Editorial
Varicella zoster virus: The potentially useful virus
Al-Anazi KA1* and Al-Jasser AM2
1Department of Hematology and Hematopoietic Stem Cell Transplantation, King Fahad Specialist Hospital, P.O. Box: 15215, Dammam 31444, Saudi Arabia
2Department of Research and Studies, General Directorate of Health Affairs in Riyadh Region, Ministry of Health, Riyadh 12822, Saudi Arabia
*Address for Correspondence: Khalid Ahmed Al-Anazi, Consultant Hemato-Oncologist and Chairman, Department of Hematology and Hematopoietic, Stem Cell Transplantation, Oncology Center, King Fahad Specialist Hospital, P.O. Box: 15215, Dammam 31444, Saudi Arabia, Fax: 966-13-8427420, Tel: 966 - 03- 8431111; Email: kaa_alanazi@yahoo.com
Dates: Submitted: 25 June 2019; Approved: 04 July 2019; Published: 05 July 2019
How to cite this article: Al-Anazi KA, Al-Jasser AM. Varicella zoster virus: The potentially useful virus. J Hematol Clin Res. 2019; 3: 011-015. DOI: 10.29328/journal.jhcr.1001009
Copyright License: © 2019 Al-Anazi KA, et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Varicella zoster virus (VZV), a double-stranded DNA virus, is a highly contagious human neurotropic virus that belongs to the alpha group of herpes viruses [1-4]. Primary VZV infection (chickenpox) occurs in childhood then the virus becomes latent in the nerve ganglia [1,5-7]. Reactivation of the virus may occur decades later and cause herpes zoster (HZ) which is manifested by a typical painful skin eruption that has characteristic dermatomal distribution [1,5]. Reactivation of VZV is usually predisposed to: old age; comorbid medical conditions such as diabetes mellitus, chronic obstructive airway disease, and end-stage renal disease; and immunosuppression due to malignancy, autoimmune disorders, immunosuppressive therapies, trauma, cytotoxic chemotherapy, hematopoietic stem cell transplantation (HSCT), and solid organ transplantation (SOT) [1,5-7].
VZV infections can cause not only transient pancytopenia but also aplastic anemia that may require allogeneic HSCT [8-12]. Additionally, VZV infections have been reported to be associated with increased risk of developing lymphoid malignancies and solid tumors [13-17]. On the contrary, there is growing evidence showing certain beneficial effects of the virus in immunocompromised individuals and these effects may be translated into prolongation of overall survival (OS) [18,19]. In a single center, case-controlled, retrospective study that included 16 episodes of VZV infections occurring in 14 patients with various types of hematologic malignancies (HMs) and bone marrow (BM) failure syndromes, Al-Anazi KA et al., reported an increase in white blood cell count, hemoglobin (Hb) level, and platelet count starting approximately 6 weeks following VZV infections and this stimulation of the 3 hematopoietic cell lines in the BM that followed VZV infections was maintained for periods longer than 3 years following the infection [18]. In another single center retrospective study that included a large number of patients with multiple myeloma subjected to high-dose melphalan followed by autologous HSCT after control of their primary disease, Kamber C et al., reported that approximately one third of these patients developed VZV infections either before or after HSCT [19]. Despite encountering VZV infections in patients with worse expected prognosis, the OS in patients who developed VZV infection was superior to that in patients who never developed the infection [19]. Recently, Al-Anazi KA et al., reported BM biopsy-proven reversal of pure red cell aplasia manifested by a gradual increase in Hb level starting 6 weeks following a localized HZ infection till the Hb level plateaued above 14g/dL fourteen months following the viral infection [4]. Additionally, several studies have demonstrated that VZV infection may trigger chronic graft versus host disease (GVHD) following allogeneic HSCT [20-22]. GVHD is associated with graft versus cancer effects and, provided GVHD is of low-grade, it can be associated with improvement in OS in patients with acute leukemia or lymphoma [23-25].
Although viruses can cause infectious complications that may be associated with significant morbidity and mortality and evolution of certain cancers, they may provide hope to effectively treat several serious medical illnesses by being utilized as: vaccines; anticancer agents in the setting of oncolytic virus therapy; as well as vectors in induced pluripotent stem cells, gene therapy for several hereditary and acquired disorders, and chimeric antigen receptor (CAR) T-cell therapy [26-33]. VZV is the only virus consistently reported to have an inverse association with glioma suggesting a protective effect of VZV infection against the tumor [34,35]. Studies have shown that: the protective effect of prior VZV infection against glioma is stronger for high-grade tumor; this protective effect may be mediated by the VZV-specific T-lymphocytes; VZV exhibits an extrinsic oncolytic potential in malignant glioma cultures; and human mesenchymal stem cells (MSCs) are suitable for delivering VZV to the sites of tumor growth [34,36,37]. However, efficacy of oncolytic virotherapy in malignant glioma has several difficulties that need to be overcome [35].
VZV pathogenesis, latency, and reactivation are difficult to study due to the fact that VZV is an exclusively human pathogen [38-40]. Numerous efforts have been made to develop adequate animal models of VZV infection but these models remain limited because all aspects of VZV infection, latency and reactivation, and understanding VZV pathology will remain not only difficult but also incomplete without a suitable model [38,40]. Despite the rare reports of breakthrough VZV infections that may become disseminated and life-threatening particularly in immunocompromised hosts, VZV vaccines including the live-attenuated ones have been shown to be generally safe and effective in: recipients of SOT as well as HSCT; patients with HMs and solid tumors; patients with diabetes mellitus, autoimmune disorders and renal disease; elderly individuals on corticosteroid maintenance therapy; and patients with history of HZ infection [41-55].
VZV infections are associated with specific alterations in the BM microenvironment induced by stress-related hematopoiesis [56-58]. Studies have shown that the following stromal and immune cells are involved the pathogenesis of VZV: (1) MSCs, the masters of hematopoiesis, which have antimicrobial and anticancer effects [59-62]; (2) dendritic cells, the essential cells for initiating antiviral immune response, which are found to transmit VZV to T-cells in order for the virus to disrupt the function of immune cells [63,64]; (3) natural killer cells, which play important roles in controlling VZV infection and eliminating malignant cells, may themselves become manipulated by the virus for its advantage [65,66]; (4) T-lymphocytes, including CD4, CD8 effector cells and memory T cells, that are involved in T cell immunity encountered following primary VZV infection and in maintenance of latency [67]; and (5) mononuclear cells, which once infected with VZV, dessiminate the virus to distal organs to produce clinical disease [68]. Additionally, the following cellular proteins are involved in the pathogenesis of VZV infections: open reading frames, glycoproteins, promyelocytic leukemia protein, chaperons and small ubiquitin-like modifier proteins [69-73]. Extracellular vesicles, exosomes, and micro-RNAS are also implicated in VZV infections [74-76]. Studies have also shown that several cytokines, chemokines and ligands are involved in VZV infections and that certain complications of VZV infections such as: postherpetic neuralgia, vasculopathy, myelopathy, encephalopathy, and acute retinal necrosis have specific cytokine profiles [77-81]. A number of signaling pathways; such as Janus kinase/signal transducer and activator of transcription proteins (JAK/STAT), extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and PI3K/Akt pathways; have been found to be activated in VZV infections [82-85].
Apparently, VZV behaves differently from other herpes viruses. Additionally, it has the following peculiar features: (1) having the smallest genome; (2) losing almost all the genes that are not essential for its survival; (3) being highly fusogenic and cell-associated; (4) having no inhibitors of autophagy; (5) being an exclusively human pathogen; (6) having a species-specific cytokine profile; (7) having an inverse relationship with glioma; (8) association with GVHD in recipients with allogeneic HSCT; and (9) having BM stimulatory effects as well as several antitumor actions in patients with HMs and BM failure syndromes [1-4,18-22,34,35,86,87].
The reported beneficial effects may develop through several direct or indirect immunological mechanisms. Hence, these beneficial effects merit thorough investigations and should encourage scientists and researchers to give this potentially useful virus the attention it deserves. The antitumor effects as well as the stimulatory effect exerted by the virus on the 3 cell lines in the BM can be explained by one or more of the suggested mechanisms or may be due to a new mechanism yet to be elucidated. The virus itself, modified or engineered versions of the virus, or specific materials obtained from the serum of patients infected with VZV may ultimately become novel therapeutic modalities in the management of these immunocompromised patients.
References
- Kennedy PGE, Gershon AA. Clinical features of varicella-zoster virus infection. Viruses. 2018; 10: E609. PubMed: https://bit.ly/2XqPx7d
- Weber DJ. Prevention and control of varicella-zoster virus in hospitals.2019.
- Cohrs RJ, Gilden DH, Mahalingam R. Varicella zoster virus latency, neurological disease and experimental models: an update. Front Biosci. 2004; 9: 751-762. PubMed: https://bit.ly/32aVMuQ
- Al-Anazi KA, Kanfar S, Aldayel A, Abduljalil O, Sayyed AH. Reversal of pure red cell aplasia by varicella zoster virus infection. J Hematol Clin Res 2019; 3: 001-010.
- Albrecht MA, Levin MJ. Epidemiology, clinical manifestations and diagnosis of herpes zoster. 2019.
- Onozawa M, Hashino S, Takahata M, Fujisawa F, Kawamura T, et al. Relationship between preexisting anti-varicella-zoster virus (VZV) antibody and clinical VZV reactivation in hematopoietic stem cell transplantation recipients. J Clin Microbiol. 2006; 44: 4441-4443. PubMed: https://bit.ly/2XnEMOc
- Cvjetković D, Jovanović J, Hrnjaković-Cvjetković I, Brkić S, Bogdanović M. Reactivation of herpes zoster infection by varicella-zoster virus. Med Pregl. 1999; 52: 125-128. PubMed: https://bit.ly/2FTuC1X
- Yetgin S, Kuşkonmaz B, Aytaç S, Cetin M. The evaluation of acquired aplastic anemia in children and unexpected frequency of varicella-zoster virus association: a single-center study. Turk J Pediatr. 2008; 50: 342-348. PubMed: https://bit.ly/2FOIURq
- Pascutti MF, Erkelens MN, Nolte MA. Impact of viral infections on hematopoiesis: from beneficial to detrimental effects on bone marrow output. Front Immunol. 2016; 7: 364. PubMed: https://bit.ly/2XrrHTX
- Kakish K, Basak RB, Al Dhuhouri J, Chakraborty S. Four year old child with breakthrough varicella leading to pancytopenia. Bahrain Med Bull. 2009; 31, 2.
- Muthu V, Kumar PS, Varma S, Malhotra P. Varicella zoster virus-related pancytopenia. Int J Infect Dis. 2013; 17: e1264. PubMed: https://bit.ly/2xqOsNw
- Kuskonmaz B, Cetin M, Uckan D, Yetgin S. Varicella zoster-associated severe aplastic anemia in a child and its successful treatment with peripheral blood stem cell transplantation from HLA-5/6-identical donor. Med Sci Monit. 2007; 13: CS128-131. PubMed: https://bit.ly/2XnY1qZ
- Ragozzino MW, Melton LJ 3rd, Kurland LT, Chu CP, Perry HO. Risk of cancer after herpes zoster: a population-based study. N Engl J Med. 1982; 307: 393-397. PubMed: https://bit.ly/2RWKYM3
- Ho JD, Xirasagar S, Lin HC. Increased risk of a cancer diagnosis after herpes zoster ophthalmicus: a nationwide population-based study. Ophthalmology. 2011; 118: 1076-1081. PubMed: https://bit.ly/2JoKbj4
- Schmidt SA, Mor A, Schønheyder HC, Sørensen HT, Dekkers OM, et al. Herpes zoster as a marker of occult cancer: A systematic review and meta-analysis. J Infect. 2017; 74: 215-235. PubMed: https://bit.ly/2XE7HC3
- Liu YC, Yang YH, Hsiao HH, Yang WC, Liu TC, et al. Herpes zoster is associated with an increased risk of subsequent lymphoid malignancies - a nationwide population-based matched-control study in Taiwan. BMC Cancer. 2012; 12: 503. PubMed: https://bit.ly/309KcOz
- Mays RM, Murthy RK, Gordon RA, Lapolla WJ, Galfione SK, et al. Diffuse Large B-Cell Lymphoma at the site of a herpes zoster scar. World J Oncol. 2012; 3: 199-203. PubMed: https://bit.ly/2Jlt7up
- Al-Anazi KA, Al-Jasser AM, Evans DA. Effect of varicella zoster virus infection on bone marrow function. Eur J Haematol. 2005; 75: 234-240. PubMed: https://bit.ly/2YwMzL7
- Kamber C, Zimmerli S, Suter-Riniker F, Mueller BU, Taleghani BM, et al. Varicella zoster virus reactivation after autologous SCT is a frequent event and associated with favorable outcome in myeloma patients. Bone Marrow Transplant. 2015; 50: 573-578. PubMed: https://bit.ly/2KXAsU3
- Kawano N, Gondo H, Kamimura T, Aoki K, Iino T, et al. Chronic graft-versus-host disease following varicella-zoster virus infection in allogeneic stem cell transplant recipients. Int J Hematol. 2003; 78: 370-373. PubMed: https://bit.ly/2RQFvXc
- Raymond AK, Singletary HL, Nelson KC, Sidhu-Malik NK. Dermatomal sclerodermoid graft-vs-host disease following varicella-zoster virus infection. Arch Dermatol. 2011; 147: 1121-1122. PubMed: https://bit.ly/2XJokfn
- Baselga E, Drolet BA, Segura AD, Leonardi CL, Esterly NB. Dermatomal lichenoid chronic graft-vs-host disease following varicella-zoster infection despite absence of viral genome. J Cutan Pathol. 1996; 23: 576-581. PubMed: https://bit.ly/2NGLdfA
- Weisdorf D, Zhang MJ, Arora M, Horowitz MM, Rizzo JD, et al. Graft-versus-host disease induced graft-versus-leukemia effect: greater impact on relapse and disease-free survival after reduced intensity conditioning. Biol Blood Marrow Transplant. 2012; 18: 1727-1733. PubMed: https://bit.ly/30a1MSG
- Yeshurun M, Weisdorf D, Rowe JM, Tallman MS, Zhang MJ, et al. The impact of the graft-versus-leukemia effect on survival in acute lymphoblastic leukemia. Blood Adv. 2019; 3: 670-680. PubMed: https://bit.ly/2XE8vqz
- Negrin RS. Graft-versus-host disease versus graft-versus-leukemia. Hematology Am Soc Hematol Educ Program. 2015; 2015: 225-30.
- Belcaid Z, Lamfers ML, van Beusechem VW, Hoeben RC. Changing faces in virology: the Dutch shift from oncogenic to oncolytic viruses. Hum Gene Ther. 2014; 25: 875-884. PubMed: https://bit.ly/2XrnYWy
- Rudd PA, Herrero LJ. Viruses: Friends and foes. In: Cartilage repair and regeneration. 2017.
- Fukuhara H, Ino Y, Todo T. Oncolytic virus therapy: A new era of cancer treatment at dawn. Cancer Sci. 2016; 107: 1373-1379. PubMed: https://bit.ly/2xqQbT0
- Ajina A, Maher J. Prospects for combined use of oncolytic viruses and CAR T-cells. J Immunother Cancer. 2017; 5: 90.
- Howells A, Marelli G Lemoine NR, Wang Y. Oncolytic viruses-interaction of virus and tumor cells in the battle to eliminate cancer. Front Oncol. 2017; 7: 195. PubMed: https://bit.ly/2LAu6th
- Guo C, Manjili MH, Subjeck JR, Sarkar D, Fisher PB, et al. Therapeutic cancer vaccines: past, present, and future. Adv Cancer Res. 2013; 119: 421-475. PubMed: https://bit.ly/2RTgtqa
- Chaurasiya S, Chen NG, Warner SG. Oncolytic virotherapy versus cancer stem cells: a review of approaches and mechanisms. Cancers. 2018; 10: E124. PubMed: https://bit.ly/2KYBwY1
- Wennier ST, Liu J, McFadden G. Bugs and drugs: oncolytic virotherapy in combination with chemotherapy. Curr Pharm Biotechnol. 2012; 13: 1817-1833. PubMed: https://bit.ly/30cuW3G
- Amirian ES, Scheurer ME, Zhou R, Wrensch MR, Armstrong GN, et al. History of chickenpox in glioma risk: a report from the glioma international case-control study (GICC). Cancer Med. 2016; 5: 1352-1358. PubMed: https://bit.ly/2Nx6Cbg
- Pundole X, Amirian ES, Scheurer ME. Role of varicella zoster virus in glioma risk: Current knowledge and future directions. OA Epidemiology. 2014; 2: 6.
- Canniff J, Donson AM, Foreman NK, Weinberg A. Cytotoxicity of glioblastoma cells mediated ex vivo by varicella-zoster virus-specific T cells. J Neurovirol. 2011; 17: 448-454. PubMed: https://bit.ly/2YCBWqg
- Leske H, Haase R, Restle F, Schichor C, Albrecht V, et al. Varicella zoster virus infection of malignant glioma cell cultures: a new candidate for oncolytic virotherapy? Anticancer Res. 2012; 32: 1137-1144.
- Haberthur K, Messaoudi I. Animal models of varicella zoster virus infection. Pathogens. 2013; 2: 364-382. PubMed: https://bit.ly/2JmPjEp
- Goodwin TJ, McCarthy M, Osterrieder N, Cohrs RJ, Kaufer BB. Three-dimensional normal human neural progenitor tissue-like assemblies: a model of persistent varicella-zoster virus infection. PLoS Pathog. 2013; 9: e1003512. PubMed: https://bit.ly/2Xsp7SM
- Shahzad A, Gilden D, Cohrs RJ. Translational medicine and varicella zoster virus: Need for disease modeling. New Horiz Transl Med. 2015; 2: 89-91. PubMed: https://bit.ly/2xsMZ9h
- Harada K, Heaton H, Chen J, Vazquez M, Meyer J. Zoster vaccine-associated primary varicella infection in an immunocompetent host. BMJ Case Rep. 2017; 2017: bcr-2017-221166. PubMed: https://bit.ly/2XJoDXz
- Leung J, Broder KR, Marin M. Severe varicella in persons vaccinated with varicella vaccine (breakthrough varicella): A systematic literature review. Expert Rev Vaccines. 2017; 16: 391-400. PubMed: https://bit.ly/2LCIscz
- Khandelwal P, Marsh RA, Scott Scmid D, Radford KW, Bleesing J, et al. Case series of vaccine associated varicella zoster virus infection in immune compromised patients. Biol Blood Marrow Transplant. 2013; 19: S250.
- Bhalla P, Forrest GN, Gershon M, Zhou Y, Chen J, et al. Disseminated, persistent, and fatal infection due to the vaccine strain of varicella-zoster virus in an adult following stem cell transplantation. Clin Infect Dis. 2015; 60: 1068-1074. PubMed: https://bit.ly/2NxuXNW
- Willis ED, Woodward M, Brown E, Popmihajlov Z, Saddier P, et al. Herpes zoster vaccine live: a 10 year review of post-marketing safety experience. Vaccine. 2017; 35: 7231-7239. PubMed: https://bit.ly/2FTwTu1
- Wang L, Verschuuren EAM, van Leer-Buter CC, Bakker SJL, de Joode AAE, et al. Herpes zoster and immunogenicity and safety of zoster vaccines in transplant patients: a narrative review of the literature. Front Immunol. 2018; 9: 1632. PubMed: https://bit.ly/2Nz1a7F
- Aoki T, Koh K, Kawano Y, Mori M, Arakawa Y, et al. Safety of live attenuated high-titer varicella-zoster virus vaccine in pediatric allogeneic hematopoietic stem cell transplantation recipients. Biol Blood Marrow Transplant. 2016; 22: 771-775. PubMed: https://bit.ly/30cIWuk
- Winston DJ, Mullane KM, Cornely OA, Boeckh MJ, Brown JW, et al.; V212 Protocol 001 Trial Team. Inactivated varicella zoster vaccine in autologous haemopoietic stem-cell transplant recipients: An international, multicentre, randomised, double-blind, placebo-controlled trial. Lancet 2018; 391: 2116-2127. PubMed: https://bit.ly/2NwjDBI
- Sasadeusz J, Prince HM, Schwarer A, Szer J, Stork A, et al. Immunogenicity and safety of a two-dose live attenuated varicella vaccine given to adults following autologous hematopoietic stem cell transplantation. Transpl Infect Dis. 2014; 16: 1024-1031. PubMed: https://bit.ly/2NA4sre
- Parrino J, McNeil SA, Lawrence SJ, Kimby E, Pagnoni MF, et al. Safety and immunogenicity of inactivated varicella-zoster virus vaccine in adults with hematologic malignancies receiving treatment with anti-CD20 monoclonal antibodies. Vaccine. 2017; 35: 1764-1769. PubMed: https://bit.ly/2Jc7XA8
- Leung TF, Li CK, Hung EC, Chan PK, Mo CW, et al. Immunogenicity of a two-dose regime of varicella vaccine in children with cancers. Eur J Haematol. 2004; 72: 353-357. PubMed: https://bit.ly/2xsqgu5
- Ohfuji S, Ito K, Inoue M, Ishibashi M, Kumashiro H, et al. Safety of live attenuated varicella-zoster vaccine in patients with underlying illnesses compared with healthy adults: A prospective cohort study. BMC Infect Dis. 2019; 19: 95. PubMed: https://bit.ly/2S2jtkt
- Eberhardson M, Hall S, Papp KA, Sterling TM, Stek JE, et al. Safety and immunogenicity of inactivated varicella-zoster virus vaccine in adults with autoimmune disease: A phase 2, randomized, double-blind, placebo-controlled clinical trial. Clin Infect Dis. 2017; 65: 1174-1182. PubMed: https://bit.ly/2Xr0DZT
- Russell AF, Parrino J, Fisher CL Jr, Spieler W, Stek JE, et al. Safety, tolerability, and immunogenicity of zoster vaccine in subjects on chronic/ maintenance corticosteroids. Vaccine. 2015; 33: 3129-3134. PubMed: https://bit.ly/2FQXMyM
- Mills R, Tyring SK, Levin MJ, Parrino J, Li X, et al. Safety, tolerability, and immunogenicity of zoster vaccine in subjects with a history of herpes zoster. Vaccine. 2010; 28: 4204-4209. PubMed: https://tinyurl.com/y6sa9nhs
- Agarwala S, Tamplin OJ. Neural crossroads in the hematopoietic stem cell niche. Trends Cell Biol. 2018; 28: 987-998. PubMed: https://tinyurl.com/yxjeyetc
- Boiko JR, Borghesi L. Hematopoiesis sculpted by pathogens: Toll-like receptors and inflammatory mediators directly activate stem cells. Cytokine. 2012; 57:1-8. PubMed: https://tinyurl.com/y4gk5xcw
- Zhao JL, Baltimore D. Regulation of stress-induced hematopoiesis. Curr Opin Hematol. 2015; 22: 286-292.
- Pleyer L, Valent P, Greil R. Mesenchymal stem and progenitor cells in normal and dysplastic hematopoiesis-masters of survival and clonality? Int J Mol Sci. 2016; 17: E1009. PubMed: https://tinyurl.com/y56v96kc
- Avanzi S, Leoni V, Rotola A, Alviano F, Solimando L, et al. Susceptibility of human placenta derived mesenchymal stromal/stem cells to human herpesviruses infection. PLoS One. 2013; 8: e71412. PubMed: https://tinyurl.com/y6s43fcl
- Auletta JJ, Deans RJ, Bartholomew AM. Emerging roles for multipotent, bone marrow-derived stromal cells in host defense. Blood. 2012; 119:1801-1809. PubMed: https://tinyurl.com/y3473tet
- Nowakowski A, Drela K, Rozycka J, Janowski M, Lukomska B. Engineered mesenchymal stem cells as an anti-cancer Trojan horse. Stem Cells Dev. 2016; 25: 1513-1531. PubMed: https://tinyurl.com/y5x8ths2
- Abendroth A, Morrow G, Cunningham AL, Slobedman B. Varicella-zoster virus infection of human dendritic cells and transmission to T cells: Implications for virus dissemination in the host. J Virol. 2001; 75: 6183-6192. PubMed: https://tinyurl.com/y3uvhnd4
- Morrow G, Slobedman B, Cunningham AL, Abendroth A. Varicella-zoster virus productively infects mature dendritic cells and alters their immune function. J. Virol. 2003; 77: 4950-4959. PubMed: https://tinyurl.com/y53kdqqs
- Handgretinger R, Lang P, André MC. Exploitation of natural killer cells for the treatment of acute leukemia. Blood. 2016; 127: 3341-3349. PubMed: https://tinyurl.com/y36xbrcu
- Campbell TM, McSharry BP, Steain M, Ashhurst TM, Slobedman B, et al. Varicella zoster virus productively infects human natural killer cells and manipulates phenotype. PLoS Pathog. 2018; 14: e1006999. PubMed: https://tinyurl.com/y6eusgrw
- Weinberg A, Levin MJ. VZV T cell-mediated immunity. Curr Top Microbiol. Immunol. 2010; 342: 341-57.
- Jones D, Como CN, Jing L, Blackmon A, Neff CP, et al. Varicella zoster virus productively infects human peripheral blood mononuclear cells to modulate expression of immunoinhibitory proteins and blocking PD-L1 enhances virus-specific CD8+ T cell effector function. PLoS Pathog. 2019; 15: e1007650. PubMed: https://tinyurl.com/y3bww5x6
- Sommer MH, Zagha E, Serrano OK, Ku CC, Zerboni L, et al. Mutational analysis of the repeated open reading frames, ORFs 63 and 70 and ORFs 64 and 69, of varicella-zoster virus. J Virol. 2001; 75: 8224-8239. PubMed: https://tinyurl.com/y29a7r7r
- Lenac Roviš T, Bailer SM, Pothineni VR, Ouwendijk WJ, Šimić H, et al. Comprehensive analysis of varicella-zoster virus proteins using a new monoclonal antibody collection. J Virol. 2013; 87: 6943-6954. PubMed: https://tinyurl.com/y3w5paex
- Lallemand-Breitenbach V, de Thé H. PML nuclear bodies. Curr Opin Cell Biol. 2018; 52: 154-161. PubMed: https://tinyurl.com/y4plm9lj
- Kyratsous CA, Silverstein SJ. BAG3, a host cochaperone, facilitates varicella-zoster virus replication. J Virol. 2007; 81: 7491-7503. PubMed: https://tinyurl.com/y5r9yg7o
- Mattoscio D, Segré CV, Chiocca S. Viral manipulation of cellular protein conjugation pathways: The SUMO lesson. World J Virol. 2013; 2: 79-90. PubMed: https://tinyurl.com/y3ntxxq8
- Bello-Morales R, López-Guerrero JA. Extracellular vesicles in herpes viral spread and immune evasion. Front Microbiol. 2018; 9: 2572. PubMed: https://tinyurl.com/y435rqj5
- Anderson MR, Kashanchi F, Jacobson S. Exosomes in viral disease. Neurotherapeutics. 2016; 13: 535-546.
- Li X, Huang Y, Zhang Y, He N. Evaluation of microRNA expression in patients with herpes zoster. Viruses. 2016; 8: E326. PubMed: https://tinyurl.com/y4fxlzdz
- Zajkowska A, Garkowski A, Świerzbińska R, Kułakowska A, Król ME, et al. Evaluation of chosen cytokine levels among patients with herpes zoster as ability to provide immune response. PLoS One. 2016; 11: e0150301. PubMed: https://tinyurl.com/y4a6gg23
- Zak-Prelich M, McKenzie RC, Sysa-Jedrzejowska A, Norval M. Local immune responses and systemic cytokine responses in zoster: relationship to the development of postherpetic neuralgia. Clin Exp Immunol. 2003; 131: 318-323. PubMed: https://tinyurl.com/y3spcyb9
- Jones D, Neff CP, Palmer BE, Stenmark K, Nagel MA. Varicella zoster virus-infected cerebrovascular cells produce a proinflammatory environment. Neurol Neuroimmunol Neuroinflamm. 2017; 4: e382. PubMed: https://tinyurl.com/y5r8jtq7
- Bubak AN, Como CN, Blackmon AM, Jones D, Nagel MA. Varicella zoster virus differentially alters morphology and suppresses proinflammatory cytokines in primary human spinal cord and hippocampal astrocytes. J Neuroinflammation. 2018; 15: 318. PubMed: https://tinyurl.com/y3uc6edb
- de Visser L, H de Boer J, T Rijkers G, Wiertz K, van den Ham HJ, et al. Cytokines and chemokines involved in acute retinal necrosis. Invest Ophthalmol Vis Sci. 2017; 58: 2139-2151. PubMed: https://tinyurl.com/y2ome53j
- Verweij MC, Wellish M, Whitmer T, Malouli D, Lapel M, et al. Varicella viruses inhibit interferon-stimulated JAK-STAT signaling through multiple mechanisms. PLoS Pathog. 2015; 11: e1004901. PubMed: https://tinyurl.com/y6nn3dsp
- Rahaus M, Desloges N, Wolff MH. Varicella-zoster virus influences the activities of components and targets of the ERK signalling pathway. J Gen Virol. 2006; 87: 749-758. PubMed: https://tinyurl.com/y2axb3fg
- Kurapati S, Sadaoka T, Rajbhandari L, Jagdish B, Shukla P, et al. Role of the JNK pathway in varicella-zoster virus lytic infection and Reactivation. J Virol. 2017; 91: e00640-17. PubMed: https://tinyurl.com/y55zksve
- Rahaus M, Desloges N, Wolff MH. Varicella-zoster virus requires a functional PI3K/Akt/GSK-3alpha/beta signaling cascade for efficient replication. Cell Signal. 2007; 19: 312-320. PubMed: https://tinyurl.com/y3magfsr
- Grigoryan S, Yee MB, Glick Y, Gerber D, Kepten E, et al. Direct transfer of viral and cellular proteins from varicella-zoster virus-infected non-neuronal cells to human axons. PLoS One. 2015; 10: e0126081. PubMed: https://tinyurl.com/y68bxr5w
- Grose C, Buckingham EM, Carpenter JE, Kunkel JP. Varicella-zoster virus infectious cycle: ER stress, autophagic flux, and amphisome-mediated trafficking. Pathogens. 2016; 5: E67. PubMed: https://tinyurl.com/y63xvhww