1
|
Walker M, Federico E, Zunt JR, Levitt MR, Johnston CM. Quantitative analysis of HSV‑1 shedding as a predictor of cerebral vasospasm severity in patients with subarachnoid hemorrhage. Biomed Rep 2024; 21:177. [PMID: 39387003 PMCID: PMC11462505 DOI: 10.3892/br.2024.1865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Cerebral vasospasm (CV) is a critical determinant of outcomes in patients with aneurysmal subarachnoid hemorrhage (aSAH). Despite advances in neurocritical care, modifiable risk factors for CV remain poorly understood, and identifying them could significantly enhance patient management and treatment strategies. The present study explored the potential link between the reactivation of herpes simplex virus type 1 (HSV-1), a common resident virus in cranial nerves, and CV severity. It was hypothesized that higher HSV-1 viral load in saliva may be associated with increased CV severity. Saliva samples were collected on days 4, 7, 10 and 14 post-aSAH, and HSV-1 DNA levels were measured using quantitative PCR. CV severity was assessed using the Lindegaard ratio (LR), with an LR >3 considered the diagnostic threshold for CV. A total of 36 patients were enrolled, and 139 saliva samples were collected. HSV-1 DNA was detected in 19.4% of samples (27/139), and 44% of patients (16/36) developed CV. HSV-1 seropositive patients made up 88.9% (32/36) of the cohort, with 50% exhibiting viral shedding during the study period. None of the HSV-1 seronegative patients (11.1%, 4/36) exhibited viral shedding or developed CV. Regression analysis showed a positive association between HSV-1 viral load and CV severity, with viral load explaining 27.8% of the variability (P=0.005). Age was also significant, with older patients experiencing less severe CV (P<0.001). Supervised machine learning identified viral load thresholds that aligned with standard LR values for moderate and severe CV. While the small sample size and observational design limit the generalizability of the results, these findings suggested that earlier detection and intervention for CV could be informed by assessing HSV-1 serostatus and monitoring viral activity through saliva samples or other non-invasive methods, highlighting the need for larger, controlled studies to validate these results.
Collapse
Affiliation(s)
- Melanie Walker
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA 98104, USA
- Stroke and Applied NeuroSciences Center, University of Washington School of Medicine, Seattle, WA 98104, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Emma Federico
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA 98104, USA
- Stroke and Applied NeuroSciences Center, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Joseph R. Zunt
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98104, USA
- Department of Medicine, Division of Infectious Disease, University of Washington, Seattle, WA 98104, USA
| | - Michael R. Levitt
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA 98104, USA
- Stroke and Applied NeuroSciences Center, University of Washington School of Medicine, Seattle, WA 98104, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98104, USA
- Department of Radiology, University of Washington School of Medicine, Seattle, WA 98104, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98104, USA
| | - Christine M. Johnston
- Department of Medicine, Division of Infectious Disease, University of Washington, Seattle, WA 98104, USA
| |
Collapse
|
2
|
Joyce JD, Moore GA, Goswami P, Harrell TL, Taylor TM, Hawks SA, Green JC, Jia M, Irwin MD, Leslie E, Duggal NK, Thompson CK, Bertke AS. SARS-CoV-2 Rapidly Infects Peripheral Sensory and Autonomic Neurons, Contributing to Central Nervous System Neuroinvasion before Viremia. Int J Mol Sci 2024; 25:8245. [PMID: 39125815 PMCID: PMC11311394 DOI: 10.3390/ijms25158245] [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: 05/21/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Neurological symptoms associated with COVID-19, acute and long term, suggest SARS-CoV-2 affects both the peripheral and central nervous systems (PNS/CNS). Although studies have shown olfactory and hematogenous invasion into the CNS, coinciding with neuroinflammation, little attention has been paid to susceptibility of the PNS to infection or to its contribution to CNS invasion. Here we show that sensory and autonomic neurons in the PNS are susceptible to productive infection with SARS-CoV-2 and outline physiological and molecular mechanisms mediating neuroinvasion. Our infection of K18-hACE2 mice, wild-type mice, and golden Syrian hamsters, as well as primary peripheral sensory and autonomic neuronal cultures, show viral RNA, proteins, and infectious virus in PNS neurons, satellite glial cells, and functionally connected CNS tissues. Additionally, we demonstrate, in vitro, that neuropilin-1 facilitates SARS-CoV-2 neuronal entry. SARS-CoV-2 rapidly invades the PNS prior to viremia, establishes a productive infection in peripheral neurons, and results in sensory symptoms often reported by COVID-19 patients.
Collapse
Affiliation(s)
- Jonathan D. Joyce
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Greyson A. Moore
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Poorna Goswami
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
| | - Telvin L. Harrell
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Tina M. Taylor
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Seth A. Hawks
- Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Jillian C. Green
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Mo Jia
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Matthew D. Irwin
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Emma Leslie
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
| | - Nisha K. Duggal
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
- Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Christopher K. Thompson
- School of Neuroscience, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Andrea S. Bertke
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| |
Collapse
|
3
|
Canova PN, Charron AJ, Leib DA. Models of Herpes Simplex Virus Latency. Viruses 2024; 16:747. [PMID: 38793628 PMCID: PMC11125678 DOI: 10.3390/v16050747] [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: 04/05/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Our current understanding of HSV latency is based on a variety of clinical observations, and in vivo, ex vivo, and in vitro model systems, each with unique advantages and drawbacks. The criteria for authentically modeling HSV latency include the ability to easily manipulate host genetics and biological pathways, as well as mimicking the immune response and viral pathogenesis in human infections. Although realistically modeling HSV latency is necessary when choosing a model, the cost, time requirement, ethical constraints, and reagent availability are also equally important. Presently, there remains a pressing need for in vivo models that more closely recapitulate human HSV infection. While the current in vivo, ex vivo, and in vitro models used to study HSV latency have limitations, they provide further insights that add to our understanding of latency. In vivo models have shed light on natural infection routes and the interplay between the host immune response and the virus during latency, while in vitro models have been invaluable in elucidating molecular pathways involved in latency. Below, we review the relative advantages and disadvantages of current HSV models and highlight insights gained through each.
Collapse
Affiliation(s)
- Paige N. Canova
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| | - Audra J. Charron
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| | - David A. Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH 03755, USA;
| |
Collapse
|
4
|
Zhang H, Yu M, Liang G, Li S, Zhao C, Jing K, Feng S. Prevalence and clinical markers of herpes simplex virus infection in oral lesions of bullous pemphigoid. Front Immunol 2024; 15:1387503. [PMID: 38698862 PMCID: PMC11063318 DOI: 10.3389/fimmu.2024.1387503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Background The manifestations of bullous pemphigoid (BP) and herpes simplex virus (HSV) infection are similar in oral mucosa, and the laboratory detection of HSV has some limitations, making it difficult to identify the HSV infection in oral lesions of BP. In addition, the treatments for BP and HSV infection have contradictory aspects. Thus, it is important to identify the HSV infection in BP patients in time. Objective To identify the prevalence and clinical markers of HSV infection in oral lesions of BP. Methods This prospective cross-sectional descriptive analytical study was conducted on 42 BP patients with oral lesions. A total of 32 BP patients without oral lesions and 41 healthy individuals were enrolled as control groups. Polymerase chain reaction was used to detect HSV. Clinical and laboratory characteristics of patients with HSV infection were compared with those without infection. Results A total of 19 (45.2%) BP patients with oral lesions, none (0.0%) BP patients without oral lesions, and four (9.8%) healthy individuals were positive for HSV on oral mucosa. Among BP patients with oral lesions, the inconsistent activity between oral and skin lesions (p=0.001), absence of blister/blood blister in oral lesions (p=0.020), and pain for oral lesions (p=0.014) were more often seen in HSV-positive than HSV-negative BP patients; the dosage of glucocorticoid (p=0.023) and the accumulated glucocorticoid dosage in the last 2 weeks (2-week AGC dosage) (p=0.018) were higher in HSV-positive BP patients. Combining the above five variables as test variable, the AUC was 0.898 (p<0.001) with HSV infection as state variable in ROC analysis. The absence of blister/blood blister in oral lesions (p=0.030) and pain for oral lesions (p=0.038) were found to be independent predictors of HSV infection in multivariable analysis. A total of 14 (73.7%) HSV-positive BP patients were treated with 2-week famciclovir and the oral mucosa BPDAI scores significantly decreased (p<0.001). Conclusion HSV infection is common in BP oral lesions. The inconsistent activity between oral and skin lesions, absence of blister in oral lesions, pain for oral lesions, higher currently used glucocorticoid dosage, and higher 2-week AGC dosage in BP patients should alert physicians to HSV infection in oral lesions and treat them with 2-week famciclovir in time.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Suying Feng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| |
Collapse
|
5
|
Dochnal SA, Whitford AL, Francois AK, Krakowiak PA, Cuddy S, Cliffe AR. c-Jun signaling during initial HSV-1 infection modulates latency to enhance later reactivation in addition to directly promoting the progression to full reactivation. J Virol 2024; 98:e0176423. [PMID: 38193709 PMCID: PMC10878265 DOI: 10.1128/jvi.01764-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/12/2023] [Indexed: 01/10/2024] Open
Abstract
Herpes simplex virus-1 (HSV-1) establishes a latent infection in peripheral neurons and periodically reactivates to permit transmission, which can result in clinical manifestations. Viral transactivators required for lytic infection are largely absent during latent infection, and therefore, HSV-1 relies on the co-option of neuronal host signaling pathways to initiate its gene expression. The activation of the neuronal c-Jun N-terminal kinase (JNK) cell stress pathway is central to initiating biphasic reactivation in response to multiple stimuli. However, how host factors work with JNK to stimulate the initial wave of gene expression (known as Phase I) or the progression to full Phase II reactivation remains unclear. Here, we found that c-Jun, the primary target downstream of neuronal JNK cell stress signaling, functions during reactivation but not during the JNK-mediated initiation of Phase I gene expression. Instead, c-Jun was required to transition from Phase I to full HSV-1 reactivation and was detected in viral replication compartments of reactivating neurons. Interestingly, we also identified a role for both c-Jun and enhanced neuronal stress during initial neuronal infection in promoting a more reactivation-competent form of HSV-1 latency. Therefore, c-Jun functions at multiple stages during the HSV latent infection of neurons to promote reactivation but not during the initial JNK-dependent Phase I. Importantly, by demonstrating that initial infection conditions can contribute to later reactivation abilities, this study highlights the potential for latently infected neurons to maintain a molecular scar of previous exposure to neuronal stressors.IMPORTANCEThe molecular mechanisms that regulate the reactivation of herpes simplex virus-1 (HSV-1) from latent infection are unknown. The host transcription and pioneer factor c-Jun is the main target of the JNK cell stress pathway that is known to be important in exit of HSV from latency. Surprisingly, we found that c-Jun does not act with JNK during exit from latency but instead promotes the transition to full reactivation. Moreover, c-Jun and enhanced neuronal stress during initial neuronal infection promoted a more reactivation-competent form of HSV-1 latency. c-Jun, therefore, functions at multiple stages during HSV-1 latent infection of neurons to promote reactivation. Importantly, this study contributes to a growing body of evidence that de novo HSV-1 infection conditions can modulate latent infection and impact future reactivation events, raising important questions on the clinical impact of stress during initial HSV-1 acquisition on future reactivation events and consequences.
Collapse
Affiliation(s)
- Sara A. Dochnal
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Abigail L. Whitford
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Alison K. Francois
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Patryk A. Krakowiak
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Sean Cuddy
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Anna R. Cliffe
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
6
|
Maezawa M, Ochi J, Kubota N, Kamoshida T, Fuji M, Tsukada Y. Herpes Simplex Virus Encephalitis after Recovery from Coronavirus Disease 2019: A Rare Case Report. Intern Med 2023; 62:3515-3518. [PMID: 37779075 PMCID: PMC10749812 DOI: 10.2169/internalmedicine.1790-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/30/2023] [Indexed: 10/03/2023] Open
Abstract
An 85-year-old woman was diagnosed with coronavirus disease 2019 (COVID-19). The patient was treated with dexamethasone, and the infection was cured. She later developed a low-grade fever and fell unconscious. Positivity for herpes simplex virus deoxyribonucleic acid polymerase chain reaction (HSV-DNA PCR) was detected in the cerebrospinal fluid, so she was diagnosed with HSV encephalitis. The patient was treated with antiviral drugs and recovered from the HSV encephalitis. This case suggests that, in patients with COVID-19 and disorders of consciousness, the possibility of HSV encephalitis should be considered along with COVID-19 encephalitis.
Collapse
Affiliation(s)
- Mari Maezawa
- Department of Pulmology, Soka Municipal Hospital, Japan
| | - Junichi Ochi
- Department of Pulmology, Soka Municipal Hospital, Japan
| | | | | | - Mayumi Fuji
- Department of Pulmology, Soka Municipal Hospital, Japan
| | | |
Collapse
|
7
|
Dochnal SA, Whitford AL, Francois AK, Krakowiak PA, Cuddy S, Cliffe AR. c-Jun Signaling During Initial HSV-1 Infection Modulates Latency to Enhance Later Reactivation in addition to Directly Promoting the Progression to Full Reactivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566462. [PMID: 37986840 PMCID: PMC10659354 DOI: 10.1101/2023.11.10.566462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Herpes simplex virus-1 (HSV-1) establishes a latent infection in peripheral neurons and can periodically reactivate to permit transmission and clinical manifestations. Viral transactivators required for lytic infection are largely absent during latent infection and therefore HSV-1 relies on the co-option of neuronal host signaling pathways to initiate its gene expression. Activation of the neuronal c-Jun N-terminal kinase (JNK) cell stress pathway is central to initiating biphasic reactivation in response to multiple stimuli. However, how host factors work with JNK to stimulate the initial wave of gene expression (known as Phase I) or the progression to full, Phase II reactivation remains unclear. Here, we found that c-Jun, the primary target downstream of neuronal JNK cell stress signaling, functions during reactivation but not during the JNK-mediated initiation of Phase I gene expression. Instead, c-Jun was required for the transition from Phase I to full HSV-1 reactivation and was detected in viral replication compartments of reactivating neurons. Interestingly, we also identified a role for both c-Jun and enhanced neuronal stress during initial neuronal infection in promoting a more reactivation-competent form of HSV-1 latency. Therefore, c-Jun functions at multiple stages during HSV latent infection of neurons to promote reactivation. Importantly, by demonstrating that initial infection conditions can contribute to later reactivation abilities, this study highlights the potential for latently infected neurons to maintain a molecular scar of previous exposure to neuronal stressors.
Collapse
Affiliation(s)
- Sara A. Dochnal
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Abigail L. Whitford
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Alison K. Francois
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Patryk A. Krakowiak
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| | - Sean Cuddy
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, 22908
| | - Anna R. Cliffe
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908
| |
Collapse
|
8
|
Stolley JM, Scott MC, Joag V, Dale AJ, Johnston TS, Saavedra F, Gavil NV, Lotfi-Emran S, Soerens AG, Weyu E, Pierson MJ, Herzberg MC, Zhang N, Vezys V, Masopust D. Depleting CD103+ resident memory T cells in vivo reveals immunostimulatory functions in oral mucosa. J Exp Med 2023; 220:e20221853. [PMID: 37097449 PMCID: PMC10130744 DOI: 10.1084/jem.20221853] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/15/2023] [Accepted: 03/31/2023] [Indexed: 04/26/2023] Open
Abstract
The oral mucosa is a frontline for microbial exposure and juxtaposes several unique tissues and mechanical structures. Based on parabiotic surgery of mice receiving systemic viral infections or co-housing with microbially diverse pet shop mice, we report that the oral mucosa harbors CD8+ CD103+ resident memory T cells (TRM), which locally survey tissues without recirculating. Oral antigen re-encounter during the effector phase of immune responses potentiated TRM establishment within tongue, gums, palate, and cheek. Upon reactivation, oral TRM triggered changes in somatosensory and innate immune gene expression. We developed in vivo methods for depleting CD103+ TRM while sparing CD103neg TRM and recirculating cells. This revealed that CD103+ TRM were responsible for inducing local gene expression changes. Oral TRM putatively protected against local viral infection. This study provides methods for generating, assessing, and in vivo depleting oral TRM, documents their distribution throughout the oral mucosa, and provides evidence that TRM confer protection and trigger responses in oral physiology and innate immunity.
Collapse
Affiliation(s)
- J. Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Milcah C. Scott
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Alexander J. Dale
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Timothy S. Johnston
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Flavia Saavedra
- School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Noah V. Gavil
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Sahar Lotfi-Emran
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Andrew G. Soerens
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Eyob Weyu
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Mark J. Pierson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Mark C. Herzberg
- School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
9
|
Obrador E, Salvador-Palmer R, López-Blanch R, Oriol-Caballo M, Moreno-Murciano P, Estrela JM. Survival Mechanisms of Metastatic Melanoma Cells: The Link between Glucocorticoids and the Nrf2-Dependent Antioxidant Defense System. Cells 2023; 12:cells12030418. [PMID: 36766760 PMCID: PMC9913432 DOI: 10.3390/cells12030418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/11/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Circulating glucocorticoids increase during stress. Chronic stress, characterized by a sustained increase in serum levels of cortisol, has been associated in different cases with an increased risk of cancer and a worse prognosis. Glucocorticoids can promote gluconeogenesis, mobilization of amino acids, fat breakdown, and impair the body's immune response. Therefore, conditions that may favor cancer growth and the acquisition of radio- and chemo-resistance. We found that glucocorticoid receptor knockdown diminishes the antioxidant protection of murine B16-F10 (highly metastatic) melanoma cells, thus leading to a drastic decrease in their survival during interaction with the vascular endothelium. The BRAFV600E mutation is the most commonly observed in melanoma patients. Recent studies revealed that VMF/PLX40-32 (vemurafenib, a selective inhibitor of mutant BRAFV600E) increases mitochondrial respiration and reactive oxygen species (ROS) production in BRAFV600E human melanoma cell lines. Early-stage cancer cells lacking Nrf2 generate high ROS levels and exhibit a senescence-like growth arrest. Thus, it is likely that a glucocorticoid receptor antagonist (RU486) could increase the efficacy of BRAF-related therapy in BRAFV600E-mutated melanoma. In fact, during early progression of skin melanoma metastases, RU486 and VMF induced metastases regression. However, treatment at an advanced stage of growth found resistance to RU486 and VMF. This resistance was mechanistically linked to overexpression of proteins of the Bcl-2 family (Bcl-xL and Mcl-1 in different human models). Moreover, melanoma resistance was decreased if AKT and NF-κB signaling pathways were blocked. These findings highlight mechanisms by which metastatic melanoma cells adapt to survive and could help in the development of most effective therapeutic strategies.
Collapse
Affiliation(s)
- Elena Obrador
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
- Correspondence: (E.O.); (J.M.E.); Tel.: +34-963864646 (J.M.E.)
| | - Rosario Salvador-Palmer
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
| | - Rafael López-Blanch
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
| | - María Oriol-Caballo
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
| | | | - José M. Estrela
- Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
- Scientia BioTech S.L., 46002 Valencia, Spain
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
- Correspondence: (E.O.); (J.M.E.); Tel.: +34-963864646 (J.M.E.)
| |
Collapse
|
10
|
Gopinath D, Koe KH, Maharajan MK, Panda S. A Comprehensive Overview of Epidemiology, Pathogenesis and the Management of Herpes Labialis. Viruses 2023; 15:225. [PMID: 36680265 PMCID: PMC9867007 DOI: 10.3390/v15010225] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/03/2023] [Accepted: 01/08/2023] [Indexed: 01/17/2023] Open
Abstract
Herpes labialis remains exceedingly prevalent and is one of the most common human viral infections throughout the world. Recurrent herpes labialis evolves from the initial viral infection by herpes simplex virus type 1 (HSV-1) which subsequently presents with or without symptoms. Reactivation of this virus is triggered by psychosocial factors such as stress, febrile environment, ultraviolet light susceptibility, or specific dietary inadequacy. This virus infection is also characterized by uninterrupted transitions between chronic-latent and acute-recurrent phases, allowing the virus to opportunistically avoid immunity and warrant the transmission to other vulnerable hosts simultaneously. This review comprehensively evaluates the current evidence on epidemiology, pathogenesis, transmission modes, clinical manifestations, and current management options of herpes labialis infections.
Collapse
Affiliation(s)
- Divya Gopinath
- Basic Medical and Dental Sciences Department, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Kim Hoe Koe
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Swagatika Panda
- Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha‘O’Anusandhan Deemed to be University, Bhubaneswar 751030, India
| |
Collapse
|
11
|
Dai J, Wang H, Liao Y, Tan L, Sun Y, Song C, Liu W, Qiu X, Ding C. RNA-seq and LC-MS/MS analysis of antiviral effects mediated by cold stress and stress hormone corticosterone in chicken DF-1 cells. Vet Microbiol 2022; 275:109580. [DOI: 10.1016/j.vetmic.2022.109580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022]
|
12
|
Hasanah NT, Hidayat W. Stress as Trigger Factor of HSV-1 Reactivation Causing Recurrent Intraoral Herpes Mimicking HAEM: A Case Report. Int Med Case Rep J 2022; 15:699-706. [DOI: 10.2147/imcrj.s388708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/09/2022] [Indexed: 12/03/2022] Open
|
13
|
Mousa SA, Dehe L, Aboryag N, Shaqura M, Beyer A, Schäfer M, Treskatsch S. Identification of glucocorticoid receptors as potential modulators of parasympathetic and sympathetic neurons within rat intracardiac ganglia. Front Neuroanat 2022; 16:902738. [PMID: 36213610 PMCID: PMC9539283 DOI: 10.3389/fnana.2022.902738] [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: 03/23/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Background Emerging evidences indicate that glucocorticoid receptors (GR) play a regulatory role in cardiac function, particularly with regard to the autonomic nervous system. Therefore, this study aimed to demonstrate the expression and the precise anatomical location of GR in relation to the parasympathetic and sympathetic innervations of the heart. Methods The present study used tissue samples from rat heart atria to perform conventional reverse-transcriptase polymerase chain reaction (RT-PCR), Western blot, and double immunofluorescence confocal analysis of GR with the neuronal markers vesicular acetylcholine transporter (VAChT), tyrosine hydroxylase (TH), calcitonin gene-related peptide (CGRP) as well as the mineralocorticoid receptor (MR). Results Double immunofluorescence labeling revealed that GRs were co-expressed with VAChT in parasympathetic principal neuronal somata and nerve terminals innervating atrium. Also, GR colocalized with the sympathetic neuronal marker TH in a cluster of small intensely fluorescent (SIF) cells, on intracardiac nerve terminals and in the atrial myocardium. GR immunoreactivity was scarcely identified on CGRP-immunoreactive sensory nerve terminals. Approximately 20% of GR immunoreactive neuronal somata co-localized with MR. Finally, conventional RT-PCR and Western blot confirmed the presence of GR and MR in rat heart atria. Conclusion This study provides evidence for the existence of GR predominantly on cardiac parasympathetic neurons and TH-immunoreactive SIF cells suggesting a functional role of cardiac GR on cardiovascular function by modulation of the cardiac autonomic nervous system.
Collapse
Affiliation(s)
- Shaaban A. Mousa
- Department of Anaesthesiology and Intensive Care Medicine, Charité – University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Lukas Dehe
- Department of Anaesthesiology and Intensive Care Medicine, Charité – University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Noureddin Aboryag
- Department of Anaesthesiology and Intensive Care Medicine, Charité – University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Mohammed Shaqura
- Department of Anaesthesiology and Intensive Care Medicine, Charité – University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Antje Beyer
- Department of Anaesthesiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Schäfer
- Department of Anaesthesiology and Intensive Care Medicine, Charité – University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Sascha Treskatsch
- Department of Anaesthesiology and Intensive Care Medicine, Charité – University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
14
|
Cairns BE. The contribution of autonomic mechanisms to pain in temporomandibular disorders: A narrative review. J Oral Rehabil 2022; 49:1115-1126. [PMID: 36098708 DOI: 10.1111/joor.13370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/22/2022] [Accepted: 09/06/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Temporomandibular disorders (TMD) are diagnosed based on symptom presentation and, like other functional pain disorders, often lack definitive pathology. There is a strong association between elevated stress levels and the severity of TMD-related pain, which suggests that alterations in autonomic tone may contribute to this pain condition. OBJECTIVES This narrative review examines the association between altered autonomic function and pain in TMD. METHODS Relevant articles were identified by searching PubMed and through the reference list of those studies. RESULTS TMD sufferers report an increased incidence of orthostatic hypotension. As in other chronic musculoskeletal pain conditions, TMD is associated with increased sympathetic tone, diminished baroreceptor reflex sensitivity and decreased parasympathetic tone. It remains to be determined whether ongoing pain drives these autonomic changes and/or is exacerbated by them. To examine whether increased sympathetic tone contributes to TMD-related pain through β2 adrenergic receptor activation, clinical trials with the beta blocker propranolol have been undertaken. Although evidence from small studies suggested propranolol reduced TMD-related pain, a larger clinical trial did not find a significant effect of propranolol treatment. This is consistent with human experimental pain studies that were unable to demonstrate an effect of β2 adrenergic receptor activation or inhibition on masticatory muscle pain. In preclinical models of temporomandibular joint arthritis, β2 adrenergic receptor activation appears to contribute to inflammation and nociception, whereas in masticatory muscle, α1 adrenergic receptor activation has been found to induce mechanical sensitization. Some agents used to treat TMD, such as botulinum neurotoxin A, antidepressants and α2 adrenergic receptor agonists, may interact with the autonomic nervous system as part of their analgesic mechanism. CONCLUSION Even if dysautonomia turns out to be a consequence rather than a causative factor of painful TMD, the study of its role has opened up a greater understanding of the pathogenesis of this condition.
Collapse
Affiliation(s)
- Brian E Cairns
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
15
|
Hakim DDL, Gurnida DA, Nuraeny N, Susilaningsih FS, Herawati DMD. Serological Evidence of Herpes Simplex Virus-1 (HSV-1) Infection among Humans from Bandung, West Java Province, Indonesia. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.10183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND: Toxoplasma gondii, Rubella virus, Cytomegalovirus, and herpes simplex virus (TORCH) infection is still a significant burden in developing countries since they potentially increase perinatal death and decrease life quality by causing congenital disorders. As part of TORCH and as one of the most common infections in humans, HSV Type 1 infection also should receive attention. HSV-1 infection induces an immediate reactive oxygen species (ROS) production, indicate that ROS plays beneficial effects in several biological functions, including innate immunity and antiviral responses. HSV-1 preferentially replicate and establish latency in different subtypes of sensory neurons and in neurons of the autonomic nervous system that are highly responsive to stress hormones, including cortisol.
AIM: The objective of the study was to detect the latent HSV-1 infection in adults population and its effect on ROS and cortisol levels.
PATIENTS AND METHODS: Subjects were enrolled with consecutive-sampling methods among the adults population age 18–40 years old, with no health complaints. We collected their blood to examined IgG HSV-1, ROS, and cortisol levels.
RESULTS: A total of 57 subjects with 27 subjects were reactive IgG HSV-1 (herpes group) and 30 subjects were non-reactive IgG HSV-1 (non herpes groups). Mean of cortisol and ROS was 223.2904 nmol/L and 2.23337 IU/mL, respectively. There was a very weak correlation between HSV-1 infection with ROS and cortisol.
CONCLUSION: There is a positive effect of latent HSV-1 infection in the adult population on cortisol ROS levels.
Collapse
|
16
|
Progressive Course of Chronic Tick-Borne Encephalitis Manifesting as Amyotrophic Lateral Sclerosis-like Syndrome 35 Years after the Acute Disease. Brain Sci 2022; 12:brainsci12081020. [PMID: 36009082 PMCID: PMC9405949 DOI: 10.3390/brainsci12081020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
The chronic form of tick-borne encephalitis (TBE) is understudied and seems to be linked exclusively to Siberian and Far Eastern TBE virus (TBEV) subtypes. There are limited clinical descriptions demonstrating that chronic TBE can resemble an amyotrophic lateral sclerosis (ALS)-like disease. Here, we present a description of a clinical case of progressive chronic TBEV infection with a relapse 35 years after the initial acute infection following a tick bite. The disease manifested as an ALS-like syndrome with bulbar signs, progressive muscle weakness and atrophy, decreased reflexes, and eventual respiratory failure and death. There is no clear differentiation between chronic TBE and postencephalitic syndrome described in European sources. The reactivation of TBEV infection was supported by the presence of anti-TBEV antibodies in serum and antibodies to E protein and to the nonstructural protein NS1 in the CSF. These findings support the diagnosis of a relapse of chronic TBE in this patient.
Collapse
|
17
|
Impact of Cultured Neuron Models on α-Herpesvirus Latency Research. Viruses 2022; 14:v14061209. [PMID: 35746680 PMCID: PMC9228292 DOI: 10.3390/v14061209] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
A signature trait of neurotropic α-herpesviruses (α-HV) is their ability to establish stable non-productive infections of peripheral neurons termed latency. This specialized gene expression program is the foundation of an evolutionarily successful strategy to ensure lifelong persistence in the host. Various physiological stresses can induce reactivation in a subset of latently-infected neurons allowing a new cycle of viral productive cycle gene expression and synthesis of infectious virus. Recurring reactivation events ensure transmission of the virus to new hosts and contributes to pathogenesis. Efforts to define the molecular basis of α-HV latency and reactivation have been notoriously difficult because the neurons harboring latent virus in humans and in experimentally infected live-animal models, are rare and largely inaccessible to study. Increasingly, researchers are turning to cultured neuron infection models as simpler experimental platforms from which to explore latency and reactivation at the molecular level. In this review, I reflect on the strengths and weaknesses of existing neuronal models and briefly summarize the important mechanistic insights these models have provided. I also discuss areas where prioritization will help to ensure continued progress and integration.
Collapse
|
18
|
Goswami P, Ives AM, Abbott ARN, Bertke AS. Stress Hormones Epinephrine and Corticosterone Selectively Reactivate HSV-1 and HSV-2 in Sympathetic and Sensory Neurons. Viruses 2022; 14:1115. [PMID: 35632856 PMCID: PMC9147053 DOI: 10.3390/v14051115] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/16/2022] Open
Abstract
Herpes simplex viruses 1 and 2 (HSV-1 and HSV-2) establish latency in sensory and autonomic neurons, from which they can reactivate to cause recurrent disease throughout the life of the host. Stress is strongly associated with HSV recurrences in humans and animal models. However, the mechanisms through which stress hormones act on the latent virus to cause reactivation are unknown. We show that the stress hormones epinephrine (EPI) and corticosterone (CORT) induce HSV-1 reactivation selectively in sympathetic neurons, but not sensory or parasympathetic neurons. Activation of multiple adrenergic receptors is necessary for EPI-induced HSV-1 reactivation, while CORT requires the glucocorticoid receptor. In contrast, CORT, but not EPI, induces HSV-2 reactivation in both sensory and sympathetic neurons through either glucocorticoid or mineralocorticoid receptors. Reactivation is dependent on different transcription factors for EPI and CORT, and coincides with rapid changes in viral gene expression, although genes differ for HSV-1 and HSV-2, and temporal kinetics differ for EPI and CORT. Thus, stress-induced reactivation mechanisms are neuron-specific, stimulus-specific and virus-specific. These findings have implications for differences in HSV-1 and HSV-2 recurrent disease patterns and frequencies, as well as development of targeted, more effective antivirals that may act on different responses in different types of neurons.
Collapse
Affiliation(s)
- Poorna Goswami
- Translational Biology Medicine and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
| | - Angela M. Ives
- Biomedical and Veterinary Science, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
| | - Amber R. N. Abbott
- Department of Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
| | - Andrea S. Bertke
- Population Health Sciences, Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| |
Collapse
|
19
|
Begazo L, Morante A, Espinoza Montes C. [Herpes simplex factors: Literature review]. REVISTA CIENTÍFICA ODONTOLÓGICA 2022; 10:e099. [PMID: 38389913 PMCID: PMC10880724 DOI: 10.21142/2523-2754-1001-2022-099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/30/2022] [Indexed: 02/24/2024] Open
Abstract
Herpes simplex virus, after the first infection, can remain dormant in the lymph nodes and reactivate at any time due to certain factors and/or stimuli causing effects in the patient. The purpose of this literature review is to update scientific information on the factors that predispose to the reactivation of herpes simplex virus. A comprehensive literature review was conducted in the LILACS, SciELO, PubMed, Scopus and the following keywords were used: herpes simplex, reactivation, latency and risks. A factor is reactivation by immunosuppression due to the intake of immunosuppressants or oncological therapy. Another factor is psychological stress, in which the hormones released act on mediating signs of reactivation. Sun exposure that reactivates the virus in infected neurons and infection in pregnant women can trigger complications in the fetus and delivery.
Collapse
Affiliation(s)
- Lisette Begazo
- Carrera de Estomatología, Facultad de Ciencias de la Vida y Salud. Universidad Científica del Sur, Lima, Perú. , , Universidad Científica del Sur Carrera de Estomatología Facultad de Ciencias de la Vida y Salud Universidad Científica del Sur Lima Peru
| | - Alysson Morante
- Carrera de Estomatología, Facultad de Ciencias de la Vida y Salud. Universidad Científica del Sur, Lima, Perú. , , Universidad Científica del Sur Carrera de Estomatología Facultad de Ciencias de la Vida y Salud Universidad Científica del Sur Lima Peru
| | - Carlos Espinoza Montes
- Carrera de Estomatología, Facultad de Ciencias de la Vida y Salud. Universidad Científica del Sur, Lima, Perú. , , Universidad Científica del Sur Carrera de Estomatología Facultad de Ciencias de la Vida y Salud Universidad Científica del Sur Lima Peru
| |
Collapse
|
20
|
Shanshal M, Ahmed HS. COVID-19 and Herpes Simplex Virus Infection: A Cross-Sectional Study. Cureus 2021; 13:e18022. [PMID: 34667693 PMCID: PMC8520410 DOI: 10.7759/cureus.18022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2021] [Indexed: 01/22/2023] Open
Abstract
Background Despite being variable and poorly characterized, the reported cutaneous manifestations of coronavirus disease 2019 (COVID-19) are of increasing concern. Methodology This study aimed to determine the prevalence and possible association between COVID-19 and herpes simplex virus (HSV) infection. A nine-item questionnaire was sent to 120 polymerase chain reaction-confirmed COVID-19 patients with a response rate of 66.67%. This cross-sectional observational study included 80 patients with mild-to-moderate COVID-19 infection who did not require hospitalization or steroid therapy. Results One or more HSV infections were observed in 28 patients (35%) with COVID-19 infection, including 10 (35.7%) males and 18 (64.29%) females. Of the 28 patients, fever was reported in 17 (75%) during COVID-19. Most of the respondents (78%) described a single HSV reactivation, 14.29% had two attacks, and 7.14% experienced three attacks. Compared to previous non-COVID-19-related HSV reactivation, the COVID-19-related attacks were more severe in 12 (42.85%) patients, equally severe in five (17.85%) patients, and less severe in one (3.57%) patient. Interestingly, 10 (35.71%) patients developed an initial symptomatic HSV attack during COVID-19 infection. Conclusions This study demonstrated a possible association between COVID-19 infection and primary HSV infection or reactivation. COVID-19 direct neuronal effect in addition to COVID-19-related psychological stress, fever, and immunological dysregulation could play a potential role in HSV reactivation or primary infection during COVID-19.
Collapse
Affiliation(s)
| | - Hayder Saad Ahmed
- Department of Dermatology and Venereology, University of Tikrit, College of Medicine, Tikrit, IRQ
| |
Collapse
|
21
|
Suzich JB, Cuddy SR, Baidas H, Dochnal S, Ke E, Schinlever AR, Babnis A, Boutell C, Cliffe AR. PML-NB-dependent type I interferon memory results in a restricted form of HSV latency. EMBO Rep 2021; 22:e52547. [PMID: 34197022 PMCID: PMC8419685 DOI: 10.15252/embr.202152547] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 01/23/2023] Open
Abstract
Herpes simplex virus (HSV) establishes latent infection in long-lived neurons. During initial infection, neurons are exposed to multiple inflammatory cytokines but the effects of immune signaling on the nature of HSV latency are unknown. We show that initial infection of primary murine neurons in the presence of type I interferon (IFN) results in a form of latency that is restricted for reactivation. We also find that the subnuclear condensates, promyelocytic leukemia nuclear bodies (PML-NBs), are absent from primary sympathetic and sensory neurons but form with type I IFN treatment and persist even when IFN signaling resolves. HSV-1 genomes colocalize with PML-NBs throughout a latent infection of neurons only when type I IFN is present during initial infection. Depletion of PML prior to or following infection does not impact the establishment latency; however, it does rescue the ability of HSV to reactivate from IFN-treated neurons. This study demonstrates that viral genomes possess a memory of the IFN response during de novo infection, which results in differential subnuclear positioning and ultimately restricts the ability of genomes to reactivate.
Collapse
Affiliation(s)
- Jon B Suzich
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Sean R Cuddy
- Neuroscience Graduate ProgramUniversity of VirginiaCharlottesvilleVAUSA
| | - Hiam Baidas
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Sara Dochnal
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Eugene Ke
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Austin R Schinlever
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Aleksandra Babnis
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| | - Chris Boutell
- MRC‐University of Glasgow Centre for Virus Research (CVR)GlasgowUK
| | - Anna R Cliffe
- Department of Microbiology, Immunology and Cancer BiologyUniversity of VirginiaCharlottesvilleVAUSA
| |
Collapse
|
22
|
Passeron T, Zouboulis CC, Tan J, Andersen ML, Katta R, Lyu X, Aguilar L, Kerob D, Morita A, Krutmann J, Peters EMJ. Adult skin acute stress responses to short-term environmental and internal aggression from exposome factors. J Eur Acad Dermatol Venereol 2021; 35:1963-1975. [PMID: 34077579 PMCID: PMC8519049 DOI: 10.1111/jdv.17432] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 12/02/2022]
Abstract
Exposome factors that lead to stressed skin can be defined as any disturbance to homeostasis from environmental (meteorological factors, solar radiation, pollution or tobacco smoke) and/or internal exposure (unhealthy diet, hormonal variations, lack of sleep, psychosocial stress). The clinical and biological impact of chronic exposome effects on skin functions has been extensively reviewed, whereas there is a paucity of information on the impact of short‐term acute exposure. Acute stress, which would typically last minutes to hours (and generally no more than a week), provokes a transient but robust neuroendocrine‐immune and tissue remodelling response in the skin and can alter the skin barrier. Firstly, we provide an overview of the biological effects of various acute stressors on six key skin functions, namely the skin physical barrier, pigmentation, defences (antioxidant, immune cell‐mediated, microbial and microbiome maintenance), structure (extracellular matrix and appendages), neuroendocrine and thermoregulation functions. Secondly, we describe the biological and clinical effects on adult skin from individual exposome factors that elicit an acute stress response and their consequences in skin health maintenance. Clinical manifestations of acutely stressed skin may include dry skin that might accentuate fine lines, oily skin, sensitive skin, pruritus, erythema, pale skin, sweating, oedema and flares of inflammatory skin conditions such as acne, rosacea, atopic dermatitis, pigmentation disorders and skin superinfection such as viral reactivation. Acute stresses can also induce scalp sensitivity, telogen effluvium and worsen alopecia.
Collapse
Affiliation(s)
- T Passeron
- Department of Dermatology, University Hospital Centre Nice, Côte d'Azur University, Nice, France.,INSERM U1065, team 12, C3M, Nice, France
| | - C C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane and Faculty of Health Sciences Brandenburg, Dessau, Germany
| | - J Tan
- Windsor Clinical Research Inc., Windsor, ON, Canada.,Department of Medicine, University of Western Ontario, London, Canada
| | - M L Andersen
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP)/Escola Paulista de Medicina, São Paulo, Brazil
| | - R Katta
- Volunteer Clinical Faculty, Baylor College of Medicine, Houston, Texas, USA.,McGovern Medical School at UT Health, Houston, Texas, USA
| | - X Lyu
- Department of Dermatology, Anzhen Hospital, Capital Medical University, Beijing, China
| | - L Aguilar
- L'Oréal Advanced Research, Aulnay-sous-bois, France
| | - D Kerob
- Laboratoires Vichy, Levallois Perret, France
| | - A Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - J Krutmann
- IUF Leibniz Research Institute for Environmental Medicine, Dusseldorf, Germany.,Medical faculty, Heinrich-Heine-University, Dusseldorf, Germany
| | - E M J Peters
- Department of Psychosomatic Medicine and Psychotherapy, Justus-Liebig University, Gießen, Germany.,Charité Center 12 (CC12) for Internal Medicine and Dermatology, Berlin, Germany
| |
Collapse
|
23
|
McDonald JA, Cherubin S, Goldberg M, Wei Y, Chung WK, Schwartz LA, Knight JA, Schooling CM, Santella RM, Bradbury AR, Buys SS, Andrulis IL, John EM, Daly MB, Terry MB. Common Childhood Viruses and Pubertal Timing: The LEGACY Girls Study. Am J Epidemiol 2021; 190:766-778. [PMID: 33128063 DOI: 10.1093/aje/kwaa240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/13/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Earlier pubertal development is only partially explained by childhood body mass index; the role of other factors, such as childhood infections, is less understood. Using data from the LEGACY Girls Study (North America, 2011-2016), we prospectively examined the associations between childhood viral infections (cytomegalovirus (CMV), Epstein-Barr virus (EBV), herpes simplex virus (HSV) 1, HSV2) and pubertal timing. We measured exposures based on seropositivity in premenarcheal girls (n = 490). Breast and pubic hair development were classified based on mother-reported Tanner Stage (TS) (TS2+ compared with TS1), adjusting for age, body mass index, and sociodemographic factors. The average age at first blood draw was 9.8 years (standard deviation, 1.9 years). The prevalences were 31% CMV+, 37% EBV+, 14% HSV1+, 0.4% HSV2+, and 16% for both CMV+/EBV+ coinfection. CMV+ infection without coinfection was associated with developing breasts an average of 7 months earlier (hazard ratio (HR) = 2.12, 95% confidence interval (CI): 1.32, 3.40). CMV infection without coinfection and HSV1 and/or HSV2 infection were associated with developing pubic hair 9 months later (HR = 0.41, 95% CI: 0.24, 0.71, and HR = 0.42, 95% CI: 0.22, 0.81, respectively). Infection was not associated with menarche. If replicated in larger cohorts with blood collection prior to any breast development, this study supports the hypothesis that childhood infections might play a role in altering pubertal timing.
Collapse
|
24
|
Colón-Thillet R, Jerome KR, Stone D. Optimization of AAV vectors to target persistent viral reservoirs. Virol J 2021; 18:85. [PMID: 33892762 PMCID: PMC8067653 DOI: 10.1186/s12985-021-01555-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
Gene delivery of antiviral therapeutics to anatomical sites where viruses accumulate and persist is a promising approach for the next generation of antiviral therapies. Recombinant adeno-associated viruses (AAV) are one of the leading vectors for gene therapy applications that deliver gene-editing enzymes, antibodies, and RNA interference molecules to eliminate viral reservoirs that fuel persistent infections. As long-lived viral DNA within specific cellular reservoirs is responsible for persistent hepatitis B virus, Herpes simplex virus, and human immunodeficiency virus infections, the discovery of AAV vectors with strong tropism for hepatocytes, sensory neurons and T cells, respectively, is of particular interest. Identification of natural isolates from various tissues in humans and non-human primates has generated an extensive catalog of AAV vectors with diverse tropisms and transduction efficiencies, which has been further expanded through molecular genetic approaches. The AAV capsid protein, which forms the virions' outer shell, is the primary determinant of tissue tropism, transduction efficiency, and immunogenicity. Thus, over the past few decades, extensive efforts to optimize AAV vectors for gene therapy applications have focused on capsid engineering with approaches such as directed evolution and rational design. These approaches are being used to identify variants with improved transduction efficiencies, alternate tropisms, reduced sequestration in non-target organs, and reduced immunogenicity, and have produced AAV capsids that are currently under evaluation in pre-clinical and clinical trials. This review will summarize the most recent strategies to identify AAV vectors with enhanced tropism and transduction in cell types that harbor viral reservoirs.
Collapse
Affiliation(s)
- Rossana Colón-Thillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
| | - Keith R Jerome
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Daniel Stone
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA.
| |
Collapse
|
25
|
Newcastle Disease Virus Induced Pathologies Severely Affect the Exocrine and Endocrine Functions of the Pancreas in Chickens. Genes (Basel) 2021; 12:genes12040495. [PMID: 33805275 PMCID: PMC8067305 DOI: 10.3390/genes12040495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 01/05/2023] Open
Abstract
Newcastle disease virus (NDV) causes a highly contagious and devastating disease in poultry. ND causes heavy economic losses to the global poultry industry by decreasing the growth rate, decrease in egg production high morbidity and mortality. Although significant advances have been made in the vaccine development, outbreaks are reported in vaccinated birds. In this study, we report the damage caused by NDV infection in the pancreatic tissues of vaccinated and specific-pathogen-free chickens. The histopathological examination of the pancreas showed severe damage in the form of partial depletion of zymogen granules, acinar cell vacuolization, necrosis, apoptosis, congestion in the large and small vessels, sloughing of epithelial cells of the pancreatic duct, and mild perivascular edema. Increased plasma levels of corticosterone and somatostatin were observed in NDV-infected chicken at three- and five- days post infection (DPI). A slight decrease in the plasma concentrations of insulin was noticed at 5 DPI. Significant changes were not observed in the plasma levels of glucagon. Furthermore, NDV infection decreased the activity and mRNA expression of amylase, lipase, and trypsin from the pancreas. Taken together, our findings highlight that NDV induces extensive tissue damage in the pancreas, decreases the activity and expression of pancreatic enzymes, and increases plasma corticosterone and somatostatin. These findings provide new insights that a defective pancreas may be one of the reasons for decreased growth performance after NDV infection in chickens.
Collapse
|
26
|
HSV-pneumonitis in a patient with lung cancer receiving check point inhibitors - a case report. Pneumonia (Nathan) 2021; 13:1. [PMID: 33487176 PMCID: PMC7830804 DOI: 10.1186/s41479-020-00079-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/15/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Herpes simplex virus (HSV) is commonly associated with oro-facial and genital manifestations. It rarely causes encephalitis and even less commonly, in heavily immunosuppressed patients, visceral disease or bronchopneumonitis. We present a case of cytologically-proven, PCR-positive HSV-1 tracheobronchitis and pneumonitis in a patient with less severe immunocompromise. CASE PRESENTATION A 64 year old white man with steroid-induced diabetes mellitus and progressive small-cell bronchial carcinoma despite chemo- and immunotherapy with two checkpoint inhibitors presented with symptoms of lower respiratory tract infection. Community-acquired pneumonia was suspected and empirical broad-spectrum antibacterial treatment was initiated. Chest CT-scan revealed ground-glass opacities and tree-in bud lesions. Cytology of BAL showed extensive cytopathic effects typically caused by infection with herpes virus and PCR confirmation of HSV-1. Acute phase HSV serology was positive for IgG and borderline for IgM. The patient deteriorated clinically due to tumor progress and infection despite high-dose acyclovir therapy and died 2 weeks after admission. CONCLUSIONS We report an unusual case of fatal HSV-1 pneumonitis due to reactivation in a patient with lung cancer, steroid-induced diabetes and treatment with two checkpoint inhibitors. In immunosuppressed patients with non-improving pneumonia invasive diagnostic procedures are warranted including cytology and molecular diagnostics.
Collapse
|
27
|
Liu ZH, Niu FJ, Xie YX, Xie SM, Liu YN, Yang YY, Zhou CZ, Wan XH. A review: Natural polysaccharides from medicinal plants and microorganisms and their anti-herpetic mechanism. Biomed Pharmacother 2020; 129:110469. [DOI: 10.1016/j.biopha.2020.110469] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
|
28
|
Powell-Doherty RD, Abbott ARN, Nelson LA, Bertke AS. Amyloid-β and p-Tau Anti-Threat Response to Herpes Simplex Virus 1 Infection in Primary Adult Murine Hippocampal Neurons. J Virol 2020; 94:e01874-19. [PMID: 32075924 PMCID: PMC7163132 DOI: 10.1128/jvi.01874-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's Disease (AD) is the sixth leading cause of death in the United States. Recent studies have established a potential link between herpes simplex virus 1 (HSV-1) infection and the development of AD. HSV-1 DNA has been detected in AD amyloid plaques in human brains, and treatment with the antiviral acyclovir (ACV) was reported to block the accumulation of the AD-associated proteins beta-amyloid (Aβ) and hyper-phosphorylated tau (p-tau) in Vero and glioblastoma cells. Our goal was to determine whether the accumulation of AD-related proteins is attributable to acute and/or latent HSV-1 infection in mature hippocampal neurons, a region of the brain severely impacted by AD. Primary adult murine hippocampal neuronal cultures infected with HSV-1, with or without antivirals, were assessed for Aβ and p-tau expression over 7 days postinfection. P-tau expression was transiently elevated in HSV-1-infected neurons, as well as in the presence of antivirals alone. Infected neurons, as well as uninfected neurons treated with antivirals, had a greater accumulation of Aβ42 than uninfected untreated neurons. Furthermore, Aβ42 colocalized with HSV-1 latency-associated transcript (LAT) expression. These studies suggest that p-tau potentially acts as an acute response to any perceived danger-associated molecular pattern (DAMP) in primary adult hippocampal neurons, while Aβ aggregation is a long-term response to persistent threats, including HSV-1 infection.IMPORTANCE Growing evidence supports a link between HSV-1 infection and Alzheimer's disease (AD). Although AD is clearly a complex multifactorial disorder, an infectious disease etiology provides alternative therapy opportunities for this devastating disease. Understanding the impact that HSV-1 has on mature neurons and the proteins most strongly associated with AD pathology may identify specific mechanisms that could be manipulated to prevent progression of neurodegeneration and dementia.
Collapse
Affiliation(s)
- Rebecca D Powell-Doherty
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Amber R N Abbott
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Laura A Nelson
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Andrea S Bertke
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
29
|
Davidson I, Altory-Natour A, Haddas R, Nagar S, Meir R, Avital-Cohen N, Rozenboim I. Evaluation of Viral-Induced Stress by Quantitating Corticosterone in Feathers of Virus-Infected Specific Pathogen-Free Chicks. J APPL POULTRY RES 2020. [DOI: 10.3382/japr/pfz027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
30
|
Disturbed Yin-Yang balance: stress increases the susceptibility to primary and recurrent infections of herpes simplex virus type 1. Acta Pharm Sin B 2020; 10:383-398. [PMID: 32140387 PMCID: PMC7049575 DOI: 10.1016/j.apsb.2019.06.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/27/2019] [Accepted: 05/31/2019] [Indexed: 12/19/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1), a neurotropic herpes virus, is able to establish a lifelong latent infection in the human host. Following primary replication in mucosal epithelial cells, the virus can enter sensory neurons innervating peripheral tissues via nerve termini. The viral genome is then transported to the nucleus where it can be maintained without producing infectious progeny, and thus latency is established in the cell. Yin–Yang balance is an essential concept in traditional Chinese medicine (TCM) theory. Yin represents stable and inhibitory factors, and Yang represents the active and aggressive factors. When the organism is exposed to stress, especially psychological stress caused by emotional stimulation, the Yin–Yang balance is disturbed and the virus can re-engage in productive replication, resulting in recurrent diseases. Therefore, a better understanding of the stress-induced susceptibility to HSV-1 primary infection and reactivation is needed and will provide helpful insights into the effective control and treatment of HSV-1. Here we reviewed the recent advances in the studies of HSV-1 susceptibility, latency and reactivation. We included mechanisms involved in primary infection and the regulation of latency and described how stress-induced changes increase the susceptibility to primary and recurrent infections.
Collapse
Key Words
- 4E-BP, eIF4E-binding protein
- AD, Alzheimer's disease
- AKT, protein kinase B
- AMPK, AMP-dependent kinase
- BCL-2, B-cell lymphoma 2
- CNS, central nervous system
- CORT, corticosterone
- CPE, cytopathic effect
- CTCF, CCCTC-binding factor
- CTL, cytotoxic T lymphocyte
- CoREST, REST corepressor 1
- DAMPs, damage-associated molecular patterns
- DCs, dendritic cells
- DEX, dexamethasone
- GREs, GR response elements
- GRs, glucocorticoid receptors
- H3K9, histone H3 on lysines 9
- HCF-1, host cell factor 1
- HDACs, histone deacetylases
- HPA axis, hypothalamo–pituitary–adrenal axis
- HPK, herpetic simplex keratitis
- HPT axis, hypothalamic–pituitary–thyroid axis
- HSV-1
- HSV-1, herpes simplex virus type 1
- Herpes simplex virus type 1
- ICP, infected cell polypeptide
- IRF3, interferon regulatory factor 3
- KLF15, Krüppel-like transcription factor 15
- LAT, latency-associated transcripts
- LRF, Luman/CREB3 recruitment factor
- LSD1, lysine-specific demethylase 1
- Latency
- MAVS, mitochondrial antiviral-signaling protein
- MOI, multiplicity of infection
- ND10, nuclear domains 10
- NGF, nerve growth factor
- NK cells, natural killer cells
- OCT-1, octamer binding protein 1
- ORFs, open reading frames
- PAMPs, pathogen-associated molecular patterns
- PDK1, pyruvate dehydrogenase lipoamide kinase isozyme 1
- PI3K, phosphoinositide 3-kinases
- PML, promyelocytic leukemia protein
- PNS, peripheral nervous system
- PRC1, protein regulator of cytokinesis 1
- PRRs, pattern-recognition receptors
- PTMs, post-translational modifications
- RANKL, receptor activator of NF-κB ligands
- REST, RE1-silencing transcription factor
- ROS, reactive oxygen species
- Reactivation
- SGKs, serum and glucocorticoid-regulated protein kinases
- SIRT1, sirtuin 1
- Stress
- Susceptibility
- T3, thyroid hormone
- TCM, traditional Chinese medicine
- TG, trigeminal ganglia
- TK, thymidine kinase
- TRIM14, tripartite motif-containing 14
- TRKA, tropomyosin receptor kinase A
- TRM, tissue resident memory T cells
- cGAS, cyclic GMP-AMP synthase
- mTOR, mammalian target of rapamycin
- sncRNAs, small non-coding RNAs
Collapse
|
31
|
Herpes Simplex Virus 2 in Autonomic Ganglia: Evidence for Spontaneous Reactivation. J Virol 2019; 93:JVI.00227-19. [PMID: 30894469 DOI: 10.1128/jvi.00227-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 01/09/2023] Open
Abstract
Herpes simplex virus 2 (HSV-2) can be transmitted in the presence or absence of lesions, allowing efficient spread among the general population. Recurrent HSV genital lesions are thought to arise from reactivated latent virus in sensory cell bodies of the dorsal root ganglia (DRG). However, HSV-2 has also been found latent in autonomic ganglia. Spontaneous reactivation or a low level of chronic infection could theoretically also occur in these peripheral nervous tissues, contributing to the presence of infectious virus in the periphery and to viral transmission. Use of a recently described, optimized virus with a monomeric mNeonGreen protein fused to viral capsid protein 26 (VP26) permitted detection of reactivating virus in explanted ganglia and cryosections of DRG and the sacral sympathetic ganglia (SSG) from latently infected guinea pigs. Immediate early, early, and late gene expression were quantified by droplet digital reverse transcription-PCR (ddRT-PCR), providing further evidence of viral reactivation not only in the expected DRG but also in the sympathetic SSG. These findings indicate that viral reactivation from autonomic ganglia is a feature of latent viral infection and that these reactivations likely contribute to viral pathogenesis.IMPORTANCE HSV-2 is a ubiquitous important human pathogen that causes recurrent infections for the life of its host. We hypothesized that the autonomic ganglia have important roles in viral reactivation, and this study sought to determine whether this is correct in the clinically relevant guinea pig vaginal infection model. Our findings indicate that sympathetic ganglia are sources of reactivating virus, helping explain how the virus causes lifelong recurrent disease.
Collapse
|
32
|
Berra LV, Armocida D, Pesce A, Di Rita A, Santoro A. Herpes Simplex Reactivation After Surgical Treatment of Trigeminal Neuralgia: A Retrospective Cohort Study. World Neurosurg 2019; 127:e16-e21. [PMID: 30771541 DOI: 10.1016/j.wneu.2019.01.226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Herpes simplex virus (HSV) reactivation after surgery for trigeminal neuralgia has long been recognized. Only a few studies to date have focused on this complication, and its actual incidence remains unknown. The aim of this study was to investigate the incidence of postoperative herpes labialis (HL) in a cohort of patients treated with either percutaneous balloon compression or microvascular decompression to identify potentially significant differences between different treatments. METHODS A total of 92 patients who were operated on for TN with microvascular decompression (group A) or percutaneous balloon compression (group B) in the period 2010-2017 were retrospectively evaluated. The 2 subgroups of patients were compared according to history of previous HL and incidence of postoperative HL. RESULTS The final cohort comprised 56 male and 36 female patients. Average age was 58.50 years; 30 male patients belonged to group A and 26 male patients belonged to group B. Lifetime incidence of episodes of HL before surgery in 18/58 patients in group A (31.0%) and 12/34 patients in group B (35.3%), with no statistically significant difference among subgroups. Postoperatively, 1/56 patients in group A (1.7%) experienced HL compared 5/34 patients in group B (14.7%), with a strongly statistically significant difference between the 2 subgroups. CONCLUSIONS In our clinical experience, herpes simplex virus reactivation after surgery for trigeminal neuralgia is not so rare and is still not completely understood. Postoperative herpes simplex virus reactivation could be due to a direct mechanical injury on gasserian ganglion neurons, which is more common after percutaneous balloon compression.
Collapse
Affiliation(s)
- Luigi Valentino Berra
- Neurosurgery Division, Human Neurosciences Department, Sapienza University of Rome, Rome, Italy
| | - Daniele Armocida
- Neurosurgery Division, Human Neurosciences Department, Sapienza University of Rome, Rome, Italy.
| | - Alessandro Pesce
- Neurosurgery Division, NESMOS Department, Sapienza University of Rome, Rome, Italy; Azienda Universitario-Ospedaliera Sant'Andrea, Rome, Italy
| | - Andrea Di Rita
- Neurosurgery Division, San Carlo Borromeo Hospital, Milan, Italy
| | - Antonio Santoro
- Neurosurgery Division, Human Neurosciences Department, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Li W, Wang XH, Luo Z, Liu LF, Yan C, Yan CY, Chen GD, Gao H, Duan WJ, Kurihara H, Li YF, He RR. Traditional Chinese Medicine as a Potential Source for HSV-1 Therapy by Acting on Virus or the Susceptibility of Host. Int J Mol Sci 2018; 19:ijms19103266. [PMID: 30347851 PMCID: PMC6213986 DOI: 10.3390/ijms19103266] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is the most common virus, with an estimated infection rate of 60–95% among the adult population. Once infected, HSV-1 can remain latent in the host for a lifetime and be reactivated in patients with a compromised immune system. Reactivation of latent HSV-1 can also be achieved by other stimuli. Though acyclovir (ACV) is a classic drug for HSV-1 infection, ACV-resistant strains have been found in immune-compromised patients and drug toxicity has also been commonly reported. Therefore, there is an urge to search for new anti-HSV-1 agents. Natural products with potential anti-HSV-1 activity have the advantages of minimal side effects, reduced toxicity, and they exert their effect by various mechanisms. This paper will not only provide a reference for the safe dose of these agents if they are to be used in humans, referring to the interrelated data obtained from in vitro experiments, but also introduce the main pharmacodynamic mechanisms of traditional Chinese medicine (TCM) against HSV-1. Taken together, TCM functions as a potential source for HSV-1 therapy by direct (blocking viral attachment/absorption/penetration/replication) or indirect (reducing the susceptibility to HSV-1 or regulating autophagy) antiviral activities. The potential of these active components in the development of anti-HSV-1 drugs will also be described.
Collapse
Affiliation(s)
- Wen Li
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Xiao-Hua Wang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Zhuo Luo
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Li-Fang Liu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Chang Yan
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Chang-Yu Yan
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Guo-Dong Chen
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Hao Gao
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Wen-Jun Duan
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Hiroshi Kurihara
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Yi-Fang Li
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Rong-Rong He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
34
|
Diderichsen F, Augusto LGDS, Perez B. Understanding social inequalities in Zika infection and its consequences: A model of pathways and policy entry-points. Glob Public Health 2018; 14:675-683. [PMID: 30301438 DOI: 10.1080/17441692.2018.1532528] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The health consequences of arbovirus infections such as dengue fever (DENV), Chikungunya (CHIKV) and Zika (ZIKV) has in recent years become a public health challenge, due to failure of prevention followed by increased incidence and pronounced social inequality in occurrence and consequences. This motivates a more systematic analysis of the potential mechanisms and pathways that generate these inequalities. We present in the paper a model that delineates five possible mechanisms driving the inequality of ZIKV and congenital Zika syndrome (CZS). They include differential exposure to bad housing and sanitary conditions, differential exposure to vector density and virus, differential vulnerability to the health effects of exposure to virus, differential intrauterine susceptibility to the teratogenic effects of ZIKV infection and differential social consequences of caring for a disabled child. For each mechanism, we present empirical evidence or need for more research as well as a discussion about policy implications.
Collapse
Affiliation(s)
- Finn Diderichsen
- a Department of Public Health, University of Copenhagen, Copenhagen , Denmark.,b Oswaldo Cruz Foundation, IAM-FIOCRUZ/PE , Recife , Brazil
| | | | - Bernadete Perez
- c Departamento de Medicina Social, Federal University , Recife , Brazil
| |
Collapse
|
35
|
Suzich JB, Cliffe AR. Strength in diversity: Understanding the pathways to herpes simplex virus reactivation. Virology 2018; 522:81-91. [PMID: 30014861 DOI: 10.1016/j.virol.2018.07.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 01/09/2023]
Abstract
Herpes simplex virus (HSV) establishes a latent infection in peripheral neurons and can periodically reactivate to cause disease. Reactivation can be triggered by a variety of stimuli that activate different cellular processes to result in increased HSV lytic gene expression and production of infectious virus. The use of model systems has contributed significantly to our understanding of how reactivation of the virus is triggered by different physiological stimuli that are correlated with recrudescence of human disease. Furthermore, these models have led to the identification of both common and distinct mechanisms of different HSV reactivation pathways. Here, we summarize how the use of these diverse model systems has led to a better understanding of the complexities of HSV reactivation, and we present potential models linking cellular signaling pathways to changes in viral gene expression.
Collapse
Affiliation(s)
- Jon B Suzich
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, United States
| | - Anna R Cliffe
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, United States.
| |
Collapse
|
36
|
Crucian BE, Choukèr A, Simpson RJ, Mehta S, Marshall G, Smith SM, Zwart SR, Heer M, Ponomarev S, Whitmire A, Frippiat JP, Douglas GL, Lorenzi H, Buchheim JI, Makedonas G, Ginsburg GS, Ott CM, Pierson DL, Krieger SS, Baecker N, Sams C. Immune System Dysregulation During Spaceflight: Potential Countermeasures for Deep Space Exploration Missions. Front Immunol 2018; 9:1437. [PMID: 30018614 PMCID: PMC6038331 DOI: 10.3389/fimmu.2018.01437] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/11/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies have established that dysregulation of the human immune system and the reactivation of latent herpesviruses persists for the duration of a 6-month orbital spaceflight. It appears certain aspects of adaptive immunity are dysregulated during flight, yet some aspects of innate immunity are heightened. Interaction between adaptive and innate immunity also seems to be altered. Some crews experience persistent hypersensitivity reactions during flight. This phenomenon may, in synergy with extended duration and galactic radiation exposure, increase specific crew clinical risks during deep space exploration missions. The clinical challenge is based upon both the frequency of these phenomena in multiple crewmembers during low earth orbit missions and the inability to predict which specific individual crewmembers will experience these changes. Thus, a general countermeasure approach that offers the broadest possible coverage is needed. The vehicles, architecture, and mission profiles to enable such voyages are now under development. These include deployment and use of a cis-Lunar station (mid 2020s) with possible Moon surface operations, to be followed by multiple Mars flyby missions, and eventual human Mars surface exploration. Current ISS studies will continue to characterize physiological dysregulation associated with prolonged orbital spaceflight. However, sufficient information exists to begin consideration of both the need for, and nature of, specific immune countermeasures to ensure astronaut health. This article will review relevant in-place operational countermeasures onboard ISS and discuss a myriad of potential immune countermeasures for exploration missions. Discussion points include nutritional supplementation and functional foods, exercise and immunity, pharmacological options, the relationship between bone and immune countermeasures, and vaccination to mitigate herpes (and possibly other) virus risks. As the immune system has sentinel connectivity within every other physiological system, translational effects must be considered for all potential immune countermeasures. Finally, we shall discuss immune countermeasures in the context of their individualized implementation or precision medicine, based on crewmember specific immunological biases.
Collapse
Affiliation(s)
- Brian E. Crucian
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, United States
| | - Alexander Choukèr
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Richard J. Simpson
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ, United States
- Department of Pediatrics, The University of Arizona, Tucson, AZ, United States
- Department of Immunobiology, The University of Arizona, Tucson, AZ, United States
| | | | - Gailen Marshall
- University of Mississippi Medical Center, Jackson, MS, United States
| | - Scott M. Smith
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, United States
| | - Sara R. Zwart
- University of Texas Medical Branch, Galveston, TX, United States
| | - Martina Heer
- Institute of Nutritional and Food Sciences, University of Bonn, Bonn, Germany
| | | | | | - Jean P. Frippiat
- Stress Immunity Pathogens Laboratory, EA7300, Lorraine University, Nancy, France
| | - Grace L. Douglas
- Human Systems Engineering and Development Division, NASA Johnson Space Center, Houston, TX, United States
| | | | - Judith-Irina Buchheim
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | | | - Geoffrey S. Ginsburg
- Duke Center for Applied Genomics and Precision Medicine, Durham, NC, United States
| | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, United States
| | - Duane L. Pierson
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, United States
| | | | - Natalie Baecker
- Institute of Nutritional and Food Sciences, University of Bonn, Bonn, Germany
| | - Clarence Sams
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, United States
| |
Collapse
|
37
|
Holzer P, Farzi A, Hassan AM, Zenz G, Jačan A, Reichmann F. Visceral Inflammation and Immune Activation Stress the Brain. Front Immunol 2017; 8:1613. [PMID: 29213271 PMCID: PMC5702648 DOI: 10.3389/fimmu.2017.01613] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022] Open
Abstract
Stress refers to a dynamic process in which the homeostasis of an organism is challenged, the outcome depending on the type, severity, and duration of stressors involved, the stress responses triggered, and the stress resilience of the organism. Importantly, the relationship between stress and the immune system is bidirectional, as not only stressors have an impact on immune function, but alterations in immune function themselves can elicit stress responses. Such bidirectional interactions have been prominently identified to occur in the gastrointestinal tract in which there is a close cross-talk between the gut microbiota and the local immune system, governed by the permeability of the intestinal mucosa. External stressors disturb the homeostasis between microbiota and gut, these disturbances being signaled to the brain via multiple communication pathways constituting the gut-brain axis, ultimately eliciting stress responses and perturbations of brain function. In view of these relationships, the present article sets out to highlight some of the interactions between peripheral immune activation, especially in the visceral system, and brain function, behavior, and stress coping. These issues are exemplified by the way through which the intestinal microbiota as well as microbe-associated molecular patterns including lipopolysaccharide communicate with the immune system and brain, and the mechanisms whereby overt inflammation in the GI tract impacts on emotional-affective behavior, pain sensitivity, and stress coping. The interactions between the peripheral immune system and the brain take place along the gut-brain axis, the major communication pathways of which comprise microbial metabolites, gut hormones, immune mediators, and sensory neurons. Through these signaling systems, several transmitter and neuropeptide systems within the brain are altered under conditions of peripheral immune stress, enabling adaptive processes related to stress coping and resilience to take place. These aspects of the impact of immune stress on molecular and behavioral processes in the brain have a bearing on several disturbances of mental health and highlight novel opportunities of therapeutic intervention.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, Graz, Austria
| | - Aitak Farzi
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Geraldine Zenz
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Angela Jačan
- CBmed GmbH-Center for Biomarker Research in Medicine, Graz, Austria
| | - Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| |
Collapse
|
38
|
Carod-Artal FJ. Infectious diseases causing autonomic dysfunction. Clin Auton Res 2017; 28:67-81. [PMID: 28730326 DOI: 10.1007/s10286-017-0452-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/10/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To review infectious diseases that may cause autonomic dysfunction. METHODS Review of published papers indexed in medline/embase. RESULTS Autonomic dysfunction has been reported in retrovirus (human immunodeficiency virus (HIV), human T-lymphotropic virus), herpes viruses, flavivirus, enterovirus 71 and lyssavirus infections. Autonomic dysfunction is relatively common in HIV-infected patients and heart rate variability is reduced even in early stages of infection. Orthostatic hypotension, urinary dysfunction and hypohidrosis have been described in tropical spastic paraparesis patients. Varicella zoster reactivation from autonomic ganglia may be involved in visceral disease and chronic intestinal pseudo-obstruction. Autonomic and peripheral nervous system dysfunction may happen in acute tick-borne encephalitis virus infections. Hydrophobia, hypersalivation, dyspnea, photophobia, and piloerection are frequently observed in human rabies. Autonomic dysfunction and vagal denervation is common in Chagas disease. Neuronal depopulation occurs mainly in chagasic heart disease and myenteric plexus, and megacolon, megaesophagus and cardiomyopathy are common complications in the chronic stage of Chagas disease. Parasympathetic autonomic dysfunction precedes left ventricle systolic dysfunction in Chagas disease. A high prevalence of subclinical autonomic neuropathy in leprosy patients has been reported, and autonomic nerve dysfunction may be an early manifestation of the disease. Autonomic dysfunction features in leprosy include anhidrosis, impaired sweating function, localised alopecia ,and reduced heart rate variability. Urinary retention and intestinal pseudo-obstruction have been described in Lyme disease. Diphtheritic polyneuropathy, tetanus and botulism are examples of bacterial infections releasing toxins that affect the autonomic nervous system. CONCLUSIONS Autonomic dysfunction may be responsible for additional morbidity in some infectious diseases.
Collapse
Affiliation(s)
- Francisco Javier Carod-Artal
- Neurology Department, Raigmore Hospital, Inverness, UK. .,Health Sciences Faculty, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| |
Collapse
|