Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 25(7): 1158-1171, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38902519

ABSTRACT

Up to 25% of individuals infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exhibit postacute cognitive sequelae. Although millions of cases of coronavirus disease 2019 (COVID-19)-mediated memory dysfunction are accumulating worldwide, the underlying mechanisms and how vaccination lowers risk are unknown. Interleukin-1 (IL-1), a key component of innate immune defense against SARS-CoV-2 infection, is elevated in the hippocampi of individuals with COVID-19. Here we show that intranasal infection of C57BL/6J mice with SARS-CoV-2 Beta variant leads to central nervous system infiltration of Ly6Chi monocytes and microglial activation. Accordingly, SARS-CoV-2, but not H1N1 influenza virus, increases levels of brain IL-1ß and induces persistent IL-1R1-mediated loss of hippocampal neurogenesis, which promotes postacute cognitive deficits. Vaccination with a low dose of adenoviral-vectored spike protein prevents hippocampal production of IL-1ß during breakthrough SARS-CoV-2 infection, loss of neurogenesis and subsequent memory deficits. Our study identifies IL-1ß as one potential mechanism driving SARS-CoV-2-induced cognitive impairment in a new mouse model that is prevented by vaccination.


Subject(s)
COVID-19 , Hippocampus , Interleukin-1beta , Memory Disorders , Mice, Inbred C57BL , Neurogenesis , SARS-CoV-2 , Animals , Interleukin-1beta/metabolism , Interleukin-1beta/immunology , Mice , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Hippocampus/immunology , Hippocampus/metabolism , Memory Disorders/immunology , Neurogenesis/immunology , Vaccination , Spike Glycoprotein, Coronavirus/immunology , COVID-19 Vaccines/immunology , Male , Humans , Microglia/immunology , Microglia/metabolism , Disease Models, Animal , Receptors, Interleukin-1 Type I/metabolism , Receptors, Interleukin-1 Type I/genetics , Monocytes/immunology , Monocytes/metabolism , Female
2.
J Neuroinflammation ; 21(1): 24, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38233868

ABSTRACT

BACKGROUND: Venezuelan Equine Encephalitis virus (VEEV) may enter the central nervous system (CNS) within olfactory sensory neurons (OSN) that originate in the nasal cavity after intranasal exposure. While it is known that VEEV has evolved several mechanisms to inhibit type I interferon (IFN) signaling within infected cells, whether this inhibits virologic control during neuroinvasion along OSN has not been studied. METHODS: We utilized an established murine model of intranasal infection with VEEV and a repository of scRNAseq data from IFN-treated OSN to assess the cellular targets and IFN signaling responses after VEEV exposure. RESULTS: We found that immature OSN, which express higher levels of the VEEV receptor LDLRAD3 than mature OSN, are the first cells infected by VEEV. Despite rapid VEEV neuroinvasion after intranasal exposure, olfactory neuroepithelium (ONE) and olfactory bulb (OB) IFN responses, as assessed by evaluation of expression of interferon signaling genes (ISG), are delayed for up to 48 h during VEEV neuroinvasion, representing a potential therapeutic window. Indeed, a single intranasal dose of recombinant IFNα triggers early ISG expression in both the nasal cavity and OB. When administered at the time of or early after infection, IFNα treatment delayed onset of sequelae associated with encephalitis and extended survival by several days. VEEV replication after IFN treatment was also transiently suppressed in the ONE, which inhibited subsequent invasion into the CNS. CONCLUSIONS: Our results demonstrate a critical and promising first evaluation of intranasal IFNα for the treatment of human encephalitic alphavirus exposures.


Subject(s)
Encephalitis Virus, Venezuelan Equine , Olfactory Receptor Neurons , Humans , Mice , Animals , Encephalitis Virus, Venezuelan Equine/genetics , Central Nervous System , Virus Replication
3.
Res Sq ; 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37790551

ABSTRACT

Up to 25% of SARS-CoV-2 patients exhibit post-acute cognitive sequelae. Although millions of cases of COVID-19-mediated memory dysfunction are accumulating worldwide, the underlying mechanisms and how vaccination lowers risk are unknown. Interleukin-1, a key component of innate immune defense against SARS-CoV-2 infection, is elevated in the hippocampi of COVID-19 patients. Here we show that intranasal infection of C57BL/6J mice with SARS-CoV-2 beta variant, leads to CNS infiltration of Ly6Chi monocytes and microglial activation. Accordingly, SARS-CoV-2, but not H1N1 influenza virus, increases levels of brain IL-1ß and induces persistent IL-1R1-mediated loss of hippocampal neurogenesis, which promotes post-acute cognitive deficits. Breakthrough infection after vaccination with a low dose of adenoviral vectored Spike protein prevents hippocampal production of IL-1ß during breakthrough SARS-CoV-2 infection, loss of neurogenesis, and subsequent memory deficits. Our study identifies IL-1ß as one potential mechanism driving SARS-CoV-2-induced cognitive impairment in a new murine model that is prevented by vaccination.

4.
Cell Rep ; 42(8): 112946, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37556325

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an encephalitic alphavirus responsible for epidemics of neurological disease across the Americas. Low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3) is a recently reported entry receptor for VEEV. Here, using wild-type and Ldlrad3-deficient mice, we define a critical role for LDLRAD3 in controlling steps in VEEV infection, pathogenesis, and neurotropism. Our analysis shows that LDLRAD3 is required for efficient VEEV infection and pathogenesis prior to and after central nervous system invasion. Ldlrad3-deficient mice survive intranasal and intracranial VEEV inoculation and show reduced infection of neurons in different brain regions. As LDLRAD3 is a determinant of pathogenesis and an entry receptor required for VEEV infection of neurons of the brain, receptor-targeted therapies may hold promise as countermeasures.


Subject(s)
Encephalomyelitis, Venezuelan Equine , Receptors, LDL , Animals , Mice , Brain/pathology , Central Nervous System , Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/pathology , Receptors, LDL/physiology
5.
bioRxiv ; 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37425867

ABSTRACT

Venezuelan Equine Encephalitis virus (VEEV) may enter the central nervous system (CNS) within olfactory sensory neurons (OSN) that originate in the nasal cavity after intranasal exposure. While it is known that VEEV has evolved several mechanisms to inhibit type I interferon (IFN) signaling within infected cells, whether this inhibits virologic control during neuroinvasion along OSN has not been studied. Here, we utilized an established murine model of intranasal infection with VEEV to assess the cellular targets and IFN signaling responses after VEEV exposure. We found that immature OSN, which express higher levels of the VEEV receptor LDLRAD3 than mature OSN, are the first cells infected by VEEV. Despite rapid VEEV neuroinvasion after intranasal exposure, olfactory neuroepithelium (ONE) and olfactory bulb (OB) IFN responses, as assessed by evaluation of expression of interferon signaling genes (ISG), are delayed for up to 48 hours during VEEV neuroinvasion, representing a potential therapeutic window. Indeed, a single intranasal dose of recombinant IFNα triggers early ISG expression in both the nasal cavity and OB. When administered at the time of or early after infection, IFNα treatment delayed onset of sequelae associated with encephalitis and extended survival by several days. VEEV replication after IFN treatment was also transiently suppressed in the ONE, which inhibited subsequent invasion into the CNS. Our results demonstrate a critical and promising first evaluation of intranasal IFNα for the treatment of human encephalitic alphavirus exposures.

6.
Proc Natl Acad Sci U S A ; 120(26): e2306318120, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37307435

ABSTRACT

Histidine-rich protein II (HRPII) is secreted by Plasmodium falciparum during the blood stage of malaria infection. High plasma levels of HRPII are associated with cerebral malaria, a severe and highly fatal complication of malaria. HRPII has been shown to induce vascular leakage, the hallmark of cerebral malaria, in blood-brain barrier (BBB) and animal models. We have discovered an important mechanism for BBB disruption that is driven by unique features of HRPII. By characterizing serum from infected patients and HRPII produced by P. falciparum parasites in culture, we found that HRPII exists in large multimeric particles of 14 polypeptides that are richly laden with up to 700 hemes per particle. Heme loading of HRPII is required for efficient binding and internalization via caveolin-mediated endocytosis in hCMEC/D3 cerebral microvascular endothelial cells. Upon acidification of endolysosomes, two-thirds of the hemes are released from acid-labile binding sites and metabolized by heme oxygenase 1, generating ferric iron and reactive oxygen species. Subsequent activation of the NLRP3 inflammasome and IL-1ß secretion resulted in endothelial leakage. Inhibition of these pathways with heme sequestration, iron chelation, or anti-inflammatory drugs protected the integrity of the BBB culture model from HRPII:heme. Increased cerebral vascular permeability was seen after injection of young mice with heme-loaded HRPII (HRPII:heme) but not with heme-depleted HRPII. We propose that during severe malaria infection, HRPII:heme nanoparticles in the bloodstream deliver an overwhelming iron load to endothelial cells to cause vascular inflammation and edema. Disrupting this process is an opportunity for targeted adjunctive therapies to reduce the morbidity and mortality of cerebral malaria.


Subject(s)
Hemeproteins , Malaria, Cerebral , Malaria, Falciparum , Animals , Mice , Histidine , Endothelial Cells , Inflammation , Heme , Iron
7.
Am J Forensic Med Pathol ; 42(3): 248-251, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33560647

ABSTRACT

ABSTRACT: Many fragmenting and frangible projectiles have been developed in the course of firearm history. In addition, partially because of the concerns of range and environmental contamination, bullets constructed without lead have become increasingly common. A case regarding a unique projectile that incorporates both features, the G2 Research Radically Invasive Projectile ammunition, is discussed in this article. Here we report a 25-year-old woman who died of multiple gunshot wounds caused by G2 Research Radically Invasive Projectile ammunition. Because of the bullet's unique design, the wounds demonstrated characteristic radiographic patterns and unique autopsy findings. Familiarity with these findings is important to forensic pathologists in terms of case documentation, projectile recovery, and personal safety.


Subject(s)
Forensic Ballistics , Wounds, Gunshot/diagnostic imaging , Wounds, Gunshot/pathology , Adult , Female , Humans , Radiography
8.
Neurooncol Adv ; 2(1): vdaa071, 2020.
Article in English | MEDLINE | ID: mdl-32666049

ABSTRACT

BACKGROUND: The blood-brain and blood-tumor barriers (BBB and BTB), which restrict the entry of most drugs into the brain and tumor, respectively, are a significant challenge in the treatment of glioblastoma. Laser interstitial thermal therapy (LITT) is a minimally invasive surgical technique increasingly used clinically for tumor cell ablation. Recent evidence suggests that LITT might locally disrupt BBB integrity, creating a potential therapeutic window of opportunity to deliver otherwise brain-impermeant agents. METHODS: We established a LITT mouse model to test if laser therapy can increase BBB/BTB permeability in vivo. Mice underwent orthotopic glioblastoma tumor implantation followed by LITT in combination with BBB tracers or the anticancer drug doxorubicin. BBB/BTB permeability was measured using fluorimetry, microscopy, and immunofluorescence. An in vitro endothelial cell model was also used to corroborate findings. RESULTS: LITT substantially disrupted the BBB and BTB locally, with increased permeability up to 30 days after the intervention. Remarkably, molecules as large as human immunoglobulin extravasated through blood vessels and permeated laser-treated brain tissue and tumors. Mechanistically, LITT decreased tight junction integrity and increased brain endothelial cell transcytosis. Treatment of mice bearing glioblastoma tumors with LITT and adjuvant doxorubicin, which is typically brain-impermeant, significantly increased animal survival. CONCLUSIONS: Together, these results suggest that LITT can locally disrupt the BBB and BTB, enabling the targeted delivery of systemic therapies, including, potentially, antibody-based agents.

9.
mBio ; 11(1)2020 02 11.
Article in English | MEDLINE | ID: mdl-32047126

ABSTRACT

Venezuelan and western equine encephalitis viruses (VEEV and WEEV, respectively) invade the central nervous system (CNS) early during infection, via neuronal and hematogenous routes. While viral replication mediates host shutoff, including expression of type I interferons (IFN), few studies have addressed how alphaviruses gain access to the CNS during established infection or the mechanisms of viral crossing at the blood-brain barrier (BBB). Here, we show that hematogenous dissemination of VEEV and WEEV into the CNS occurs via caveolin-1 (Cav-1)-mediated transcytosis (Cav-MT) across an intact BBB, which is impeded by IFN and inhibitors of RhoA GTPase. Use of reporter and nonreplicative strains also demonstrates that IFN signaling mediates viral restriction within cells comprising the neurovascular unit (NVU), differentially rendering brain endothelial cells, pericytes, and astrocytes permissive to viral replication. Transmission and immunoelectron microscopy revealed early events in virus internalization and Cav-1 association within brain endothelial cells. Cav-1-deficient mice exhibit diminished CNS VEEV and WEEV titers during early infection, whereas viral burdens in peripheral tissues remained unchanged. Our findings show that alphaviruses exploit Cav-MT to enter the CNS and that IFN differentially restricts this process at the BBB.IMPORTANCE VEEV, WEEV, and eastern equine encephalitis virus (EEEV) are emerging infectious diseases in the Americas, and they have caused several major outbreaks in the human and horse population during the past few decades. Shortly after infection, these viruses can infect the CNS, resulting in severe long-term neurological deficits or death. Neuroinvasion has been associated with virus entry into the CNS directly from the bloodstream; however, the underlying molecular mechanisms have remained largely unknown. Here, we demonstrate that following peripheral infection alphavirus augments vesicular formation/trafficking at the BBB and utilizes Cav-MT to cross an intact BBB, a process regulated by activators of Rho GTPases within brain endothelium. In vivo examination of early viral entry in Cav-1-deficient mice revealed significantly lower viral burdens in the brain than in similarly infected wild-type animals. These studies identify a potentially targetable pathway to limit neuroinvasion by alphaviruses.


Subject(s)
Blood-Brain Barrier/virology , Caveolae/virology , Encephalitis Virus, Venezuelan Equine/physiology , Encephalitis Virus, Western Equine/physiology , Transcytosis , Virus Internalization , Animals , Caveolin 1/genetics , Cell Line , Central Nervous System/virology , Endothelial Cells/virology , Male , Mice, Inbred C57BL , Virus Replication
10.
Neuron ; 103(5): 771-783, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31487528

ABSTRACT

CNS infections continue to rise in incidence in conjunction with increases in immunocompromised populations or conditions that contribute to the emergence of pathogens, such as global travel, climate change, and human encroachment on animal territories. The severity and complexity of these diseases is impacted by the diversity of etiologic agents and their routes of neuroinvasion. In this review, we present historical, clinical, and molecular concepts regarding the mechanisms of pathogen invasion of the CNS. We also discuss the structural components of CNS compartments that influence pathogen entry and recent discoveries of the pathways exploited by pathogens to facilitate CNS infections. Advances in our understanding of the CNS invasion mechanisms of different neurotropic pathogens may enable the development of strategies to control their entry and deliver drugs to mitigate established infections.


Subject(s)
Axonal Transport , Blood-Brain Barrier/microbiology , Central Nervous System Infections/microbiology , Leukocytes/microbiology , Transcytosis , Transendothelial and Transepithelial Migration , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Central Nervous System Infections/physiopathology , Humans
11.
Immunology ; 155(1): 112-122, 2018 09.
Article in English | MEDLINE | ID: mdl-29749614

ABSTRACT

Innate immune cells are integral to the pathogenesis of several diseases of the central nervous system (CNS), including multiple sclerosis (MS). Dendritic cells (DCs) are potent CD11c+ antigen-presenting cells that are critical regulators of adaptive immune responses, particularly in autoimmune diseases such as MS. The regulation of DC function in both the periphery and CNS compartment has not been fully elucidated. One limitation to studying the role of CD11c+ DCs in the CNS is that microglia can upregulate CD11c during inflammation, making it challenging to distinguish bone marrow-derived DCs (BMDCs) from microglia. Selective expression of microRNAs (miRNAs) has been shown to distinguish populations of innate cells and regulate their function within the CNS during neuro-inflammation. Using the experimental autoimmune encephalomyelitis (EAE) murine model of MS, we characterized the expression of miRNAs in CD11c+ cells using a non-biased murine array. Several miRNAs, including miR-31, were enriched in CD11c+ cells within the CNS during EAE, but not LysM+ microglia. Moreover, to distinguish CD11c+ DCs from microglia that upregulate CD11c, we generated bone marrow chimeras and found that miR-31 expression was specific to BMDCs. Interestingly, miR-31-binding sites were enriched in mRNAs downregulated in BMDCs that migrated into the CNS, and a subset was confirmed to be regulated by miR-31. Finally, miR-31 was elevated in DCs migrating through an in vitro blood-brain barrier. Our findings suggest miRNAs, including miR-31, may regulate entry of DCs into the CNS during EAE, and could potentially represent therapeutic targets for CNS autoimmune diseases such as MS.


Subject(s)
Central Nervous System/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , MicroRNAs/immunology , Multiple Sclerosis/immunology , Animals , Dendritic Cells/cytology , Disease Models, Animal , Inflammation/immunology , Mice , Mice, Inbred C57BL
12.
J Neuroimmunol ; 308: 118-130, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28501330

ABSTRACT

Viral infections of the central nervous system (CNS) are often associated with blood-brain barrier (BBB) disruption, yet the impact of virus replication and immune cell recruitment on BBB integrity are incompletely understood. Using two-photon microscopy, we demonstrate that Venezuelan equine encephalitis virus (VEEV) strain TC83-GFP, a GFP expressing, attenuated strain with a G3A mutation within the 5' UTR that is associated with increased sensitivity to type I interferons (IFNs), does not directly impact BBB permeability. Following intranasal infection of both wild-type and IFN-induced protein with tetratricopeptide repeats 1 (IFIT1)-deficient mice, which fail to block TC83-specific RNA translation, virus spreads to the olfactory bulb and cortex via migration along axonal tracts of neurons originating from the olfactory neuroepithelium. Global dissemination of virus in the CNS by 2days post-infection (dpi) was associated with increased BBB permeability in the olfactory bulb, but not in the cortex or hindbrain, where permeability only increased after the recruitment of CX3CR1+ and CCR2+ mononuclear cells on 6 dpi, which corresponded with tight junction loss and claudin 5 redistribution. Importantly, despite higher levels of viral replication, similar results were obtained in IFIT1-deficient mice. These findings indicate that TC83 gains CNS access via anterograde axonal migration without directly altering BBB function and that mononuclear and endothelial cell interactions may underlie BBB disruption during alphavirus encephalitis.


Subject(s)
Alphavirus Infections/pathology , Blood-Brain Barrier/physiopathology , Brain/metabolism , Brain/virology , Virus Replication/physiology , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/metabolism , Alphavirus Infections/genetics , Animals , Animals, Newborn , Blood-Brain Barrier/ultrastructure , Blood-Brain Barrier/virology , CX3C Chemokine Receptor 1 , Capillary Permeability/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cricetinae , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/physiology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Epithelial Cells/ultrastructure , Epithelial Cells/virology , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Virus Internalization
13.
J Biol Chem ; 292(24): 9906-9918, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28432124

ABSTRACT

The R7 regulator of G protein signaling family (R7-RGS) critically regulates nervous system development and function. Mice lacking all R7-RGS subtypes exhibit diverse neurological phenotypes, and humans bearing mutations in the retinal R7-RGS isoform RGS9-1 have vision deficits. Although each R7-RGS subtype forms heterotrimeric complexes with Gß5 and R7-RGS-binding protein (R7BP) that regulate G protein-coupled receptor signaling by accelerating deactivation of Gi/o α-subunits, several neurological phenotypes of R7-RGS knock-out mice are not readily explained by dysregulated Gi/o signaling. Accordingly, we used tandem affinity purification and LC-MS/MS to search for novel proteins that interact with R7-RGS heterotrimers in the mouse brain. Among several proteins detected, we focused on Gα13 because it had not been linked to R7-RGS complexes before. Split-luciferase complementation assays indicated that Gα13 in its active or inactive state interacts with R7-RGS heterotrimers containing any R7-RGS isoform. LARG (leukemia-associated Rho guanine nucleotide exchange factor (GEF)), PDZ-RhoGEF, and p115RhoGEF augmented interaction between activated Gα13 and R7-RGS heterotrimers, indicating that these effector RhoGEFs can engage Gα13·R7-RGS complexes. Because Gα13/R7-RGS interaction required R7BP, we analyzed phenotypes of neuronal cell lines expressing RGS7 and Gß5 with or without R7BP. We found that neurite retraction evoked by Gα12/13-dependent lysophosphatidic acid receptors was augmented in R7BP-expressing cells. R7BP expression blunted neurite formation evoked by serum starvation by signaling mechanisms involving Gα12/13 but not Gαi/o These findings provide the first evidence that R7-RGS heterotrimers interact with Gα13 to augment signaling pathways that regulate neurite morphogenesis. This mechanism expands the diversity of functions whereby R7-RGS complexes regulate critical aspects of nervous system development and function.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurons/metabolism , RGS Proteins/metabolism , Amino Acid Substitution , Animals , Brain/cytology , Brain/enzymology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , GTP-Binding Protein alpha Subunits, G12-G13/chemistry , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurites/enzymology , Neurons/cytology , Neurons/enzymology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , RGS Proteins/chemistry , RGS Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction
14.
Neurotherapeutics ; 13(3): 514-34, 2016 07.
Article in English | MEDLINE | ID: mdl-27220616

ABSTRACT

Arboviruses are arthropod-borne viruses that exhibit worldwide distribution, contributing to systemic and neurologic infections in a variety of geographical locations. Arboviruses are transmitted to vertebral hosts during blood feedings by mosquitoes, ticks, biting flies, mites, and nits. While the majority of arboviral infections do not lead to neuroinvasive forms of disease, they are among the most severe infectious risks to the health of the human central nervous system. The neurologic diseases caused by arboviruses include meningitis, encephalitis, myelitis, encephalomyelitis, neuritis, and myositis in which virus- and immune-mediated injury may lead to severe, persisting neurologic deficits or death. Here we will review the major families of emerging arboviruses that cause neurologic infections, their neuropathogenesis and host neuroimmunologic responses, and current strategies for treatment and prevention of neurologic infections they cause.


Subject(s)
Arboviruses/physiology , Encephalitis, Arbovirus/diagnosis , Encephalitis, Arbovirus/physiopathology , Animals , Arboviruses/immunology , Brain/virology , Disease Models, Animal , Encephalitis, Arbovirus/complications , Encephalitis, Arbovirus/immunology , Humans , Immunity, Innate , Mice , Neuroglia/virology , Neurons/virology
15.
Acad Forensic Pathol ; 6(1): 103-108, 2016 Mar.
Article in English | MEDLINE | ID: mdl-31239877

ABSTRACT

INTRODUCTION: Autopsy reports are often complex, with ample opportunity for errors and inconsistencies. These reports are often scrutinized by both families and attorneys. Identification of errors by proofreading physicians or clerical staff can be improved by utilizing a computer program to examine reports for discrepancies. METHODS: A webpage to review demographic consistency, organ descriptions, and pertinent information regarding gunshot wounds was developed to proofread reports. Thirty completed reports were analyzed from the Jefferson County Coroner/Medical Examiner Office. Additionally, a separate individual was instructed to sabotage reports and then determine if the software could detect the alterations. RESULTS: Of the 30 completed reports analyzed, no errors/omissions were identified; however, these reports were accurate upon manual inspection. Rarely, the computer triggered a warning that an organ should be confirmed if the author used a description that differed from the software's vocabulary (e.g., the author discussed "uterine wall" instead of "uterus"). The webpage detected eight out of ten errors supplied to the five sabotaged cases. These errors ranged from inconsistent age, race, and gender, to incomplete gunshot wound descriptions. CONCLUSION: Identification of errors by a computer proofreading program can improve autopsy report quality. The webpage has been designed so that additional modules, such as strangulation proofreading, could easily be added. Furthermore, the ability of the software to detect errors will continue to improve as more words are added to its vocabulary. The webpage is freely available and can be adapted to other medical examiner offices needs from the GitHub website.

16.
Acad Forensic Pathol ; 6(2): 281-290, 2016 Jun.
Article in English | MEDLINE | ID: mdl-31239899

ABSTRACT

INTRODUCTION: Drug use is a constantly evolving public health challenge. We present the use of Google Fusion Tables and Google Maps for the surveillance of drug-related deaths and discuss its potential large-scale use. METHODS: Demographic and geographic data for deaths related to cocaine and heroin use occurring from 2012-2014 was queried from the Jefferson County Coroner/Medical Examiner's Office. Fusion Tables was then used to plot each address onto a map. RESULTS: We developed a method to automatically import data into Fusion Tables. Individualized icons were then used to denote specific demographics and locations on a Google Map where filters could be applied to visualize findings such as only 18-25 year old white males. These features provided an easy means to visualize the relative high frequency of heroin-related deaths in white males. Furthermore, the date filters highlighted heroin-related deaths more than doubled in 2014 compared to previous years. CONCLUSIONS: Mapping of health-related data has a long-standing history in public health, though its use in the forensic pathology community is limited. Fusion Tables provides users with the ability to visualize and analyze patterns and share data. Many geocoding solutions exist; however, we found this software to be user friendly and flexible. Our office now includes these visualizations into the annual summary that is provided to local law enforcement, public health officials, and the general public.

18.
Pediatr Dev Pathol ; 18(3): 203-9, 2015.
Article in English | MEDLINE | ID: mdl-25634794

ABSTRACT

Fetal and infant autopsy yields information regarding cause of death and the risk of recurrence, and it provides closure for parents. A significant number of perinatal evaluations are performed by general practice pathologists or trainees, who often find them time-consuming and/or intimidating. We sought to create a program that would enable pathologists to conduct these examinations with greater ease and to produce reliable, informative reports. We developed software that automatically generates a set of expected anthropometric and organ weight ranges by gestational age (GA)/postnatal age (PA) and a correlative table with the GA/PA that best matches the observed anthropometry. The program highlights measurement and organ weight discrepancies, enabling users to identify abnormalities. Furthermore, a Web page provides options for exporting and saving the data. Pathology residents utilized the program to determine ease of usage and benefits. The average time using conventional methods (ie, reference books and Internet sites) was compared to the average time using our Web page. Average time for novice and experienced residents using conventional methods was 26.7 minutes and 15 minutes, respectively. Using the Web page program, these times were reduced to an average of 3.2 minutes (P < 0.046 and P < 0.02, respectively). Participants found our program simple to use and the corrective features beneficial. This novel application saves time and improves the quality of fetal and infant autopsy reports. The software allows data exportation to reports and data storage for future analysis. Finalization of our software to enable usage by both university and private practice groups is in progress.


Subject(s)
Anthropometry/methods , Autopsy/methods , Pathology, Clinical/methods , Software , Fetus , Gestational Age , Humans , Infant, Newborn , Organ Size
19.
PLoS One ; 8(12): e82276, 2013.
Article in English | MEDLINE | ID: mdl-24349243

ABSTRACT

In the outer retina, G protein-coupled receptor (GPCR) signaling mediates phototransduction and synaptic transmission between photoreceptors and ON bipolar cells. In contrast, the functions of modulatory GPCR signaling networks in the inner retina are less well understood. We addressed this question by determining the consequences of augmenting modulatory Gi/o signaling driven by endogenous transmitters. This was done by analyzing the effects of genetically ablating the R7 RGS-binding protein (R7BP), a membrane-targeting protein and positive allosteric modulator of R7-RGS (regulator of the G protein signaling 7) family that deactivates Gi/oα subunits. We found that R7BP is expressed highly in starburst amacrine cells and retinal ganglion cells (RGCs). As indicated by electroretinography and multielectrode array recordings of adult retina, ablation of R7BP preserved outer retina function, but altered the firing rate and latency of ON RGCs driven by rods and cones but not rods alone. In developing retina, R7BP ablation increased the burst duration of glutamatergic waves whereas cholinergic waves were unaffected. This effect on glutamatergic waves did not result in impaired segregation of RGC projections to eye-specific domains of the dorsal lateral geniculate nucleus. R7BP knockout mice exhibited normal spatial contrast sensitivity and visual acuity as assessed by optomotor reflexes. Taken together these findings indicate that R7BP-dependent regulation of R7-RGS proteins shapes specific aspects of light-evoked and spontaneous activity of RGCs in mature and developing retina.


Subject(s)
Glutamates/metabolism , Light , RGS Proteins/metabolism , Retina/metabolism , Retina/radiation effects , Action Potentials/radiation effects , Allosteric Regulation/radiation effects , Amacrine Cells/metabolism , Amacrine Cells/radiation effects , Animals , Animals, Newborn , Choline/metabolism , Contrast Sensitivity/radiation effects , Gene Deletion , Mice , Mice, Inbred C57BL , RGS Proteins/deficiency , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/radiation effects , Retinal Cone Photoreceptor Cells/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Retinal Rod Photoreceptor Cells/metabolism , Transcription Factor Brn-3A/metabolism , Visual Acuity/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...