Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
J Med Virol ; 96(8): e29854, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39135475

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) has a high mortality rate compared to other infectious diseases. SFTS is particularly associated with a high risk of mortality in immunocompromised individuals, while most patients who die of SFTS exhibit symptoms of severe encephalitis before death. However, the region of brain damage and mechanisms by which the SFTS virus (SFTSV) causes encephalitis remains unknown. Here, we revealed that SFTSV infects the brainstem and spinal cord, which are regions of the brain associated with respiratory function, and motor nerves in IFNAR1-/- mice. Further, we show that A1-reactive astrocytes are activated, causing nerve cell death, in infected mice. Primary astrocytes of SFTSV-infected IFNAR1-/- mice also induced neuronal cell death through the activation of A1-reactive astrocytes. Herein, we showed that SFTSV induces fatal neuroinflammation in the brain regions important for respiratory function and motor nerve, which may underlie mortality in SFTS patients. This study provides new insights for the treatment of SFTS, for which there is currently no therapeutic approach.


Subject(s)
Astrocytes , Bunyaviridae Infections , Mice, Knockout , Phlebovirus , Receptor, Interferon alpha-beta , Animals , Astrocytes/virology , Astrocytes/pathology , Mice , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/deficiency , Phlebovirus/genetics , Phlebovirus/physiology , Phlebovirus/pathogenicity , Bunyaviridae Infections/virology , Bunyaviridae Infections/pathology , Bunyaviridae Infections/immunology , Brain/virology , Brain/pathology , Brain/immunology , Spinal Cord/virology , Spinal Cord/pathology , Disease Models, Animal , Neurons/virology , Neurons/pathology , Mice, Inbred C57BL , Brain Stem/virology , Brain Stem/pathology , Cell Death
2.
J Exp Med ; 221(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-39023559

ABSTRACT

Inherited deficiency of the RNA lariat-debranching enzyme 1 (DBR1) is a rare etiology of brainstem viral encephalitis. The cellular basis of disease and the range of viral predisposition are unclear. We report inherited DBR1 deficiency in a 14-year-old boy who suffered from isolated SARS-CoV-2 brainstem encephalitis. The patient is homozygous for a previously reported hypomorphic and pathogenic DBR1 variant (I120T). Consistently, DBR1 I120T/I120T fibroblasts from affected individuals from this and another unrelated kindred have similarly low levels of DBR1 protein and high levels of RNA lariats. DBR1 I120T/I120T human pluripotent stem cell (hPSC)-derived hindbrain neurons are highly susceptible to SARS-CoV-2 infection. Exogenous WT DBR1 expression in DBR1 I120T/I120T fibroblasts and hindbrain neurons rescued the RNA lariat accumulation phenotype. Moreover, expression of exogenous RNA lariats, mimicking DBR1 deficiency, increased the susceptibility of WT hindbrain neurons to SARS-CoV-2 infection. Inborn errors of DBR1 impair hindbrain neuron-intrinsic antiviral immunity, predisposing to viral infections of the brainstem, including that by SARS-CoV-2.


Subject(s)
Brain Stem , COVID-19 , Neurons , SARS-CoV-2 , Humans , Male , SARS-CoV-2/genetics , COVID-19/genetics , COVID-19/virology , Brain Stem/pathology , Brain Stem/virology , Brain Stem/metabolism , Adolescent , Neurons/metabolism , Neurons/pathology , Encephalitis, Viral/genetics , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Fibroblasts/metabolism , Rhombencephalon/metabolism
3.
Virus Res ; 347: 199420, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38880336

ABSTRACT

Human alphaherpesvirus 1 (HSV-1) establishes life-long latency in sensory neurons in trigeminal ganglia (TG), brainstem neurons, and other CNS neurons. Two important segments of the brainstem were examined in this study: principal sensory nucleus of the spinal trigeminal tract (Pr5) because it receives direct afferent inputs from TG, and locus coeruleus (LC) because it is indirectly connected to Pr5 and LC sends axonal projections to cortical structures, which may facilitate viral spread from brainstem to the brain. The only viral gene abundantly expressed during latency is the latency associated transcript (LAT). Previous studies revealed 8-week old female C57Bl/6 mice infected with a LAT null mutant (dLAT2903) versus wild-type (wt) HSV-1 exhibit higher levels of senescence markers and inflammation in LC of females. New studies revealed 1-year old mice latently infected with wt HSV-1 or dLAT2903 contained differences in neuroinflammation and senescence in Pr5 and LC versus young mice. In summary, these studies confirm HSV-1 promotes neuro-inflammation in the brainstem, which may accelerate neurodegenerative disease.


Subject(s)
Brain Stem , Herpesvirus 1, Human , Mice, Inbred C57BL , Neuroinflammatory Diseases , Virus Latency , Animals , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/genetics , Brain Stem/virology , Brain Stem/pathology , Mice , Female , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/pathology , Herpes Simplex/virology , Herpes Simplex/pathology , Aging , Humans , Latent Infection/virology , Trigeminal Ganglion/virology , Disease Models, Animal
4.
J Virol ; 97(10): e0095923, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37772825

ABSTRACT

IMPORTANCE: Viral encephalomyelitis outcome is dependent on host responses to neuronal infection. Interferon (IFN) is an important component of the innate response, and IFN regulatory factor (IRF) 7 is an inducible transcription factor for the synthesis of IFN-α. IRF7-deficient mice develop fatal paralysis after CNS infection with Sindbis virus, while wild-type mice recover. Irf7 -/- mice produce low levels of IFN-α but high levels of IFN-ß with induction of IFN-stimulated genes, so the reason for this difference is not understood. The current study shows that Irf7 -/- mice developed inflammation earlier but failed to clear virus from motor neuron-rich regions of the brainstem and spinal cord. Levels of IFN-γ and virus-specific antibody were comparable, indicating that IRF7 deficiency does not impair expression of these known viral clearance factors. Therefore, IRF7 is either necessary for the neuronal response to currently identified mediators of clearance or enables the production of additional antiviral factor(s) needed for clearance.


Subject(s)
Alphavirus Infections , Encephalomyelitis , Interferon Regulatory Factor-7 , Sindbis Virus , Animals , Mice , Alphavirus Infections/immunology , Alphavirus Infections/virology , Brain Stem/virology , Encephalomyelitis/immunology , Encephalomyelitis/virology , Inflammation/virology , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Motor Neurons/virology , Sindbis Virus/immunology , Spinal Cord/virology
5.
J Virol ; 96(17): e0108122, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35975996

ABSTRACT

Following acute infection, herpes simplex virus 1 (HSV-1) establishes lifelong latency in neurons. The latency associated transcript (LAT) is the only viral gene abundantly expressed during latency. Wild-type (WT) HSV-1 reactivates more efficiently than LAT mutants because LAT promotes establishment and maintenance of latency. While sensory neurons in trigeminal ganglia (TG) are important sites for latency, brainstem is also a site for latency and reactivation from latency. The principal sensory nucleus of the spinal trigeminal tract (Pr5) likely harbors latent HSV-1 because it receives afferent inputs from TG. The locus coeruleus (LC), an adjacent brainstem region, sends axonal projections to cortical structures and is indirectly linked to Pr5. Senescent cells accumulate in the nervous system during aging and accelerate neurodegenerative processes. Generally senescent cells undergo irreversible cell cycle arrest and produce inflammatory cytokines and chemokines. Based on these observations, we hypothesized HSV-1 influences senescence and inflammation in Pr5 and LC of latently infected mice. This hypothesis was tested using a mouse model of infection. Strikingly, female but not age-matched male mice latently infected with a LAT null mutant (dLAT2903) exhibited significantly higher levels of senescence markers and inflammation in LC, including cell cycle inhibitor p16, NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3), IL-1α, and IL-ß. Conversely, Pr5 in female but not male mice latently infected with WT HSV-1 or dLAT2903 exhibited enhanced expression of important inflammatory markers. The predilection of HSV-1 to induce senescence and inflammation in key brainstem regions of female mice infers that enhanced neurodegeneration occurs. IMPORTANCE HSV-1 (herpes simplex virus 1), an important human pathogen, establishes lifelong latency in neurons in trigeminal ganglia and the central nervous system. In contrast to productive infection, the only viral transcript abundantly expressed in latently infected neurons is the latency associated transcript (LAT). The brainstem, including principal sensory nucleus of the spinal trigeminal tract (Pr5) and locus coeruleus (LC), may expedite HSV-1 spread from trigeminal ganglia to the brain. Enhanced senescence and expression of key inflammatory markers were detected in LC of female mice latently infected with a LAT null mutant (dLAT2903) relative to age-matched male or female mice latently infected with wild-type HSV-1. Conversely, wild-type HSV-1 and dLAT2903 induced higher levels of senescence and inflammatory markers in Pr5 of latently infected female mice. In summary, enhanced inflammation and senescence in LC and Pr5 of female mice latently infected with HSV-1 are predicted to accelerate neurodegeneration.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Neuroinflammatory Diseases , Animals , Brain Stem/virology , Cellular Senescence , Female , Herpes Simplex/pathology , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Inflammation , Male , Mice , Mice, Inbred NOD , Neuroinflammatory Diseases/virology , Trigeminal Ganglion/virology , Virus Latency
6.
Nat Commun ; 12(1): 5809, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34608167

ABSTRACT

SARS-CoV-2 has caused a global pandemic of COVID-19 since its emergence in December 2019. The infection causes a severe acute respiratory syndrome and may also spread to central nervous system leading to neurological sequelae. We have developed and characterized two new organotypic cultures from hamster brainstem and lung tissues that offer a unique opportunity to study the early steps of viral infection and screening antivirals. These models are not dedicated to investigate how the virus reaches the brain. However, they allow validating the early tropism of the virus in the lungs and demonstrating that SARS-CoV-2 could infect the brainstem and the cerebellum, mainly by targeting granular neurons. Viral infection induces specific interferon and innate immune responses with patterns specific to each organ, along with cell death by apoptosis, necroptosis, and pyroptosis. Overall, our data illustrate the potential of rapid modeling of complex tissue-level interactions during infection by a newly emerged virus.


Subject(s)
Brain Stem/virology , Lung/virology , Models, Biological , SARS-CoV-2/pathogenicity , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alveolar Epithelial Cells/virology , Animals , Antiviral Agents/pharmacology , Brain Stem/cytology , Brain Stem/immunology , Brain Stem/pathology , Cricetinae , Immunity, Innate , Inflammation , Lung/cytology , Lung/immunology , Lung/pathology , Neurons/virology , Organ Culture Techniques , Regulated Cell Death , SARS-CoV-2/drug effects , Viral Tropism
7.
J Neurol ; 268(12): 4486-4491, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34003372

ABSTRACT

INTRODUCTION: SARS-CoV-2 might spread through the nervous system, reaching respiratory centers in the brainstem. Because we recently reported neurophysiological brainstem reflex abnormalities in COVID-19 patients, we here neuropathologically assessed structural brainstem damage in two COVID-19 patients. MATERIALS AND METHODS: We assessed neuropathological features in two patients who died of COVID-19 and in two COVID-19 negative patients as controls. Neuronal damage and corpora amylacea (CA) numbers /mm2 were histopathologically assessed. Other features studied were the immunohistochemical expression of the SARS-CoV-2 nucleoprotein (NP) and the Iba-1 antigen for glial activation. RESULTS: Autopsies showed normal gross brainstem anatomy. Histopathological examination demonstrated increased neuronal and CA damage in Covid-19 patients' medulla oblongata. Immunohistochemistry disclosed SARS-CoV-2 NP in brainstem neurons and glial cells, and in cranial nerves. Glial elements also exhibited a widespread increase in Iba-1 expression. Sars-Co-V2 was immunohistochemically detected in the vagus nerve fibers. DISCUSSION: Neuropathologic evidence showing SARS-CoV-2 in the brainstem and medullary damage in the area of respiratory centers strongly suggests that the pathophysiology of COVID-19-related respiratory failure includes a neurogenic component. Sars-Co-V2 detection in the vagus nerve, argues for viral trafficking between brainstem and lung.


Subject(s)
Brain Stem/virology , COVID-19 , Lung/virology , Nervous System Diseases , Humans , Nervous System Diseases/virology , SARS-CoV-2
8.
BMJ Case Rep ; 14(2)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33542002

ABSTRACT

Subacute sclerosing panencephalitis (SSPE) is a progressive lethal neurological inflammatory disease due to persistent, wild measles virus infection in the central nervous system that is seen most frequently in children and young adolescents. Atypical presentations are seen in up to 10% of cases. Most frequently and severely affected region in the brain is the parieto-occipital region of the brain. Less commonly involved organs are the cerebellum, basal ganglia and corpus callosum. Brainstem involvement is rare and usually occurs when other areas of brain are involved along with it. Here, we describe an unusual male patient of 15 years age, having SSPE with MRI of brain showing extensive involvement of brainstem with no significant involvement of other cortical structures of the brain. It is very rarely described in SSPE, but one should be vigilant about such involvement of brainstem and cerebellum, and SSPE should not be missed when brainstem hyperintensities are seen in MRI brain with or without other region of the brain to avoid misdiagnosis.


Subject(s)
Brain Stem/physiopathology , Subacute Sclerosing Panencephalitis/diagnosis , Adolescent , Brain Stem/virology , Electroencephalography , Facial Paralysis/etiology , Humans , Magnetic Resonance Imaging , Male , Measles/virology , Myoclonus/etiology
9.
ACS Chem Neurosci ; 12(4): 573-580, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33538586

ABSTRACT

Long-COVID is a postviral illness that can affect survivors of COVID-19, regardless of initial disease severity or age. Symptoms of long-COVID include fatigue, dyspnea, gastrointestinal and cardiac problems, cognitive impairments, myalgia, and others. While the possible causes of long-COVID include long-term tissue damage, viral persistence, and chronic inflammation, the review proposes, perhaps for the first time, that persistent brainstem dysfunction may also be involved. This hypothesis can be split into two parts. The first is the brainstem tropism and damage in COVID-19. As the brainstem has a relatively high expression of ACE2 receptor compared with other brain regions, SARS-CoV-2 may exhibit tropism therein. Evidence also exists that neuropilin-1, a co-receptor of SARS-CoV-2, may be expressed in the brainstem. Indeed, autopsy studies have found SARS-CoV-2 RNA and proteins in the brainstem. The brainstem is also highly prone to damage from pathological immune or vascular activation, which has also been observed in autopsy of COVID-19 cases. The second part concerns functions of the brainstem that overlap with symptoms of long-COVID. The brainstem contains numerous distinct nuclei and subparts that regulate the respiratory, cardiovascular, gastrointestinal, and neurological processes, which can be linked to long-COVID. As neurons do not readily regenerate, brainstem dysfunction may be long-lasting and, thus, is long-COVID. Indeed, brainstem dysfunction has been implicated in other similar disorders, such as chronic pain and migraine and myalgic encephalomyelitis or chronic fatigue syndrome.


Subject(s)
Brain Diseases/physiopathology , Brain Stem/physiopathology , COVID-19/complications , Inflammation/physiopathology , Thrombosis/physiopathology , Angiotensin-Converting Enzyme 2/metabolism , Brain Diseases/metabolism , Brain Diseases/virology , Brain Stem/blood supply , Brain Stem/metabolism , Brain Stem/virology , COVID-19/metabolism , COVID-19/physiopathology , Humans , Inflammation/metabolism , Inflammation/virology , Neuropilin-1/metabolism , RNA, Viral/isolation & purification , RNA, Viral/metabolism , Receptors, Coronavirus/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Thrombosis/metabolism , Thrombosis/virology , Viral Tropism , Post-Acute COVID-19 Syndrome
11.
Neurol India ; 68(5): 989-993, 2020.
Article in English | MEDLINE | ID: mdl-33109839

ABSTRACT

BACKGROUND: A terrible pandemic, Covid-19, has captivated scientists to investigate if SARS-CoV-2 virus infects the central nervous system (CNS). A crucial question is if acute respiratory distress syndrome (ARDS), the main cause of death in this pandemic, and often refractory to treatments, can be explained by respiratory center dysfunction. OBJECTIVE: To discuss that ARDS can be caused by SARS-CoV-2 infection of the respiratory center in the brainstem. MATERIALS AND METHODS: I reviewed literature about SARS-CoV-2 mechanisms to infect the respiratory center in the brainstem. RESULTS AND CONCLUSIONS: An increasing amount of reports demonstrates that neurotropism is a common feature of coronavirus, which have been found in the brains of patients and experimental models, where the brainstem was severely infested. Recent studies have provided tremendous indication of the incidence of acute respiratory failure due to SARS-CoV-2 infection of the brainstem. SARS-CoV-2 might infect the CNS through the olfactory bulb, spreading from the olfactory nerves to the rhinencephalon, and finally reaching the brainstem. Hence, the virus infection causes respiratory center dysfunctions, leading to ARDS in COVID-19 patients. I conclude that acute ARDS in Covid-19 can be caused by SARS-CoV-2 invasion of brainstem respiratory center, suggesting the needs of more specific and aggressive treatments, with the direct participation of neurologists and neurointensivists.


Subject(s)
Coronavirus Infections/physiopathology , Pneumonia, Viral/physiopathology , Respiratory Center/physiopathology , Respiratory Distress Syndrome/physiopathology , Betacoronavirus , Brain Stem/physiopathology , Brain Stem/virology , COVID-19 , Humans , Hypoxia/physiopathology , Pandemics , Respiratory Center/virology , SARS-CoV-2 , Viral Tropism
12.
Mol Neurobiol ; 57(12): 5263-5275, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32869183

ABSTRACT

Similar to its predecessors, coronavirus disease 2019 (COVID-19) exhibits neurotrophic properties, which lead to progression of neurologic sequelae. Besides direct viral invasion to the central nervous system (CNS), indirect CNS involvement through viral-mediated immune response is plausible. Aberrant immune pathways such as extreme release of cytokines (cytokine storm), autoimmunity mediated by cross-reactivity between CNS components and viral particles, and microglial activation propagate CNS damage in these patients. Here, we review the currently available evidence to discuss the plausible immunologic pathways that may contribute to the development of COVID-19 neurological complications, namely Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis, Guillain-Barre syndrome, seizure, and brainstem involvement.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Nervous System Diseases/etiology , Pandemics , Pneumonia, Viral/complications , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , Brain Stem/physiopathology , Brain Stem/virology , COVID-19 , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytopathogenic Effect, Viral , Disease Outbreaks , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/immunology , Humans , Mice , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Nerve Tissue Proteins/physiology , Nervous System Diseases/immunology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/immunology , Neuroglia/pathology , Neuroglia/virology , Neurons/pathology , Neurons/virology , Peptidyl-Dipeptidase A/physiology , Pneumonia, Viral/immunology , Receptors, Virus/physiology , Respiratory Insufficiency/etiology , Respiratory Insufficiency/physiopathology , SARS-CoV-2 , Seizures/etiology , Seizures/immunology , Severe Acute Respiratory Syndrome/complications , Severe Acute Respiratory Syndrome/epidemiology , Stroke/etiology , Stroke/immunology
13.
J Neurovirol ; 26(6): 846-862, 2020 12.
Article in English | MEDLINE | ID: mdl-32910432

ABSTRACT

Previous studies showed that persons living with HIV (PLWH) demonstrate higher brain prefrontal cortex neuroinflammation and immunoproteasome expression compared to HIV-negative individuals; these associate positively with HIV levels. Lower expression of the antioxidant enzyme heme oxygenase 1 (HO-1) was observed in PLWH with HIV-associated neurocognitive impairment (HIV-NCI) compared to neurocognitively normal PLWH. We hypothesized that similar expression patterns occur throughout cortical, subcortical, and brainstem regions in PLWH, and that neuroinflammation and immunoproteasome expression associate with lower expression of neuronal markers. We analyzed autopsied brains (15 regions) from 9 PLWH without HIV-NCI and 7 matched HIV-negative individuals. Using Western blot and RT-qPCR, we quantified synaptic, inflammatory, immunoproteasome, endothelial, and antioxidant biomarkers, including HO-1 and its isoform heme oxygenase 2 (HO-2). In these PLWH without HIV-NCI, we observed higher expression of neuroinflammatory, endothelial, and immunoproteasome markers in multiple cortical and subcortical regions compared to HIV-negative individuals, suggesting a global brain inflammatory response to HIV. Several regions, including posterior cingulate cortex, globus pallidus, and cerebellum, showed a distinct pattern of higher type I interferon (IFN)-stimulated gene and immunoproteasome expression. PLWH without HIV-NCI also had (i) stable or higher HO-1 expression and positive associations between (ii) HO-1 and HIV levels (CSF, plasma) and (iii) HO-1 expression and neuroinflammation, in multiple cortical, subcortical, and brainstem regions. We observed no differences in synaptic marker expression, suggesting little, if any, associated neuronal injury. We speculate that this may reflect a neuroprotective effect of a concurrent HO-1 antioxidant response despite global neuroinflammation, which will require further investigation.


Subject(s)
Cerebral Cortex/metabolism , Cognitive Dysfunction/genetics , HIV Infections/genetics , HIV-1/pathogenicity , Heme Oxygenase-1/genetics , Aged , Amygdala/metabolism , Amygdala/virology , Autopsy , Biomarkers/metabolism , Brain Stem/metabolism , Brain Stem/virology , Case-Control Studies , Caudate Nucleus/metabolism , Caudate Nucleus/virology , Cerebral Cortex/virology , Cognitive Dysfunction/complications , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/virology , Female , Gene Expression Regulation , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/virology , Heme Oxygenase-1/metabolism , Humans , Inflammation , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Male , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
14.
Mol Med ; 26(1): 58, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546125

ABSTRACT

In light of the present therapeutic situation in COVID-19, any measure to improve course and outcome of seriously affected individuals is of utmost importance. We recap here evidence that supports the use of human recombinant erythropoietin (EPO) for ameliorating course and outcome of seriously ill COVID-19 patients. This brief expert review grounds on available subject-relevant literature searched until May 14, 2020, including Medline, Google Scholar, and preprint servers. We delineate in brief sections, each introduced by a summary of respective COVID-19 references, how EPO may target a number of the gravest sequelae of these patients. EPO is expected to: (1) improve respiration at several levels including lung, brainstem, spinal cord and respiratory muscles; (2) counteract overshooting inflammation caused by cytokine storm/ inflammasome; (3) act neuroprotective and neuroregenerative in brain and peripheral nervous system. Based on this accumulating experimental and clinical evidence, we finally provide the research design for a double-blind placebo-controlled randomized clinical trial including severely affected patients, which is planned to start shortly.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/drug therapy , Cytokine Release Syndrome/prevention & control , Erythropoietin/therapeutic use , Neuroprotective Agents/therapeutic use , Pneumonia, Viral/drug therapy , Respiratory System Agents/therapeutic use , Brain Stem/drug effects , Brain Stem/immunology , Brain Stem/virology , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/virology , Double-Blind Method , Humans , Lung/drug effects , Lung/immunology , Lung/virology , Pandemics , Phrenic Nerve/drug effects , Phrenic Nerve/immunology , Phrenic Nerve/virology , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Proof of Concept Study , Randomized Controlled Trials as Topic , Recombinant Proteins/therapeutic use , Respiratory Muscles/drug effects , Respiratory Muscles/immunology , Respiratory Muscles/virology , SARS-CoV-2 , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/virology
15.
Neurol Sci ; 41(7): 1663-1665, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32472516

ABSTRACT

Respiratory failure is the most worrisome problem of COVID-19. Patients may develop severe pneumonia requiring invasive mechanical ventilation and a significant proportion of them dies. It has been suggested that brainstem might play a role in severe respiratory failure of COVID-19 patients. We described three COVID-19 patients in ICU at Federico II Hospital in Naples that, although had recovered from pneumonia, could not be weaned from invasive mechanical ventilation. Our clinical evaluation was consistent with an involvement of the brainstem and especially of respiratory centre thus possibly explaining the weaning failure in patients that were awake and had recovered from lung involvement. Our data, though limited, indicate that brainstem involvement may play a role in respiratory failure and perhaps in the high death rate of COVID-19 patients. Moreover, the weaning failure from mechanical ventilation due to central respiratory drive depression might underlie the unusual long stay in ICU reported for COVID-19 patients.


Subject(s)
Betacoronavirus , Brain Stem/virology , Coronavirus Infections/virology , Pneumonia, Viral/virology , Respiratory Insufficiency/virology , COVID-19 , Coronavirus Infections/etiology , Coronavirus Infections/mortality , Humans , Pandemics , Pneumonia, Viral/etiology , Pneumonia, Viral/mortality , Respiration, Artificial , SARS-CoV-2
16.
Cancer Gene Ther ; 27(3-4): 246-255, 2020 04.
Article in English | MEDLINE | ID: mdl-30918335

ABSTRACT

Primary malignant central nervous system (CNS) tumors are the leading cause of childhood cancer-related death and morbidity. While advances in surgery, radiation, and chemotherapy have improved the survival rates in children with malignant brain tumors, mortality persists in certain subpopulations and current therapies are associated with extreme morbidity. This is especially true for children with malignant infratentorial tumors. Accordingly, G207, a genetically engineered herpes simplex virus (HSV-1) capable of selectively targeting cancer cells has emerged as a promising therapeutic option for this patient population. Herein, we demonstrate that cerebellar inoculation of G207 was systemically non-toxic in an immunocompetent, HSV-1 sensitive mouse strain (CBA/J). Mice had neither abnormal brain/organ pathology nor evidence of G207 replication by immunohistochemistry at days 7 and 30 after cerebellar G207 inoculation. While a minute amount viral DNA was recovered in the cerebellum and brainstem of mice at day 7, no viral DNA persisted at day 30. Critically, G207 delivered to the cerebellum was able to target/treat the highly aggressive MYC-overexpressed group 3 murine medulloblastoma increasing survival vs controls. These results provide critical safety and efficacy data to support the translation of G207 for pediatric clinical trials in intractable cerebellar malignancies.


Subject(s)
Cerebellar Neoplasms/therapy , Herpesvirus 1, Human/immunology , Medulloblastoma/therapy , Oncolytic Virotherapy/methods , Animals , Brain Stem/pathology , Brain Stem/virology , Cell Line, Tumor/transplantation , Cerebellar Neoplasms/immunology , Cerebellar Neoplasms/pathology , Cerebellum/pathology , Cerebellum/virology , DNA, Viral/isolation & purification , Disease Models, Animal , Genetic Engineering , Herpesvirus 1, Human/genetics , Humans , Injections, Intralesional , Medulloblastoma/immunology , Medulloblastoma/pathology , Mice , Mice, Inbred CBA , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology
17.
BMJ Case Rep ; 12(11)2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31732545

ABSTRACT

Enterovirus D68 (EV-D68) causes respiratory illnesses such as pneumonia, and has been reported to cause acute flaccid myelitis. Enterovirus A71 (EV-A71) is known to cause cardiopulmonary failure due to brainstem encephalitis, but there have been few reports of these conditions being associated with EV-D68. Outbreaks of EV-D68 infection have occurred in the USA, Canada, Europe and Asia. Clinical management is largely supportive and there are no specific antivirals available. The case patient, a 4-year-old girl, had cardiopulmonary failure due to brainstem encephalitis. EV-D68 was isolated from a throat swab. On admission, she had cardiopulmonary failure, which required intensive care using a ventilator and inotropic agents. Her cardiac function improved, but she had residual bulbar paralysis and limb weakness, which resolved over a 6-month period. This case confirms that EV-D68, may cause severe illness due to brainstem encephalitis, similar to that caused by EV-A71.


Subject(s)
Brain Stem/virology , Encephalitis, Viral/complications , Enterovirus D, Human , Enterovirus Infections/complications , Heart Failure/virology , Respiratory Insufficiency/virology , Bulbar Palsy, Progressive/therapy , Bulbar Palsy, Progressive/virology , Child, Preschool , Female , Heart Failure/therapy , Humans , Respiratory Insufficiency/therapy
18.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597775

ABSTRACT

Demyelinating central nervous system (CNS) disorders like multiple sclerosis (MS) and acute disseminated encephalomyelitis (ADEM) have been difficult to study and treat due to the lack of understanding of their etiology. Numerous cases point to the link between herpes simplex virus (HSV) infection and multifocal CNS demyelination in humans; however, convincing evidence from animal models has been missing. In this work, we found that HSV-1 infection of the cotton rat Sigmodon hispidus via a common route (lip abrasion) can cause multifocal CNS demyelination and inflammation. Remyelination occurred shortly after demyelination in HSV-1-infected cotton rats but could be incomplete, resulting in "scars," further supporting an association between HSV-1 infection and multifocal demyelinating disorders. Virus was detected sequentially in the lip, trigeminal ganglia, and brain of infected animals. Brain pathology developed primarily on the ipsilateral side of the brain stem, in the cerebellum, and contralateral side of the forebrain/midbrain, suggesting that the changes may ascend along the trigeminal lemniscus pathway. Neurologic defects occasionally detected in infected animals (e.g., defective whisker touch and blink responses and compromised balance) could be representative of the brain stem/cerebellum dysfunction. Immunization of cotton rats with a split HSV-1 vaccine protected animals against viral replication and brain pathology, suggesting that vaccination against HSV-1 may protect against demyelinating disorders.IMPORTANCE Our work demonstrates for the first time a direct association between infection with herpes simplex virus 1, a ubiquitous human pathogen generally associated with facial cold sores, and multifocal brain demyelination in an otherwise normal host, the cotton rat Sigmodon hispidus For a long time, demyelinating diseases were considered to be autoimmune in nature and were studied by indirect methods, such as immunizing animals with myelin components or feeding them toxic substances that induce demyelination. Treatment against demyelinating diseases has been elusive, partially because of their unknown etiology. This work provides the first experimental evidence for the role of HSV-1 as the etiologic agent of multifocal brain demyelination in a normal host and suggests that vaccination against HSV-1 can help to combat demyelinating disorders.


Subject(s)
Demyelinating Diseases/prevention & control , Encephalitis/prevention & control , Herpes Simplex Virus Vaccines/administration & dosage , Herpes Simplex/prevention & control , Herpesvirus 1, Human/drug effects , Animals , Brain Stem/drug effects , Brain Stem/immunology , Brain Stem/pathology , Brain Stem/virology , Cerebellum/drug effects , Cerebellum/immunology , Cerebellum/pathology , Cerebellum/virology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Disease Models, Animal , Encephalitis/immunology , Encephalitis/pathology , Encephalitis/virology , Female , Herpes Simplex/immunology , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Humans , Male , Prosencephalon/drug effects , Prosencephalon/immunology , Prosencephalon/pathology , Prosencephalon/virology , Sigmodontinae , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/immunology , Trigeminal Ganglion/pathology , Trigeminal Ganglion/virology , Vaccination , Viral Load/drug effects
19.
BMJ Case Rep ; 12(8)2019 Aug 26.
Article in English | MEDLINE | ID: mdl-31451462

ABSTRACT

Rituximab and other B cell depleting agents are increasingly used for haematological, immunological and neurological diseases. In a small minority, immunosuppression leads to increased virulence of normally mild infections. Brainstem encephalitis has been described occurring after infection from enteroviruses, more commonly in the paediatric population, but also in immunosuppressed adults. In this paper, we describe an enteroviral brainstem encephalitis in a rituximab-immunosuppressed patient. The enterovirus identified was Coxsackie A16, which has never yet been reported to cause brainstem encephalitis in an adult.


Subject(s)
Cerebrospinal Fluid/virology , Coxsackievirus Infections , Encephalitis , Enterovirus/isolation & purification , Immunoglobulins, Intravenous/administration & dosage , Rituximab , Adult , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/immunology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Stem/diagnostic imaging , Brain Stem/virology , Coxsackievirus Infections/diagnosis , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/therapy , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Encephalitis/diagnosis , Encephalitis/physiopathology , Encephalitis/therapy , Encephalitis/virology , Humans , Immunocompromised Host , Immunologic Factors/administration & dosage , Lymphoma, Follicular/drug therapy , Magnetic Resonance Imaging/methods , Male , Prednisone/administration & dosage , Rituximab/administration & dosage , Rituximab/adverse effects , Rituximab/immunology , Treatment Outcome , Vincristine/administration & dosage
20.
Rev Med Virol ; 29(5): e2058, 2019 09.
Article in English | MEDLINE | ID: mdl-31237061

ABSTRACT

Subacute sclerosing panencephalitis (SSPE) is a slowly progressive brain disorder caused by mutant measles virus. SSPE affects younger age groups. SSPE incidence is proportional to that of measles. High-income countries have seen substantial decline in SSPE incidence following universal vaccination against measles. SSPE virus differs from wild measles virus. Measles virus genome recovered from the autopsied brain tissues demonstrates clustered mutations in virus genome particularly in the M gene. These mutations destroy the structure and functioning of the encoded proteins. Complete infectious virus particle has rarely been recovered from the brain. Human neurons lack required receptor for entry of measles virus inside the neurons. Recent in vitro studies suggest that mutations in F protein confer hyperfusogenic properties to measles virus facilitating transneuronal viral spread. The inflammatory response in the brain leads to extensive tissue damage. Clinically, SSPE is characterized by florid panencephalitis. Clinically, SSPE is characterized by cognitive decline, periodic myoclonus, gait abnormalities, vision loss, and ultimately to a vegetative state. Chorioretinitis is a common ocular abnormality. Electroencephalography (EEG) shows characteristic periodic discharges. Neuroimaging demonstrates periventricular white matter signal abnormalities. In advanced stages, there is marked cerebral atrophy. Definitive diagnosis requires demonstration of elevated measles antibody titers in cerebrospinal fluid (CSF). Many drugs have been used to stabilize the course of the disease but without evidence from randomized clinical trials. Six percent of patients may experience prolonged spontaneous remission. Fusion inhibitor peptide may, in the future, be exploited to treat SSPE. A universal vaccination against measles is the only proven way to tackle this menace currently.


Subject(s)
Subacute Sclerosing Panencephalitis/diagnosis , Subacute Sclerosing Panencephalitis/etiology , Biomarkers , Brain/diagnostic imaging , Brain/pathology , Brain/virology , Brain Stem/diagnostic imaging , Brain Stem/pathology , Brain Stem/virology , Diagnosis, Differential , Disease Management , Disease Susceptibility , Electroencephalography , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Measles virus/physiology , Neuroimaging/methods , Phenotype , Pregnancy , Prognosis , Subacute Sclerosing Panencephalitis/epidemiology , Subacute Sclerosing Panencephalitis/therapy , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL