Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Cureus ; 15(3): e36787, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36998916

ABSTRACT

Familial hemophagocytic lymphohistiocytosis is a rare and potentially life-threatening genetic condition characterized by unsuppressed immune activation and hypercytokinemia. Chronic Lymphocytic Inflammation with Pontine Perivascular Enhancement Responsive to Steroids (CLIPPERS) is a central nervous system inflammatory disorder characterized by punctate and curvilinear gadolinium-enhancing lesions in the brainstem, cerebellum, and spinal cord, which responds well to corticosteroid treatment. Hemophagocytic lymphohistiocytosis has been known to mimic CLIPPERS on neuroimaging, and patients previously diagnosed with CLIPPERS may carry familial hemophagocytic lymphohistiocytosis-related gene mutations that serve as predisposing factors. In this article, we describe a case initially diagnosed with CLIPPERS based on characteristic magnetic resonance imaging features and clinical course, who was later diagnosed with hemophagocytic lymphohistiocytosis based on a heterozygous familial hemophagocytic lymphohistiocytosis-associated PRF1 gene mutation.

2.
Radiol Case Rep ; 18(3): 1334-1336, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36704365

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a rare, often fatal, demyelinating disease of the central nervous system. The disease almost exclusively presents in immunosuppressed patients, such as those with acquired immunodeficiency syndrome, a hematopoietic malignancy, or a transplanted organ; it is extremely rare in patients without immunosuppression. We present a case of a 74-year-old female with radiographic and histopathological findings consistent with PML that possibly arose in the setting of Sjögren's-related vasculitis but no immunosuppression.

3.
Res Social Adm Pharm ; 18(12): 4138-4143, 2022 12.
Article in English | MEDLINE | ID: mdl-35871146

ABSTRACT

BACKGROUND: In response to the COVID-19 pandemic, the CDC issued guidance advising patients and providers to adopt social distancing practices such as home-based infusions (H-BI). METHODS: We performed a mixed methods evaluation to summarize perceptions, concerns, and experiences with H-BI among all inflammatory bowel disease patients 18-90 years of age who transitioned to home-based infliximab or vedolizumab infusions between March to July 2020 at a tertiary care center. Semi-structured interviews were conducted and analyzed using an iterative, inductive thematic approach. Baseline characteristics and outcome on safety, COVID-19 transmission, delays in infusions, and H-BI persistence were collected. RESULTS: Of the 57 participants who transitioned to H-BI, 20 (33%) responded. Four major categories and six major themes related to expectations, experience, perceived safety, and logistical factors were identified. Initial perceptions were mixed, however these resolved. One patient developed COVID-19, one patient experienced an adverse event, 12 (21%) patients experienced an infusion delay, and 6 (11%) patients transitioned from H-BI. DISCUSSION: Despite mixed initial perceptions, respondents had a positive experience with most respondents planning to continue H-BI after the pandemic resolves. Several real-world actionable barriers were identified related to scheduling, communication between stakeholders, and nursing quality. No major safety concerns were identified.


Subject(s)
COVID-19 , Inflammatory Bowel Diseases , Humans , COVID-19/epidemiology , Pandemics , Inflammatory Bowel Diseases/drug therapy , Infliximab , Chronic Disease
4.
Nat Commun ; 11(1): 3871, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32747712

ABSTRACT

Relapses in multiple sclerosis can result in irreversible nervous system tissue injury. If these events could be detected early, targeted immunotherapy could potentially slow disease progression. We describe the use of engineered biomaterial-based immunological niches amenable to biopsy to provide insights into the phenotype of innate immune cells that control disease activity in a mouse model of multiple sclerosis. Differential gene expression in cells from these niches allow monitoring of disease dynamics and gauging the effectiveness of treatment. A proactive treatment regimen, given in response to signal within the niche but before symptoms appeared, substantially reduced disease. This technology offers a new approach to monitor organ-specific autoimmunity, and represents a platform to analyze immune dysfunction within otherwise inaccessible target tissues.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Immunotherapy/methods , Monitoring, Physiologic/methods , Multiple Sclerosis/therapy , Animals , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Gene Expression Profiling/methods , Humans , Mice, Inbred Strains , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Recurrence , Treatment Outcome
5.
Clin Immunol ; 189: 29-33, 2018 04.
Article in English | MEDLINE | ID: mdl-27664933

ABSTRACT

Eotaxins are C-C motif chemokines first identified as potent eosinophil chemoattractants. They facilitate eosinophil recruitment to sites of inflammation in response to parasitic infections as well as allergic and autoimmune diseases such as asthma, atopic dermatitis, and inflammatory bowel disease. The eotaxin family currently includes three members: eotaxin-1 (CCL11), eotaxin-2 (CCL24), and eotaxin-3 (CCL26). Despite having only ~30% sequence homology to one another, each was identified based on its ability to bind the chemokine receptor, CCR3. Beyond their role in innate immunity, recent studies have shown that CCL11 and related molecules may directly contribute to degenerative processes in the central nervous system (CNS). CCL11 levels increase in the plasma and cerebrospinal fluid of both mice and humans as part of normal aging. In mice, these increases are associated with declining neurogenesis and impaired cognition and memory. In humans, elevated plasma levels of CCL11 have been observed in Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease, and secondary progressive multiple sclerosis when compared to age-matched, healthy controls. Since CCL11 is capable of crossing the blood-brain barrier of normal mice, it is plausible that eotaxins generated in the periphery may exert physiological and pathological actions in the CNS. Here, we briefly review known functions of eotaxin family members during innate immunity, and then focus on whether and how these molecules might participate in the progression of neurodegenerative diseases.


Subject(s)
Chemokine CCL11/immunology , Chemokine CCL24/immunology , Chemokine CCL26/immunology , Immunity, Innate/immunology , Neurodegenerative Diseases/immunology , Aging/immunology , Animals , Chemokine CCL11/blood , Chemokine CCL11/cerebrospinal fluid , Chemokine CCL24/blood , Chemokine CCL24/cerebrospinal fluid , Chemokine CCL26/blood , Chemokine CCL26/cerebrospinal fluid , Humans , Neurodegenerative Diseases/blood , Neurodegenerative Diseases/cerebrospinal fluid , Receptors, CCR3/immunology , Receptors, CCR3/metabolism
6.
Ann Clin Transl Neurol ; 4(2): 145-150, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28168214

ABSTRACT

Oxidative damage occurs in multiple sclerosis, but is difficult to identify antemortem and remains an unknown contributor to disease progression. Carbonylation is a quantitative measure of protein oxidation. Cerebrospinal fluid samples from multiple sclerosis patients showed elevated carbonylated protein levels compared to controls. In experimental autoimmune encephalomyelitis, carbonylated protein levels in cerebrospinal fluid correlated tightly with those found in inflamed spinal cord tissues. Furthermore, concentrations in cerebrospinal fluid and spinal cord responded in parallel to an antioxidant intervention that also attenuated disease symptoms. These data suggest that carbonylated cerebrospinal fluid proteins could be a quantitative, sensitive, and disease-relevant biomarker in multiple sclerosis.

7.
J Neuroinflammation ; 13(1): 196, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27562117

ABSTRACT

BACKGROUND: Alphaviruses can cause fatal encephalitis in humans. Natural infections occur via the bite of infected mosquitos, but aerosol transmissibility makes some of these viruses potential bioterrorism agents. Central nervous system (CNS) host responses contribute to alphavirus pathogenesis in experimental models and are logical therapeutic targets. We investigated whether reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity within the CNS contributes to fatal alphavirus encephalitis in mice. METHODS: Infected animals were treated systemically with the angiotensin receptor-blocking drug, telmisartan, given its ability to cross the blood-brain barrier, selectively block type-1 angiotensin receptors (AT1R), and inhibit Nox-derived ROS production in vascular smooth muscle and other extraneural tissues. Clinical, virological, biochemical, and histopathological outcomes were followed over time. RESULTS: The importance of the angiotensin II (Ang II)/AT1R axis in disease pathogenesis was confirmed by demonstrating increased Ang II levels in the CNS following infection, enhanced disease survival when CNS Ang II production was suppressed, increased AT1R expression on microglia and tissue-infiltrating myeloid cells, and enhanced disease survival in AT1R-deficient mice compared to wild-type (WT) controls. Systemic administration of telmisartan protected WT mice from lethal encephalitis caused by two different alphaviruses in a dose-dependent manner without altering virus replication or exerting any anti-inflammatory effects in the CNS. Infection triggered up-regulation of multiple Nox subunits in the CNS, while drug treatment inhibited local Nox activity, ROS production, and oxidative neuronal damage. Telmisartan proved ineffective in Nox-deficient mice, demonstrating that this enzyme is its main target in this experimental setting. CONCLUSIONS: Nox-derived ROS, likely arising from CNS myeloid cells triggered by AT1R signaling, are pathogenic during fatal alphavirus encephalitis in mice. Systemically administered telmisartan at non-hypotensive doses targets Nox activity in the CNS to exert a neuroprotective effect. Disruption of this pathway may have broader implications for the treatment of related infections as well as for other CNS diseases driven by oxidative injury.


Subject(s)
Central Nervous System/pathology , Encephalomyelitis, Equine/pathology , Myeloid Cells/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , ATPases Associated with Diverse Cellular Activities , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Benzoates/pharmacology , CX3C Chemokine Receptor 1 , Central Nervous System/drug effects , Central Nervous System/virology , DNA Helicases/genetics , DNA Helicases/metabolism , Disease Models, Animal , Encephalomyelitis, Equine/drug therapy , Encephalomyelitis, Equine/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/classification , Myeloid Cells/ultrastructure , Myeloid Cells/virology , Neurons/pathology , Neurons/ultrastructure , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Telmisartan
8.
J Biol Chem ; 291(6): 3030-42, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26601958

ABSTRACT

UBE2W ubiquitinates N termini of proteins rather than internal lysine residues, showing a preference for substrates with intrinsically disordered N termini. The in vivo functions of this intriguing E2, however, remain unknown. We generated Ube2w germ line KO mice that proved to be susceptible to early postnatal lethality without obvious developmental abnormalities. Although the basis of early death is uncertain, several organ systems manifest changes in Ube2w KO mice. Newborn Ube2w KO mice often show altered epidermal maturation with reduced expression of differentiation markers. Mirroring higher UBE2W expression levels in testis and thymus, Ube2w KO mice showed a disproportionate decrease in weight of these two organs (~50%), suggesting a functional role for UBE2W in the immune and male reproductive systems. Indeed, Ube2w KO mice displayed sustained neutrophilia accompanied by increased G-CSF signaling and testicular vacuolation associated with decreased fertility. Proteomic analysis of a vulnerable organ, presymptomatic testis, showed a preferential accumulation of disordered proteins in the absence of UBE2W, consistent with the view that UBE2W preferentially targets disordered polypeptides. These mice further allowed us to establish that UBE2W is ubiquitously expressed as a single isoform localized to the cytoplasm and that the absence of UBE2W does not alter cell viability in response to various stressors. Our results establish that UBE2W is an important, albeit not essential, protein for early postnatal survival and normal functioning of multiple organ systems.


Subject(s)
Epidermis , Skin Abnormalities , Ubiquitin-Conjugating Enzymes , Animals , Epidermis/abnormalities , Epidermis/enzymology , Epidermis/immunology , Leukocyte Disorders/congenital , Leukocyte Disorders/enzymology , Leukocyte Disorders/genetics , Leukocyte Disorders/immunology , Male , Mice , Mice, Knockout , Skin Abnormalities/enzymology , Skin Abnormalities/genetics , Skin Abnormalities/immunology , Testis/enzymology , Testis/immunology , Thymus Gland/enzymology , Thymus Gland/immunology , Ubiquitin-Conjugating Enzymes/deficiency , Ubiquitin-Conjugating Enzymes/immunology
9.
J Clin Cell Immunol ; 7(5)2016 Oct.
Article in English | MEDLINE | ID: mdl-28603659

ABSTRACT

The chemokine, C-X-C motif ligand 13 (CXCL13), is constitutively expressed in lymphoid organs and controls the recruitment and compartmentalization of lymphocytes and antigen presenting cells within these specialized structures. Recent data, however, also show induction of this molecule under a variety of circumstances during central nervous system (CNS) inflammation. While its role(s) in the pathogenesis of neoplastic, infectious and autoimmune disorders of the CNS remain incompletely understood, growing evidence suggests that CXCL13 could become a relevant therapeutic target in at least some of these conditions. This review focuses on the diseases, cellular sources and external factors known to regulate CXCL13 production in the inflamed CNS.

10.
Front Neurosci ; 9: 344, 2015.
Article in English | MEDLINE | ID: mdl-26500475

ABSTRACT

The matricellular proteins, secreted protein acidic and rich in cysteine (SPARC) and SPARC-like 1 (SPARCL1), are produced by astrocytes and control excitatory synaptogenesis in the central nervous system. While SPARCL1 directly promotes excitatory synapse formation in vitro and in the developing nervous system in vivo, SPARC specifically antagonizes the synaptogenic actions of SPARCL1. We hypothesized these proteins also help maintain existing excitatory synapses in adult hosts, and that local inflammation in the spinal cord alters their production in a way that dynamically modulates motor synapses and impacts the severity of paralysis during experimental autoimmune encephalomyelitis (EAE) in mice. Using a spontaneously remitting EAE model, paralysis severity correlated inversely with both expression of synaptic proteins and the number of synapses in direct contact with the perikarya of motor neurons in spinal gray matter. In both remitting and non-remitting EAE models, paralysis severity also correlated inversely with sparcl1:sparc transcript and SPARCL1:SPARC protein ratios directly in lumbar spinal cord tissue. In vitro, astrocyte production of both SPARCL1 and SPARC was regulated by T cell-derived cytokines, causing dynamic modulation of the SPARCL1:SPARC expression ratio. Taken together, these data support a model whereby proinflammatory cytokines inhibit SPARCL1 and/or augment SPARC expression by astrocytes in spinal gray matter that, in turn, cause either transient or sustained synaptic retraction from lumbar spinal motor neurons thereby regulating hind limb paralysis during EAE. Ongoing studies seek ways to alter this SPARCL1:SPARC expression ratio in favor of synapse reformation/maintenance and thus help to modulate neurologic deficits during times of inflammation. This could identify new astrocyte-targeted therapies for diseases such as multiple sclerosis.

12.
Cytokine ; 74(2): 228-36, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25691330

ABSTRACT

A series of controlled clinical trials have shown that exogenous interferon-beta (IFN-ß) benefits patients with relapsing-remitting multiple sclerosis (RRMS) by reducing relapse rate, disability progression, and the formation of new brain and spinal cord lesions on magnetic resonance imaging (MRI) scans. Unfortunately, however, the effectiveness of IFN-ß is limited in this setting by the occurrence of treatment non-responsiveness in nearly 25% of patients. Furthermore, clinicians who care for RRMS patients remain unable to accurately identify IFN-ß non-responders prior to the initiation of therapy, causing delays in the use of alternative treatments and sometimes requiring that patients turn to medications with more significant side effects to control their disease. Progress has been made toward understanding how both endogenous and exogenous IFN-ß act to slow RRMS as well as the related mouse model, experimental autoimmune encephalomyelitis (EAE). Most studies point to its inhibitory actions on circulating immune cells as being important for suppressing both disorders, but multiple potential target cells and inflammatory pathways have been implicated and those essential to confer its benefits remain undefined. This review focuses on the role of both endogenous and exogenous IFN-ß in RRMS, paying particular attention to the issue of why certain individuals appear refractory to its disease-modifying effects. A continued goal in this field remains the identification of a convenient biomarker that accurately predicts IFN-ß treatment non-responsiveness in individual RRMS patients. Development of such an assay will allow clinicians to customize therapy for patients with this complex disorder.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Interferon-beta , Multiple Sclerosis , Animals , Clinical Trials as Topic , Encephalomyelitis, Autoimmune, Experimental/diagnostic imaging , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Interferon-beta/immunology , Interferon-beta/therapeutic use , Mice , Mice, Knockout , Multiple Sclerosis/diagnostic imaging , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Radiography
13.
J Neurovirol ; 21(1): 43-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25361697

ABSTRACT

While alphaviruses spread naturally via mosquito vectors, some can also be transmitted as aerosols making them potential bioterrorism agents. One such pathogen, western equine encephalitis virus (WEEV), causes fatal human encephalitis via multiple routes of infection and thus presumably via multiple mechanisms. Although WEEV also produces acute encephalitis in non-human primates, a small animal model that recapitulates features of human disease would be useful for both pathogenesis studies and to evaluate candidate antiviral therapies. We have optimized conditions to infect mice with a low passage isolate of WEEV, thereby allowing detailed investigation of virus tropism, replication, neuroinvasion, and neurovirulence. We find that host factors strongly influence disease outcome, and in particular, that age, gender, and genetic background all have significant effects on disease susceptibility independent of virus tropism or replication within the central nervous system. Our data show that experimental variables can be adjusted in mice to recapitulate disease features known to occur in both non-human primates and humans, thus aiding further study of WEEV pathogenesis and providing a realistic therapeutic window for antiviral drug delivery.


Subject(s)
Alphavirus Infections/pathology , Encephalitis Virus, Western Equine/pathogenicity , RNA, Viral/blood , Seizures/pathology , Administration, Intranasal , Alphavirus Infections/virology , Animals , Behavior, Animal , Cognition , Disease Models, Animal , Encephalitis Virus, Western Equine/physiology , Host Specificity , Injections, Intraperitoneal , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Seizures/virology , Species Specificity , Viral Load , Virus Replication
14.
Adv Neuroimmune Biol ; 6(1): 1-8, 2015.
Article in English | MEDLINE | ID: mdl-26855687

ABSTRACT

The chemokine, C-X-C motif ligand 13 (CXCL13), is constitutively expressed in lymphoid organs and controls the recruitment and compartmentalization of lymphocytes and antigen presenting cells within these specialized structures. Recent data, however, also find induction of this molecule during central nervous system (CNS) inflammation under a variety of circumstances. While its role(s) in the pathogenesis of neoplastic, infectious and autoimmune disorders of the CNS remain incompletely understood, several lines of evidence suggest that CXCL13 could become a relevant therapeutic target in at least some of these diseases. This review focuses on how CXCL13 contributes to the pathogenesis of selected CNS disorders involving both experimental animals and humans, paying particular attention to the issue of whether (and if so, how) blockade of this ligand or its receptor might benefit the host. Current blocking strategies largely involve the use of monoclonal antibodies, but an improved understanding of downstream signaling pathways makes small molecule inhibition a future possibility.

15.
Front Oncol ; 4: 328, 2014.
Article in English | MEDLINE | ID: mdl-25431758

ABSTRACT

The central nervous system (CNS), once viewed as an immune-privileged site protected by the blood-brain barrier (BBB), is now known to be a dynamic immunological environment through which immune cells migrate to prevent and respond to events such as localized infection. During these responses, endogenous glial cells, including astrocytes and microglia, become highly reactive and may secrete inflammatory mediators that regulate BBB permeability and recruit additional circulating immune cells. Here, we discuss the various roles played by astrocytes, microglia, and infiltrating immune cells during host immunity to non-tumor antigens in the CNS, focusing first on bacterial and viral infections, and then turning to responses directed against self-antigens in the setting of CNS autoimmunity.

16.
Neurology ; 83(17): 1500-7, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25253754

ABSTRACT

OBJECTIVE: In the current exploratory study, we longitudinally measured immune parameters in the blood of individuals with relapsing-remitting multiple sclerosis (RRMS) and secondary progressive multiple sclerosis (SPMS), and investigated their relationship to disease duration and clinical and radiologic measures of CNS injury. METHODS: Peripheral blood mononuclear cells (PBMCs) and plasma were obtained from subjects with RRMS, SPMS, and from healthy controls on a monthly basis over the course of 1 year. MRI and Expanded Disability Status Scale evaluations were performed serially. PBMCs were analyzed by enzyme-linked immunosorbent spot assay to enumerate myelin basic protein-specific interleukin (IL)-17- and interferon (IFN)-γ-producing cells. Plasma concentrations of proinflammatory factors were measured using customized Luminex panels. RESULTS: Frequencies of myelin basic protein-specific IL-17- and IFN-γ-producing PBMCs were higher in individuals with RRMS and SPMS compared to healthy controls. Patients with SPMS expressed elevated levels of IL-17-inducible chemokines that activate and recruit myeloid cells. In the cohort of patients with SPMS without inflammatory activity, upregulation of myeloid-related factors correlated directly with MRI T2 lesion burden and inversely with brain parenchymal tissue volume. CONCLUSIONS: The results of this exploratory study raise the possibility that Th17 responses and IL-17-inducible myeloid factors are elevated during SPMS compared with RRMS, and correlate with lesion burden. Our data endorse further investigation of Th17- and myeloid-related factors as candidate therapeutic targets in SPMS.


Subject(s)
Cytokines/blood , Leukocytes, Mononuclear/metabolism , Multiple Sclerosis, Chronic Progressive/blood , Multiple Sclerosis, Chronic Progressive/pathology , Myelin Basic Protein/blood , Adult , Aged , Disability Evaluation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interferon-gamma/blood , Interleukin-17/blood , Interleukin-23/blood , Leukocytes, Mononuclear/pathology , Longitudinal Studies , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/blood
17.
J Virol ; 88(19): 11199-214, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25031353

ABSTRACT

Neurotropic alphaviruses, including western, eastern, and Venezuelan equine encephalitis viruses, cause serious and potentially fatal central nervous system infections in humans for which no currently approved therapies exist. We previously identified a series of thieno[3,2-b]pyrrole derivatives as novel inhibitors of neurotropic alphavirus replication, using a cell-based phenotypic assay (W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275), and subsequently developed second- and third-generation indole-2-carboxamide derivatives with improved potency, solubility, and metabolic stability (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). In this report, we describe the antiviral activity of the most promising third-generation lead compound, CCG205432, and closely related analogs CCG206381 and CCG209023. These compounds have half-maximal inhibitory concentrations of ∼1 µM and selectivity indices of >100 in cell-based assays using western equine encephalitis virus replicons. Furthermore, CCG205432 retains similar potency against fully infectious virus in cultured human neuronal cells. These compounds show broad inhibitory activity against a range of RNA viruses in culture, including members of the Togaviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Although their exact molecular target remains unknown, mechanism-of-action studies reveal that these novel indole-based compounds target a host factor that modulates cap-dependent translation. Finally, we demonstrate that both CCG205432 and CCG209023 dampen clinical disease severity and enhance survival of mice given a lethal western equine encephalitis virus challenge. These studies demonstrate that indole-2-carboxamide compounds are viable candidates for continued preclinical development as inhibitors of neurotropic alphaviruses and, potentially, of other RNA viruses. IMPORTANCE There are currently no approved drugs to treat infections with alphaviruses. We previously identified a novel series of compounds with activity against these potentially devastating pathogens (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). We have now produced third-generation compounds with enhanced potency, and this manuscript provides detailed information on the antiviral activity of these advanced-generation compounds, including activity in an animal model. The results of this study represent a notable achievement in the continued development of this novel class of antiviral inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Encephalitis Virus, Western Equine/drug effects , Encephalomyelitis, Equine/drug therapy , Indoles/pharmacology , Pyridines/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/chemical synthesis , Bunyaviridae/drug effects , Bunyaviridae/growth & development , Cell Line , Encephalitis Virus, Western Equine/growth & development , Encephalitis Virus, Western Equine/pathogenicity , Encephalomyelitis, Equine/mortality , Encephalomyelitis, Equine/virology , Female , Indoles/chemical synthesis , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/virology , Paramyxoviridae/drug effects , Paramyxoviridae/growth & development , Picornaviridae/drug effects , Picornaviridae/growth & development , Protein Biosynthesis/drug effects , Pyridines/chemical synthesis , Replicon/drug effects , Structure-Activity Relationship , Survival Analysis
18.
Glia ; 62(9): 1452-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24829092

ABSTRACT

Lymphoid chemokines are crucial for the development and maintenance of lymphoid organs, but their ectopic expression in non-lymphoid tissues is implicated in both local response to infection and chronic organ-specific autoimmunity. Production of one such chemokine, C-X-C motif ligand 13 (CXCL13), within the central nervous system (CNS) has been linked to the pathogenesis of multiple sclerosis (MS), although little is known about factors controlling its expression in different neural cell types and across a range of disease states. We provoked acute neuroinflammation in experimental animals without causing any associated demyelination using neuroadapted Sindbis virus (NSV) to better understand the sources and regulators of this chemokine in the CNS. We found that mice genetically deficient in the transcription factor, interferon (IFN) regulatory factor-7 (IRF7), made significantly higher CXCL13 protein levels in the CNS compared with wild-type (WT) controls. Microglia proved to be the main producer of CXCL13 in the brain during infection of both WT and IRF7(-/-) mice, and primary microglia cultured in vitro generated CXCL13 following stimulation with either virus particles or synthetic Toll-like receptor (TLR) ligands. Microglia cultured from IRF7(-/-) mice selectively overproduced CXCL13, and manipulation of extracellular type-I IFN levels demonstrated the existence of a negative feedback loop whereby type-I IFN receptor signaling specifically suppressed microglial CXCL13 release. Since IFN-ß is used to treat patients with relapsing-remitting MS and yet acts through unknown mechanisms, we speculate that suppressed lymphoid chemokine production by microglia could contribute to its therapeutic effects.


Subject(s)
Brain/immunology , Chemokine CXCL13/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Type I/metabolism , Microglia/metabolism , Alphavirus Infections/immunology , Animals , Cells, Cultured , Chemokine CXCL13/genetics , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalomyelitis/immunology , Interferon Regulatory Factor-7/genetics , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation/physiology , Sindbis Virus , Toll-Like Receptors/metabolism
19.
PLoS One ; 8(12): e82318, 2013.
Article in English | MEDLINE | ID: mdl-24349254

ABSTRACT

Natural products provide a vast array of chemical structures to explore in the discovery of new medicines. Although secondary metabolites produced by microbes have been developed to treat a variety of diseases, including bacterial and fungal infections, to date there has been limited investigation of natural products with antiviral activity. In this report, we used a phenotypic cell-based replicon assay coupled with an iterative biochemical fractionation process to identify, purify, and characterize antiviral compounds produced by marine microbes. We isolated a compound from Streptomyces kaviengensis, a novel actinomycetes isolated from marine sediments obtained off the coast of New Ireland, Papua New Guinea, which we identified as antimycin A1a. This compound displays potent activity against western equine encephalitis virus in cultured cells with half-maximal inhibitory concentrations of less than 4 nM and a selectivity index of greater than 550. Our efforts also revealed that several antimycin A analogues display antiviral activity, and mechanism of action studies confirmed that these Streptomyces-derived secondary metabolites function by inhibiting the cellular mitochondrial electron transport chain, thereby suppressing de novo pyrimidine synthesis. Furthermore, we found that antimycin A functions as a broad spectrum agent with activity against a wide range of RNA viruses in cultured cells, including members of the Togaviridae, Flaviviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Finally, we demonstrate that antimycin A reduces central nervous system viral titers, improves clinical disease severity, and enhances survival in mice given a lethal challenge with western equine encephalitis virus. Our results provide conclusive validation for using natural product resources derived from marine microbes as source material for antiviral drug discovery, and they indicate that host mitochondrial electron transport is a viable target for the continued development of broadly active antiviral compounds.


Subject(s)
Actinobacteria/chemistry , Antiviral Agents/pharmacology , Geologic Sediments/microbiology , Animals , Antimycin A/chemistry , Antimycin A/pharmacology , Antimycin A/therapeutic use , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Biological Products/chemistry , Biological Products/isolation & purification , Biological Products/pharmacology , Cell Line , Central Nervous System/drug effects , Central Nervous System/pathology , Central Nervous System/virology , Chemical Fractionation , Electron Transport/drug effects , Encephalitis Viruses/drug effects , Encephalitis, Arbovirus/drug therapy , Encephalitis, Arbovirus/pathology , Encephalitis, Arbovirus/virology , High-Throughput Screening Assays , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Viral/metabolism , Reference Standards , Reproducibility of Results , Streptomyces/chemistry , Survival Analysis , Transcription, Genetic/drug effects
20.
J Med Chem ; 56(22): 9222-41, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24151954

ABSTRACT

Neurotropic alphaviruses, which include western equine encephalitis virus (WEEV) and Fort Morgan virus, are mosquito-borne pathogens that infect the central nervous system causing acute and potentially fatal encephalitis. We previously reported a novel series of indole-2-carboxamides as alphavirus replication inhibitors, one of which conferred protection against neuroadapted Sindbis virus infection in mice. We describe here further development of this series, resulting in 10-fold improvement in potency in a WEEV replicon assay and up to 40-fold increases in half-lives in mouse liver microsomes. Using a rhodamine123 uptake assay in MDR1-MDCKII cells, we were able to identify structural modifications that markedly reduce recognition by P-glycoprotein, the key efflux transporter at the blood-brain barrier. In a preliminary mouse PK study, we were able to demonstrate that two new analogues could achieve higher and/or longer plasma drug exposures than our previous lead and that one compound achieved measurable drug levels in the brain.


Subject(s)
Drug Design , Encephalitis Virus, Western Equine/drug effects , Encephalitis Virus, Western Equine/physiology , Indoles/chemistry , Indoles/pharmacology , Virus Replication/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Humans , Hydrophobic and Hydrophilic Interactions , Indoles/metabolism , Indoles/pharmacokinetics , Mice , Molecular Weight , Solubility , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...