Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Proteomics ; 22(4): 100523, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36870567

RESUMEN

Neurologic manifestations are among the most frequently reported complications of COVID-19. However, given the paucity of tissue samples and the highly infectious nature of the etiologic agent of COVID-19, we have limited information to understand the neuropathogenesis of COVID-19. Therefore, to better understand the impact of COVID-19 on the brain, we used mass-spectrometry-based proteomics with a data-independent acquisition mode to investigate cerebrospinal fluid (CSF) proteins collected from two different nonhuman primates, Rhesus Macaque and African Green Monkeys, for the neurologic effects of the infection. These monkeys exhibited minimal to mild pulmonary pathology but moderate to severe central nervous system (CNS) pathology. Our results indicated that CSF proteome changes after infection resolution corresponded with bronchial virus abundance during early infection and revealed substantial differences between the infected nonhuman primates and their age-matched uninfected controls, suggesting these differences could reflect altered secretion of CNS factors in response to SARS-CoV-2-induced neuropathology. We also observed the infected animals exhibited highly scattered data distributions compared to their corresponding controls indicating the heterogeneity of the CSF proteome change and the host response to the viral infection. Dysregulated CSF proteins were preferentially enriched in functional pathways associated with progressive neurodegenerative disorders, hemostasis, and innate immune responses that could influence neuroinflammatory responses following COVID-19. Mapping these dysregulated proteins to the Human Brain Protein Atlas found that they tended to be enriched in brain regions that exhibit more frequent injury following COVID-19. It, therefore, appears reasonable to speculate that such CSF protein changes could serve as signatures for neurologic injury, identify important regulatory pathways in this process, and potentially reveal therapeutic targets to prevent or attenuate the development of neurologic injuries following COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Humanos , Chlorocebus aethiops , Proteínas del Líquido Cefalorraquídeo , Proteoma , Macaca mulatta
2.
Am J Pathol ; 191(2): 274-282, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33171111

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces a wide range of disease severity, ranging from asymptomatic infection to a life-threating illness, particularly in the elderly population and individuals with comorbid conditions. Among individuals with serious coronavirus 2019 (COVID-19) disease, acute respiratory distress syndrome (ARDS) is a common and often fatal presentation. Animal models of SARS-CoV-2 infection that manifest severe disease are needed to investigate the pathogenesis of COVID-19-induced ARDS and evaluate therapeutic strategies. We report two cases of ARDS in two aged African green monkeys (AGMs) infected with SARS-CoV-2 that had pathological lesions and disease similar to severe COVID-19 in humans. We also report a comparatively mild COVID-19 phenotype characterized by minor clinical, radiographic, and histopathologic changes in the two surviving, aged AGMs and four rhesus macaques (RMs) infected with SARS-CoV-2. Notable increases in circulating cytokines were observed in three of four infected, aged AGMs but not in infected RMs. All the AGMs had increased levels of plasma IL-6 compared with baseline, a predictive marker and presumptive therapeutic target in humans infected with SARS-CoV-2. Together, our results indicate that both RMs and AGMs are capable of modeling SARS-CoV-2 infection and suggest that aged AGMs may be useful for modeling severe disease manifestations, including ARDS.


Asunto(s)
COVID-19/etiología , Pulmón/virología , SARS-CoV-2/patogenicidad , Envejecimiento , Animales , Chlorocebus aethiops/virología , Infecciones por Coronavirus/tratamiento farmacológico , Citocinas/metabolismo , Humanos , Pulmón/patología , Macaca mulatta/virología , Carga Viral/métodos
3.
Am J Respir Cell Mol Biol ; 64(1): 79-88, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32991819

RESUMEN

Preclinical mouse models that recapitulate some characteristics of coronavirus disease (COVID-19) will facilitate focused study of pathogenesis and virus-host responses. Human agniotensin-converting enzyme 2 (hACE2) serves as an entry receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to infect people via binding to envelope spike proteins. Herein we report development and characterization of a rapidly deployable COVID-19 mouse model. C57BL/6J (B6) mice expressing hACE2 in the lung were transduced by oropharyngeal delivery of the recombinant human adenovirus type 5 that expresses hACE2 (Ad5-hACE2). Mice were infected with SARS-CoV-2 at Day 4 after transduction and developed interstitial pneumonia associated with perivascular inflammation, accompanied by significantly higher viral load in lungs at Days 3, 6, and 12 after infection compared with Ad5-empty control group. SARS-CoV-2 was detected in pneumocytes in alveolar septa. Transcriptomic analysis of lungs demonstrated that the infected Ad5-hACE mice had a significant increase in IFN-dependent chemokines Cxcl9 and Cxcl10, and genes associated with effector T-cell populations including Cd3 g, Cd8a, and Gzmb. Pathway analysis showed that several Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were enriched in the data set, including cytokine-cytokine receptor interaction, the chemokine signaling pathway, the NOD-like receptor signaling pathway, the measles pathway, and the IL-17 signaling pathway. This response is correlative to clinical response in lungs of patients with COVID-19. These results demonstrate that expression of hACE2 via adenovirus delivery system sensitized the mouse to SARS-CoV-2 infection and resulted in the development of a mild COVID-19 phenotype, highlighting the immune and inflammatory host responses to SARS-CoV-2 infection. This rapidly deployable COVID-19 mouse model is useful for preclinical and pathogenesis studies of COVID-19.


Asunto(s)
Células Epiteliales Alveolares/inmunología , COVID-19/inmunología , Expresión Génica , SARS-CoV-2/inmunología , Transducción de Señal/inmunología , Adenoviridae/genética , Adenoviridae/metabolismo , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Enzima Convertidora de Angiotensina 2/biosíntesis , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Animales , COVID-19/genética , COVID-19/metabolismo , COVID-19/patología , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Transducción de Señal/genética , Transducción Genética
4.
J Cell Physiol ; 235(10): 7383-7391, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32052452

RESUMEN

Models of macrophage subtypes require molecular characterization to reliably facilitate their differentiation. Although CD16+ (Fc-gamma III receptor) monocytes that express CD163 (a hemoglobin/haptoglobin receptor) have been implicated in a variety of disease states, the conditions necessary to establish lineages of these cell subtypes remains unknown. The current investigations utilize a cell line derived from acute myelogenous leukemia lineage, MonoMac-1, to interrogate the factors that promote the development of CD16+ macrophages that express CD163. Results implicate the glucocorticoid pathway as well as c-fms signaling based on the action of dexamethasone and macrophage colony-stimulating factor-1 in promoting CD16+ expression, in addition to phorbol myristate acetate and lipopolysaccharides treatment. The ability of glucocorticoid and c-fms receptor antagonists to inhibit CD16+ cell formation further establishes the role of these pathways in CD16 expression in this cell line. In view of the inherent difficulty in working with primary cells as well as donor variation, cell lines may be preferable to primary cells for their consistency. We envision that the process we use to induce CD16 expression in this cell type will be useful for screening and identification of drug candidates potentially useful for the treatment of diseases where the etiology involves the expansion of the CD16+ monocytes subset or the accumulation of CD163+ tissue macrophages.


Asunto(s)
Diferenciación Celular/fisiología , Glucocorticoides/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal/fisiología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/fisiología , Genes fms/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de IgG/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Ann Neurol ; 83(2): 406-417, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29369399

RESUMEN

OBJECTIVE: CD16+ /CD163+ macrophages (MΦs) and microglia accumulate in the brains of patients with human immunodeficiency virus (HIV) encephalitis (HIVE), a neuropathological correlate of the most severe form of HIV-associated neurocognitive disorders, HIV-associated dementia. Recently, we found that some parenchymal microglia in brain of HIV+ subjects without encephalitis (HIV/noE) but with varying degrees of neurocognitive impairment express CD16 and CD163, even in the absence of detectable virus production. To further our understanding of microglial activation in HIV, we investigated expression of specific genes by profiling parenchymal microglia from archival brain tissue of patients with HIVE and HIV/noE, and HIV- controls. METHODS: Single-population microarray analyses were performed on ∼2,500 laser capture microdissected CD163+ , CD16+ , or CD68+ MΦs/microglia per case, using terminal continuation RNA amplification and a custom-designed array platform. RESULTS: Several classes of microglial transcripts in HIVE and HIV/noE were altered, relative to HIV- subjects, including factors related to cell stress, immune activation, and apoptosis. Additionally, several neurotrophic factors were reduced in HIV infection, suggesting an additional mechanism of neuropathogenesis. The majority of transcripts altered in HIVE displayed intermediate changes in HIV/noE. INTERPRETATION: Our results support the notion that microglia contribute to the maintenance of brain homeostasis and their potential loss of function in the context of chronic inflammation contributes to neuropathogenesis. Furthermore, they indicate the utility of profiling MΦs/microglia to increase our understanding of microglia function, as well as to ascertain alterations in specific pathways, genes, and potentially, encoded proteins that may be amenable to targeted treatment modalities in diseases affecting the brain. Ann Neurol 2018;83:406-417.


Asunto(s)
Complejo SIDA Demencia/inmunología , Disfunción Cognitiva/inmunología , Infecciones por VIH/inmunología , Microglía/inmunología , Complejo SIDA Demencia/complicaciones , Complejo SIDA Demencia/patología , Adulto , Femenino , Perfilación de la Expresión Génica , Infecciones por VIH/patología , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
6.
Respir Res ; 18(1): 8, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28061907

RESUMEN

BACKGROUND: Long-acting muscarinic antagonist/long-acting ß2-agonist combinations are recommended for patients whose chronic obstructive pulmonary disease (COPD) is not managed with monotherapy. We assessed the efficacy and safety of glycopyrrolate (GP)/formoterol fumarate (FF) fixed-dose combination delivered via a Co-Suspension™ Delivery Technology-based metered dose inhaler (MDI) (GFF MDI). METHODS: This was a Phase IIb randomized, multicenter, placebo-controlled, double-blind, chronic-dosing (7 days), crossover study in patients with moderate-to-very severe COPD ( NCT01085045 ). Treatments included GFF MDI twice daily (BID) (GP/FF 72/9.6 µg or 36/9.6 µg), GP MDI 36 µg BID, FF MDI 7.2 and 9.6 µg BID, placebo MDI, and open-label formoterol dry powder inhaler (FF DPI) 12 µg BID or tiotropium DPI 18 µg once daily. The primary endpoint was forced expiratory volume in 1 s area under the curve from 0 to 12 h (FEV1 AUC0-12) on Day 7 relative to baseline FEV1. Secondary endpoints included pharmacokinetics and safety. RESULTS: GFF MDI 72/9.6 µg or 36/9.6 µg led to statistically significant improvements in FEV1 AUC0-12 after 7 days' treatment versus monocomponent MDIs, placebo MDI, tiotropium, or FF DPI (p ≤ 0.0002). GFF MDI 36/9.6 µg was non-inferior to GFF MDI 72/9.6 µg and monocomponent MDIs were non-inferior to open-label comparators. Pharmacokinetic results showed glycopyrrolate and formoterol exposure were decreased following administration via fixed-dose combination versus monocomponent MDIs; however, this was not clinically meaningful. GFF MDI was well tolerated. CONCLUSIONS: GFF MDI 72/9.6 µg and 36/9.6 µg BID improve lung function and are well tolerated in patients with moderate-to-very severe COPD. TRIAL REGISTRATION: ClinicalTrials.gov NCT01085045 . Registered 9 March 2010.


Asunto(s)
Fumarato de Formoterol/administración & dosificación , Glicopirrolato/administración & dosificación , Inhaladores de Dosis Medida , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Administración por Inhalación , Agonistas de Receptores Adrenérgicos beta 2/administración & dosificación , Adulto , Anciano , Australia , Método Doble Ciego , Combinación de Medicamentos , Diseño de Equipo , Análisis de Falla de Equipo , Femenino , Glicopirrolato/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Antagonistas Muscarínicos/administración & dosificación , Nueva Zelanda , Efecto Placebo , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Estados Unidos
7.
Respir Res ; 17(1): 109, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27586537

RESUMEN

BACKGROUND: This study forms part of the first complete characterization of the dose-response curve for glycopyrrolate (GP) delivered using Co-Suspension™ Delivery Technology via a metered dose inhaler (MDI). We examined the lower GP MDI dose range to determine an optimal dose for patients with moderate-to-severe chronic obstructive pulmonary disease (COPD). METHODS: This randomized, double-blind, chronic-dosing, balanced incomplete-block, placebo-controlled, crossover study compared six doses of GP MDI (18, 9, 4.6, 2.4, 1.2, and 0.6 µg, twice daily [BID]) with placebo MDI BID and open-label tiotropium dry powder inhaler (18 µg, once daily [QD]) in patients with moderate-to-severe COPD. Patients were randomized into 1 of 120 treatment sequences. Each sequence included 4 of 8 treatments administered for 14-day periods separated by 7- to 21-day washout periods. The primary efficacy endpoint was change from baseline in forced expiratory volume in 1 s area under the curve from 0 to 12 h (FEV1 AUC0-12) on Day 14. Secondary efficacy endpoints included peak change from baseline (post-dose) in FEV1 and inspiratory capacity (IC) on Days 1, 7, and 14; change from baseline in morning pre-dose trough FEV1 on Days 7 and 14; change from baseline in 12-h post-dose trough FEV1 on Day 14; time to onset of action (≥10 % improvement in mean FEV1) and the proportion of patients achieving ≥12 % improvement in FEV1 on Day 1; and pre-dose trough IC on Days 7 and 14. Safety and tolerability were also assessed. RESULTS: GP MDI 18, 9, 4.6, and 2.4 µg demonstrated statistically significant and clinically relevant increases in FEV1 AUC0-12 compared with placebo MDI following 14 days of treatment (modified intent-to-treat population = 120). GP MDI 18 µg was non-inferior to open-label tiotropium for peak change in FEV1 on Day 1 and morning pre-dose trough FEV1 on Day 14. All doses of GP MDI were well tolerated with no unexpected safety findings. CONCLUSIONS: These efficacy and safety results support GP MDI 18 µg BID as the most appropriate dose for evaluation in Phase III trials in patients with moderate-to-severe COPD. TRIAL REGISTRATION: ClinicalTrials.gov NCT01566773 . Registered 27 March 2012.


Asunto(s)
Broncodilatadores/administración & dosificación , Glicopirrolato/administración & dosificación , Pulmón/efectos de los fármacos , Inhaladores de Dosis Medida , Antagonistas Muscarínicos/administración & dosificación , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Administración por Inhalación , Anciano , Broncodilatadores/efectos adversos , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Diseño de Equipo , Femenino , Volumen Espiratorio Forzado , Glicopirrolato/efectos adversos , Humanos , Pulmón/fisiopatología , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Antagonistas Muscarínicos/efectos adversos , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Índice de Severidad de la Enfermedad , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos , Capacidad Vital
8.
J Neuroinflammation ; 12: 58, 2015 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-25886134

RESUMEN

BACKGROUND: Macrophage-colony stimulating factor (M-CSF) has been implicated in HIV neuropathogenesis through its ability to modulate activation of macrophages (MΦs) and microglia, as well as enhance the susceptibility of these cells to infection and promote virus production. We have recently reported that MΦs accumulating perivascularly and within nodular lesions in archival brain tissue of simian immunodeficiency virus (SIV)-infected rhesus macaques with encephalitis (SIVE) express M-CSF. In contrast, IL-34, which shares the same receptor, cFMS, was observed more often in parenchymal cells. METHODS: Frontal white and grey matter from non-infected and SIV-infected rhesus macaques with and without SIVE were examined by single- and double-label immunohistochemistry for M-CSF, IL-34, and CD163 expression. Primary rhesus macaque and human peripheral blood mononuclear cells were cultured with and without 2.5 ng/ml M-CSF or IL-34 alone and with 470 nM or 4.7 µM of GW2580, a receptor tyrosine kinase inhibitor with high specificity for cFMS. After 24 h, cells were analyzed by flow cytometry to examine the effect of these cytokines on promoting an M2 monocyte/MΦ phenotype. RESULTS: Here, we demonstrate that in SIVE brain, accumulating M-CSF(+) MΦs are also CD163(+), while IL-34 does not appear to co-localize significantly with CD163 in the parenchyma. We further demonstrate that M-CSF and IL-34 are expressed by neurons in normal brain but are altered in SIV and SIVE. Through in vitro studies, we show that M-CSF and IL-34 upregulate CD163, a marker for type 2 activation of MΦs (M2), by primary monocytes, which is attenuated by the addition of GW2580. CONCLUSIONS: Together, these data suggest that both cFMS ligands may promote and/or prolong M2 activation of MΦs and microglia in brains of SIV-infected animals with encephalitis. As such, cFMS signaling may be an attractive target for eliminating long-lived MΦ reservoirs of HIV infection in brain, as well as other tissues.


Asunto(s)
Lóbulo Frontal/patología , Regulación de la Expresión Génica/fisiología , Macrófagos/patología , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Análisis de Varianza , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Polaridad Celular/fisiología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Macaca mulatta , Factor Estimulante de Colonias de Macrófagos/metabolismo , Transducción de Señal/fisiología , Sustancia Blanca/metabolismo
9.
J Neurovirol ; 21(3): 249-56, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25060299

RESUMEN

Combination antiretroviral therapy (cART) has improved the longevity and quality of life for people living with HIV; however, it does not target virus that persists in long-lived cells, such as macrophages (MΦs). This allows for the development of viral reservoirs in various anatomical compartments where these cells reside, including the central nervous system (CNS), where perivascular MΦs and resident microglia constitute the principle cellular reservoir of HIV. How HIV persists in MΦs/microglia is not completely understood; however, prosurvival signaling that protects infected MΦs/microglia from apoptosis is likely important to viral persistence. Macrophage colony-stimulating factor (M-CSF) is an important factor in MΦ survival and has been implicated in HIV neuropathogenesis through its ability to enhance the susceptibility of MΦs to infection and promote virus production. While M-CSF has been detected in cerebrospinal fluid of HIV-infected patients, the cellular source of M-CSF in the CNS is unknown. Here, we demonstrate, for the first time, that MΦs comprising perivascular cuffs and nodular lesions in SIV encephalitis (SIVE) brain are the principle source of M-CSF. These cells also serve as the primary reservoir of productive SIV infection in the brain. We further demonstrate that M-CSF and IL-34, which signal through the same receptor, cFMS, enhance HIV-1 production by microglia in vitro. This is attenuated by the addition of a receptor tyrosine kinase inhibitor with high specificity for cFMS, GW2580. Together, these data suggest that cFMS signaling may be an attractive target for eliminating long-lived MΦ reservoirs of HIV in the brain and other tissues.


Asunto(s)
Complejo SIDA Demencia/virología , Encéfalo/virología , Reservorios de Enfermedades/virología , Microglía/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Complejo SIDA Demencia/inmunología , Animales , Encéfalo/inmunología , VIH-1/inmunología , Humanos , Inmunohistoquímica , Hibridación in Situ , Interleucinas/inmunología , Macaca mulatta , Factor Estimulante de Colonias de Macrófagos/biosíntesis , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/virología , Microglía/inmunología , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Transducción de Señal/fisiología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología
10.
BMC Pulm Med ; 14: 118, 2014 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-25027304

RESUMEN

BACKGROUND: Bronchodilator medications are central to the symptomatic management of chronic obstructive pulmonary disease (COPD). Metered-dose inhalers (MDIs) are the most commonly used devices to deliver treatment to patients with COPD and asthma, comprising approximately 70% of bronchodilator prescriptions. Proprietary porous-particle technology permits the formulation of long-acting muscarinic antagonists, long-acting ß2-agonists, and a combination of both in hydrofluoroalkane (HFA) MDIs, providing a solution to formulation challenges inherent to the development of HFA MDIs, which have contributed to the development of dry-powder inhalers. METHODS: In this randomized, double-blind, 4-period, 6-treatment, placebo- and active-controlled, multicenter, crossover study, 4 ascending single doses of a proprietary glycopyrronium (GP) MDI were evaluated compared with Placebo MDI and open-label tiotropium (TIO) in study patients with COPD. Thirty-three study patients were enrolled and received single-dose administration of 4 of the 6 treatments (Placebo MDI, TIO 18 µg, or GP MDI at 14.4, 28.8, 57.6, and 115.2 µg ex-actuator) with an interval of 1 to 3 weeks between doses. The primary efficacy endpoint was peak change in forced expiratory volume in 1 second (FEV1). RESULTS: All 4 doses of GP MDI showed statistically superior efficacy compared with Placebo MDI for peak FEV1 (differences of 146 to 248 mL; P<.001), with a clear dose ordering of the response. Statistically significant differences compared with Placebo MDI were noted at almost all doses for the secondary FEV1 parameters (P ≤ .049) except 24-hour trough FEV1 at 28.8 µg. All doses were safe and well tolerated in this study; the most frequently reported adverse event was dry mouth (0-14.3% across doses; 9.5% for Placebo MDI, and 9.1% for TIO). CONCLUSIONS: This study demonstrated superior bronchodilatory efficacy of GP MDI compared with Placebo MDI at all doses tested, and no serious adverse events were reported. This study supports the further evaluation of GP MDI in study patients with COPD. In addition, these findings indicate that the correct dosage of glycopyrronium is no more than 115.2 µg total daily dose, or 57.6 µg twice daily based on comparisons with the active comparator. TRIAL REGISTRATION: This clinical trial was registered on ClinicalTrials.gov, Identifier: NCT00871182.


Asunto(s)
Broncodilatadores/uso terapéutico , Glicopirrolato/uso terapéutico , Antagonistas Muscarínicos/uso terapéutico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Adulto , Anciano , Área Bajo la Curva , Broncodilatadores/efectos adversos , Broncodilatadores/farmacocinética , Estudios Cruzados , Método Doble Ciego , Femenino , Volumen Espiratorio Forzado , Glicopirrolato/efectos adversos , Glicopirrolato/farmacocinética , Humanos , Masculino , Inhaladores de Dosis Medida , Persona de Mediana Edad , Antagonistas Muscarínicos/efectos adversos , Antagonistas Muscarínicos/farmacocinética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Derivados de Escopolamina/uso terapéutico , Bromuro de Tiotropio , Xerostomía/inducido químicamente
11.
Front Cell Neurosci ; 18: 1360195, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38550920

RESUMEN

The blood brain barrier (BBB) plays a crucial role in maintaining brain homeostasis by selectively preventing the entry of substances from the peripheral blood into the central nervous system (CNS). Comprised of endothelial cells, pericytes, and astrocytes, this highly regulated barrier encompasses the majority of the brain's vasculature. In addition to its protective function, the BBB also engages in significant crosstalk with perivascular macrophages (MΦ) and microglia, the resident MΦ of the brain. These interactions play a pivotal role in modulating the activation state of cells comprising the BBB, as well as MΦs and microglia, themselves. Alterations in systemic metabolic and inflammatory states can promote endothelial cell dysfunction, reducing the integrity of the BBB and potentially allowing peripheral blood factors to leak into the CNS compartment. This may mediate activation of perivascular MΦs, microglia, and astrocytes, and initiate further immune responses within the brain parenchyma, suggesting neuroinflammation can be triggered by signaling from the periphery, without primary injury or disease originating within the CNS. The intricate interplay between the periphery and the CNS through the BBB highlights the importance of understanding the role of microglia in mediating responses to systemic challenges. Despite recent advancements, our understanding of the interactions between microglia and the BBB is still in its early stages, leaving a significant gap in knowledge. However, emerging research is shedding light on the involvement of microglia at the BBB in various conditions, including systemic infections, diabetes, and ischemic stroke. This review aims to provide a comprehensive overview of the current research investigating the intricate relationship between microglia and the BBB in health and disease. By exploring these connections, we hope to advance our understanding of the role of brain immune responses to systemic challenges and their impact on CNS health and pathology. Uncovering these interactions may hold promise for the development of novel therapeutic strategies for neurological conditions that involve immune and vascular mechanisms.

12.
Elife ; 122023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37417740

RESUMEN

Infection with the etiological agent of COVID-19, SARS-CoV-2, appears capable of impacting cognition in some patients with post-acute sequelae of SARS-CoV-2 (PASC). To evaluate neuropathophysiological consequences of SARS-CoV-2 infection, we examine transcriptional and cellular signatures in the Brodmann area 9 (BA9) of the frontal cortex and the hippocampal formation (HF) in SARS-CoV-2, Alzheimer's disease (AD), and SARS-CoV-2-infected AD individuals compared to age- and gender-matched neurological cases. Here, we show similar alterations of neuroinflammation and blood-brain barrier integrity in SARS-CoV-2, AD, and SARS-CoV-2-infected AD individuals. Distribution of microglial changes reflected by the increase in Iba-1 reveals nodular morphological alterations in SARS-CoV-2-infected AD individuals. Similarly, HIF-1α is significantly upregulated in the context of SARS-CoV-2 infection in the same brain regions regardless of AD status. The finding may help in informing decision-making regarding therapeutic treatments in patients with neuro-PASC, especially those at increased risk of developing AD.


Asunto(s)
Enfermedad de Alzheimer , COVID-19 , Humanos , SARS-CoV-2 , Barrera Hematoencefálica , Cognición , Progresión de la Enfermedad
13.
Nat Commun ; 14(1): 4414, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37479685

RESUMEN

Elevation in soluble urokinase receptor (suPAR) and proteinuria are common signs in patients with moderate to severe coronavirus disease 2019 (COVID-19). Here we characterize a new type of proteinuria originating as part of a viral response. Inoculation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes increased suPAR levels and glomerulopathy in African green monkeys. Using an engineered mouse model with high suPAR expression, inhaled variants of SARS-CoV-2 spike S1 protein elicite proteinuria that could be blocked by either suPAR antibody or SARS-CoV-2 vaccination. In a cohort of 1991 COVID-19 patients, suPAR levels exhibit a stepwise association with proteinuria in non-Omicron, but not in Omicron infections, supporting our findings of biophysical and functional differences between variants of SARS-CoV-2 spike S1 protein and their binding to podocyte integrins. These insights are not limited to SARS-CoV-2 and define viral response proteinuria (VRP) as an innate immune mechanism and co-activation of podocyte integrins.


Asunto(s)
COVID-19 , Podocitos , Animales , Ratones , Chlorocebus aethiops , Humanos , Vacunas contra la COVID-19 , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , SARS-CoV-2 , Integrinas , Proteinuria
15.
bioRxiv ; 2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36451886

RESUMEN

Infection with the etiological agent of COVID-19, SARS-CoV-2, appears capable of impacting cognition, which some patients with Post-acute Sequelae of SARS-CoV-2 (PASC). To evaluate neuro-pathophysiological consequences of SARS-CoV-2 infection, we examine transcriptional and cellular signatures in the Broadman area 9 (BA9) of the frontal cortex and the hippocampal formation (HF) in SARS-CoV-2, Alzheimer's disease (AD) and SARS-CoV-2 infected AD individuals, compared to age- and gender-matched neurological cases. Here we show similar alterations of neuroinflammation and blood-brain barrier integrity in SARS-CoV-2, AD, and SARS-CoV-2 infected AD individuals. Distribution of microglial changes reflected by the increase of Iba-1 reveal nodular morphological alterations in SARS-CoV-2 infected AD individuals. Similarly, HIF-1α is significantly upregulated in the context of SARS-CoV-2 infection in the same brain regions regardless of AD status. The finding may help to inform decision-making regarding therapeutic treatments in patients with neuro-PASC, especially those at increased risk of developing AD. Teaser: SARS-CoV-2 and Alzheimer's disease share similar neuroinflammatory processes, which may help explain neuro-PASC.

16.
Nat Commun ; 13(1): 1745, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35365631

RESUMEN

Neurological manifestations are a significant complication of coronavirus disease (COVID-19), but underlying mechanisms aren't well understood. The development of animal models that recapitulate the neuropathological findings of autopsied brain tissue from patients who died from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are critical for elucidating the neuropathogenesis of infection and disease. Here, we show neuroinflammation, microhemorrhages, brain hypoxia, and neuropathology that is consistent with hypoxic-ischemic injury in SARS-CoV-2 infected non-human primates (NHPs), including evidence of neuron degeneration and apoptosis. Importantly, this is seen among infected animals that do not develop severe respiratory disease, which may provide insight into neurological symptoms associated with "long COVID". Sparse virus is detected in brain endothelial cells but does not associate with the severity of central nervous system (CNS) injury. We anticipate our findings will advance our current understanding of the neuropathogenesis of SARS-CoV-2 infection and demonstrate SARS-CoV-2 infected NHPs are a highly relevant animal model for investigating COVID-19 neuropathogenesis among human subjects.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Encéfalo , Células Endoteliales , Humanos , Primates
17.
Front Aging Neurosci ; 13: 653694, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34408638

RESUMEN

The outbreak of the novel and highly infectious severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has resulted in hundreds of millions of infections and millions of deaths globally. Infected individuals that progress to coronavirus disease-19 (COVID-19) experience upper and lower respiratory complications that range in severity and may lead to wide-spread inflammation and generalized hypoxia or hypoxemia that impacts multiple organ systems, including the central and peripheral nervous systems. Since the SARS-CoV-2 outbreak, multiple reports continue to emerge that detail neurological symptoms, ranging from relatively mild (e.g., impaired taste and/or smell) to severe (e.g., stroke), suggesting SARS-CoV-2 may be neurotropic and/or contribute to nervous system injury through direct and/or indirect mechanisms. To gain insight into the types of neurological complications associated with SARS-CoV-2 infection and their possible relationship with age, sex, COVID-19 severity, and comorbidities, we performed a systematic review of case reports and series published in 2020 - April 4, 2021 of infected patients with neurological manifestations. Meta-analyses were conducted using individual patient data from reports where these data could be extracted. Here, we report neurological injury occurs across the lifespan in the context of infection, with and without known comorbidities, and with all disease severities, including asymptomatic patients. Older individuals, however, are more susceptible to developing life-threatening COVID-19 and cerebrovascular disease (CVD), such as stroke. A mild but inverse correlation with age was seen with CNS inflammatory diseases, such as encephalitis, as well as taste and/or smell disorders. When reported, increased age was also associated with comorbid cardiovascular risk factors, including hypertension, diabetes mellitus, and lipid disorders, but not with obesity. Obesity did correlate with development of critical COVID-19. Discussion into potential pathophysiological mechanisms by which neurological symptoms arise and long-term consequences of infection to the nervous system is also provided.

18.
JCI Insight ; 6(16)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34241597

RESUMEN

Evidence suggests an association between severe acute respiratory syndrome-cornavirus-2 (SARS-CoV-2) infection and the occurrence of new-onset diabetes. We examined pancreatic expression of angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2), the cell entry factors for SARS-CoV-2, using publicly available single-cell RNA sequencing data sets, and pancreatic tissue from control male and female nonhuman primates (NHPs) and humans. We also examined SARS-CoV-2 immunolocalization in pancreatic cells of SARS-CoV-2-infected NHPs and patients who had died from coronavirus disease 2019 (COVID-19). We report expression of ACE2 in pancreatic islet, ductal, and endothelial cells in NHPs and humans. In pancreata from SARS-CoV-2-infected NHPs and COVID-19 patients, SARS-CoV-2 infected ductal, endothelial, and islet cells. These pancreata also exhibited generalized fibrosis associated with multiple vascular thrombi. Two out of 8 NHPs developed new-onset diabetes following SARS-CoV-2 infection. Two out of 5 COVID-19 patients exhibited new-onset diabetes at admission. These results suggest that SARS-CoV-2 infection of the pancreas may promote acute and especially chronic pancreatic dysfunction that could potentially lead to new-onset diabetes.


Asunto(s)
COVID-19/complicaciones , Diabetes Mellitus/etiología , Páncreas/virología , SARS-CoV-2/aislamiento & purificación , Trombosis/etiología , Enzima Convertidora de Angiotensina 2/análisis , Animales , Chlorocebus aethiops , Femenino , Fibrosis , Humanos , Macaca mulatta , Masculino , Serina Endopeptidasas/análisis
19.
Theranostics ; 10(16): 7448-7464, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32642005

RESUMEN

The COVID-19 pandemic is an emerging threat to global public health. While our current understanding of COVID-19 pathogenesis is limited, a better understanding will help us develop efficacious treatment and prevention strategies for COVID-19. One potential therapeutic target is angiotensin converting enzyme 2 (ACE2). ACE2 primarily catalyzes the conversion of angiotensin I (Ang I) to a nonapeptide angiotensin or the conversion of angiotensin II (Ang II) to angiotensin 1-7 (Ang 1-7) and has direct effects on cardiac function and multiple organs via counter-regulation of the renin-angiotensin system (RAS). Significant to COVID-19, ACE2 is postulated to serve as a major entry receptor for SARS-CoV-2 in human cells, as it does for SARS-CoV. Many infected individuals develop COVID-19 with fever, cough, and shortness of breath that can progress to pneumonia. Disease progression promotes the activation of immune cells, platelets, and coagulation pathways that can lead to multiple organ failure and death. ACE2 is expressed by epithelial cells of the lungs at high level, a major target of the disease, as seen in post-mortem lung tissue of patients who died with COVID-19, which reveals diffuse alveolar damage with cellular fibromyxoid exudates bilaterally. Comparatively, ACE2 is expressed at low level by vascular endothelial cells of the heart and kidney but may also be targeted by the virus in severe COVID-19 cases. Interestingly, SARS-CoV-2 infection downregulates ACE2 expression, which may also play a critical pathogenic role in COVID-19. Importantly, targeting ACE2/Ang 1-7 axis and blocking ACE2 interaction with the S protein of SARS-CoV-2 to curtail SARS-CoV-2 infection are becoming very attractive therapeutics potential for treatment and prevention of COVID-19. Here, we will discuss the following subtopics: 1) ACE2 as a receptor of SARS-CoV-2; 2) clinical and pathological features of COVID-19; 3) role of ACE2 in the infection and pathogenesis of SARS; 4) potential pathogenic role of ACE2 in COVID-19; 5) animal models for pathological studies and therapeutics; and 6) therapeutics development for COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/metabolismo , Neumonía Viral/virología , Receptores Virales/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Antivirales/uso terapéutico , Betacoronavirus/química , Betacoronavirus/patogenicidad , Betacoronavirus/fisiología , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/terapia , Modelos Animales de Enfermedad , Interacciones Microbiota-Huesped/fisiología , Humanos , Ratones , Modelos Biológicos , Pandemias , Neumonía Viral/terapia , Sistema Renina-Angiotensina/fisiología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Nanomedicina Teranóstica , Vacunas Virales/aislamiento & purificación , Internalización del Virus
20.
Nat Commun ; 11(1): 2280, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32385245

RESUMEN

Renal macrophages (RMs) participate in tissue homeostasis, inflammation and repair. RMs consist of embryo-derived (EMRMs) and bone marrow-derived RMs (BMRMs), but the fate, dynamics, replenishment, functions and metabolic states of these two RM populations remain unclear. Here we investigate and characterize RMs at different ages by conditionally labeling and ablating RMs populations in several transgenic lines. We find that RMs expand and mature in parallel with renal growth after birth, and are mainly derived from fetal liver monocytes before birth, but self-maintain through adulthood with contribution from peripheral monocytes. Moreover, after the RMs niche is emptied, peripheral monocytes rapidly differentiate into BMRMs, with the CX3CR1/CX3CL1 signaling axis being essential for the maintenance and regeneration of both EMRMs and BMRMs. Lastly, we show that EMRMs have a higher capacity for scavenging immune complex, and are more sensitive to immune challenge than BMRMs, with this difference associated with their distinct glycolytic capacities.


Asunto(s)
Células de la Médula Ósea/citología , Linaje de la Célula , Riñón/embriología , Macrófagos/citología , Animales , Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/sangre , Quimiocina CX3CL1/metabolismo , Femenino , Feto/citología , Hígado/embriología , Masculino , Ratones , Monocitos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA