixed positioning of the activation segment in the active conformation through its own aminoterminal region suggests a regulation at the transcriptional level making a regulation by second messengers unlikely. The high conservation of the catalytic subunit from yeast to man and its role in the tetrameric complex supports this notion. The regulatory beta-subunit has been far less conserved throughout evolution. Furthermore the existence of different CK2beta-related proteins together with the observation of deregulated CK2beta levels in tumor cells and the reported association of CK2beta protein with key proteins in signal transduction, e.g. A-Raf, Mos, pg90rsk etc. are suggestive for an additional physiological role of CK2beta protein beside being the regulatory compound in the tetrameric holoenzyme.
Collapse
Affiliation(s)
- B Guerra
- Biokemisk Institut, Odense Universitet, Denmark
| | | |
Collapse
|