Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.466
Filtrar
1.
Nature ; 628(8009): 835-843, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38600381

RESUMEN

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Asunto(s)
Necroptosis , Infecciones por Orthomyxoviridae , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Animales , Necroptosis/efectos de los fármacos , Ratones , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/complicaciones , Femenino , Masculino , Humanos , Lesión Pulmonar/prevención & control , Lesión Pulmonar/virología , Lesión Pulmonar/patología , Lesión Pulmonar/tratamiento farmacológico , Virus de la Influenza A/fisiología , Virus de la Influenza A/efectos de los fármacos , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/virología , Células Epiteliales Alveolares/metabolismo , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Gripe Humana/virología , Gripe Humana/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/virología , Síndrome de Dificultad Respiratoria/prevención & control , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico
2.
Stem Cell Res Ther ; 15(1): 109, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38637891

RESUMEN

BACKGROUND: The STROMA-CoV-2 study was a French phase 2b, multicenter, double-blind, randomized, placebo-controlled clinical trial that did not identify a significant efficacy of umbilical cord-derived mesenchymal stromal cells in patients with SARS-CoV-2-induced acute respiratory distress syndrome. Safety on day 28 was found to be good. The aim of our extended study was to assess the 6- and 12-month safety of UC-MSCs administration in the STROMA-CoV-2 cohort. METHODS: A detailed multi-domain assessment was conducted at 6 and 12 months following hospital discharge focusing on adverse events, lung computed tomography-scan, pulmonary and muscular functional status, and quality of life in the STROMA-CoV-2 cohort including SARS-CoV-2-related early (< 96 h) mild-to-severe acute respiratory distress syndrome. RESULTS: Between April 2020 and October 2020, 47 patients were enrolled, of whom 19 completed a 1-year follow-up. There were no significant differences in any endpoints or adverse effects between the UC-MSCs and placebo groups at the 6- and 12-month assessments. Ground-glass opacities persisted at 1 year in 5 patients (26.3%). Furthermore, diffusing capacity for carbon monoxide remained altered over 1 year, although no patient required oxygen or non-invasive ventilatory support. Quality of life revealed declines in mental, emotional and physical health throughout the follow-up period, and the six-minute walking distance remained slightly impaired at the 1-year patient assessment. CONCLUSIONS: This study suggests a favorable safety profile for the use of intravenous UC-MSCs in the context of the first French wave of SARS-CoV-2-related moderate-to-severe acute respiratory distress syndrome, with no adverse effects observed at 1 year.


Asunto(s)
COVID-19 , Células Madre Mesenquimatosas , Síndrome de Dificultad Respiratoria , Humanos , COVID-19/terapia , Método Doble Ciego , Calidad de Vida , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , SARS-CoV-2 , Resultado del Tratamiento , Cordón Umbilical
3.
Respir Res ; 25(1): 151, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38561798

RESUMEN

INTRODUCTION: EXO-CD24 are exosomes genetically manipulated to over-express Cluster of Differentiation (CD) 24. It consists of two breakthrough technologies: CD24, the drug, as a novel immunomodulator that is smarter than steroids without any side effects, and exosomes as the ideal natural drug carrier. METHODS: A randomized, single blind, dose-finding phase IIb trial in hospitalized patients with mild to moderate Coronavirus disease 2019 (COVID-19) related Acute Respiratory Distress Syndrome (ARDS) was carried out in two medical centers in Athens. Patients received either 109 or 1010 exosome particles of EXO-CD24, daily, for five consecutive days and monitored for 28 days. Efficacy was assessed at day 7 among 91 patients who underwent randomization. The outcome was also compared in a post-hoc analysis with an income control group (n = 202) that fit the inclusion and exclusion criteria. RESULTS: The mean age was 49.4 (± 13.2) years and 74.4% were male. By day 7, 83.7% showed improved respiratory signs and 64% had better oxygen saturation (SpO2) (p < 0.05). There were significant reductions in all inflammatory markers, most notably in C-reactive protein (CRP), lactate dehydrogenase (LDH), ferritin, fibrinogen and an array of cytokines. Conversely, levels of the anti-inflammatory cytokine Interleukin-10 (IL-10) were increased (p < 0.05). Of all the documented adverse events, none were considered treatment related. No drug-drug interactions were noted. Two patients succumbed to COVID-19. Post-hoc analysis revealed that EXO-CD24 patients exhibited greater improvements in clinical and laboratory outcomes compared to an observational income control group. CONCLUSIONS: EXO-CD24 presents a promising therapeutic approach for hyper-inflammatory state and in particular ARDS. Its unique combination of exosomes, as a drug carrier, and CD24, as an immunomodulator, coupled with inhalation administration, warrants further investigation in a larger, international, randomized, quadri-blind trial against a placebo.


Asunto(s)
COVID-19 , Exosomas , Síndrome de Dificultad Respiratoria , Humanos , Masculino , Persona de Mediana Edad , Femenino , SARS-CoV-2 , Método Simple Ciego , Factores Inmunológicos , Síndrome de Dificultad Respiratoria/diagnóstico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/genética , Portadores de Fármacos , Resultado del Tratamiento , Antígeno CD24
4.
Ther Adv Respir Dis ; 18: 17534666241244974, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38616385

RESUMEN

Nanoparticles have attracted extensive attention due to their high degree of cell targeting, biocompatibility, controllable biological activity, and outstanding pharmacokinetics. Changing the size, morphology, and surface chemical groups of nanoparticles can increase the biological distribution of agents to achieve precise tissue targeting and optimize therapeutic effects. Examples of their use include nanoparticles designed for increasing antigen-specific immune responses, developing vaccines, and treating inflammatory diseases. Nanoparticles show the potential to become a new generation of therapeutic agents for regulating inflammation. Recently, many nanomaterials with targeted properties have been developed to treat acute lung injury/acute respiratory distress syndrome (ALI/ARDS). In this review, we provide a brief explanation of the pathological mechanism underlying ALI/ARDS and a systematic overview of the latest technology and research progress in nanomedicine treatments of ALI, including improved nanocarriers, nanozymes, and nanovaccines for the targeted treatment of lung injury. Ultimately, these nanomedicines will be used for the clinical treatment of ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Síndrome de Dificultad Respiratoria , Humanos , Nanomedicina , Lesión Pulmonar Aguda/tratamiento farmacológico , Movimiento Celular , Inflamación , Síndrome de Dificultad Respiratoria/tratamiento farmacológico
5.
Front Immunol ; 15: 1330373, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38596679

RESUMEN

Introduction: Indole-3-carbinol (I3C) is found in cruciferous vegetables and used as a dietary supplement. It is known to act as a ligand for aryl hydrocarbon receptor (AhR). In the current study, we investigated the role of AhR and the ability of I3C to attenuate LPS-induced Acute Respiratory Distress Syndrome (ARDS). Methods: To that end, we induced ARDS in wild-type C57BL/6 mice, Ccr2gfp/gfp KI/KO mice (mice deficient in the CCR2 receptor), and LyZcreAhRfl/fl mice (mice deficient in the AhR on myeloid linage cells). Additionally, mice were treated with I3C (65 mg/kg) or vehicle to investigate its efficacy to treat ARDS. Results: I3C decreased the neutrophils expressing CXCR2, a receptor associated with neutrophil recruitment in the lungs. In addition, LPS-exposed mice treated with I3C revealed downregulation of CCR2+ monocytes in the lungs and lowered CCL2 (MCP-1) protein levels in serum and bronchoalveolar lavage fluid. Loss of CCR2 on monocytes blocked the recruitment of CXCR2+ neutrophils and decreased the total number of immune cells in the lungs during ARDS. In addition, loss of the AhR on myeloid linage cells ablated I3C-mediated attenuation of CXCR2+ neutrophils and CCR2+ monocytes in the lungs from ARDS animals. Interestingly, scRNASeq showed that in macrophage/monocyte cell clusters of LPS-exposed mice, I3C reduced the expression of CXCL2 and CXCL3, which bind to CXCR2 and are involved in neutrophil recruitment to the disease site. Discussion: These findings suggest that CCR2+ monocytes are involved in the migration and recruitment of CXCR2+ neutrophils during ARDS, and the AhR ligand, I3C, can suppress ARDS through the regulation of immune cell trafficking.


Asunto(s)
Indoles , Monocitos , Síndrome de Dificultad Respiratoria , Ratones , Animales , Monocitos/metabolismo , Lipopolisacáridos/farmacología , Neutrófilos/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Ligandos , Ratones Endogámicos C57BL , Pulmón/metabolismo , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo
6.
Crit Care Clin ; 40(2): 275-289, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432696

RESUMEN

Acute respiratory failure relies on supportive care using non-invasive and invasive oxygen and ventilatory support. Pharmacologic therapies for the most severe form of respiratory failure, acute respiratory distress syndrome (ARDS), are limited. This review focuses on the most promising therapies for ARDS, targeting different mechanisms that contribute to dysregulated inflammation and resultant hypoxemia. Significant heterogeneity exists within the ARDS population. Treatment requires prompt recognition of ARDS and an understanding of which patients may benefit most from specific pharmacologic interventions. The key to finding effective pharmacotherapies for ARDS may rely on deeper understanding of pathophysiology and bedside identification of ARDS subphenotypes.


Asunto(s)
Síndrome de Dificultad Respiratoria , Humanos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Inflamación , Oxígeno
8.
Colloids Surf B Biointerfaces ; 237: 113869, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38522285

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are serious lung injuries caused by various factors, leading to increased permeability of the alveolar-capillary barrier, reduced stability of the alveoli, inflammatory response, and hypoxemia. Despite several decades of research since ARDS was first formally described in 1967, reliable clinical treatment options are still lacking. Currently, supportive therapy and mechanical ventilation are prioritized, and there is no medication that can be completely effective in clinical treatment. In recent years, nanomedicine has developed rapidly and has exciting preclinical treatment capabilities. Using a drug delivery system based on nanobiotechnology, local drugs can be continuously released in lung tissue at therapeutic levels, reducing the frequency of administration and improving patient compliance. Furthermore, this novel drug delivery system can target specific sites and reduce systemic side effects. Currently, many nanomedicine treatment options for ARDS have demonstrated efficacy. This review briefly introduces the pathophysiology of ARDS, discusses various research progress on using nanomedicine to treat ARDS, and anticipates future developments in related fields.


Asunto(s)
Nanomedicina , Síndrome de Dificultad Respiratoria , Humanos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Pulmón , Sistemas de Liberación de Medicamentos
9.
Thromb Res ; 236: 161-166, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38452448

RESUMEN

Direct thrombin inhibitors, including argatroban, are increasingly used for anticoagulation during venovenous extracorporeal membrane oxygenation (VV ECMO). In many centers activated partial thromboplastin time (aPTT) is used for monitoring, but it can be affected by several confounders. The aim of this study was to evaluate the safety and efficacy of anticoagulation with argatroban titrated according to diluted thrombin time targets (hemoclot™ assay) compared to anti-Xa guided anticoagulation with unfractionated heparin (UFH). METHODS: This cohort study included adults at two tertiary care centers who required VV ECMO for severe COVID-19-related acute respiratory distress syndrome (CARDS). Patients received center-dependent argatroban or UFH for anticoagulation during ECMO. Argatroban was guided following a hemoclot™ target range of 0.4-0.6 µg/ml. UFH was guided by anti-factor Xa (antiXa) levels (0.2-0.3 IU/ml). The primary outcome was safety of argatroban compared to UFH, assessed by time to first clinically relevant bleeding event or death during ECMO. Secondary outcomes included efficacy (time to thromboembolism) and feasibility (proportion of anticoagulation targets within range). RESULTS: From 2019 to 2021 57 patients were included in the study with 27 patients (47 %) receiving argatroban and 30 patients (53 %) receiving UFH. The time to the first clinically relevant bleeding or death during ECMO was similar between groups (HR (argatroban vs. UFH): 1.012, 95 % CI 0.44-2.35, p = 0.978). Argatroban was associated with a decreased risk for thromboembolism compared to UFH (HR 0.494 (95 % CI 0.26-0.95; p = 0.034)). The overall proportion of anticoagulation within target ranges was not different between groups (46 % (23-54 %) vs. 46 % (37 %-57 %), p = 0.45). CONCLUSION: Anticoagulation with argatroban according to hemoclot™ targets (0.4-0.6 µg/ml) compared to antiXa guided UFH (0.2-0.3 IU/ml) is safe and may prolong thromboembolism-free time in patients with severe ARDS requiring VV ECMO.


Asunto(s)
Arginina/análogos & derivados , Oxigenación por Membrana Extracorpórea , Ácidos Pipecólicos , Síndrome de Dificultad Respiratoria , Sulfonamidas , Tromboembolia , Adulto , Humanos , Heparina/uso terapéutico , Heparina/farmacología , Anticoagulantes/uso terapéutico , Estudios de Cohortes , Heparina de Bajo-Peso-Molecular , Hemorragia , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Estudios Retrospectivos
10.
J Tradit Chin Med ; 44(2): 381-387, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38504544

RESUMEN

OBJECTIVE: To evaluate the efficacy of Qidong Huoxue decoction (,QDHX) in treating acute lung injury and acute respiratory distress syndrome (ALI/ARDS) when used as an adjunctive treatment. METHODS: ALI/ARDS patients admitted to our medical intensive care unit were randomly allocated to the control group or the QDHX group and received standard therapy. The QDHX group received QDHX (50 mL per day for 14 d) orally or via a gastric tube. The primary outcome was measured according to Traditional Chinese Medicine (TCM) syndrome scores, with partial pressure of oxygen/fraction of inspired oxygen (PaO2/FiO2) levels as the secondary outcome. RESULTS: A total of 73 patients completed the study (36 in the TCM and 37 in the conventional group), and their records were analyzed. After 14-d treatment, the TCM group showed a significant decrease in TCM syndrome scores (P < 0.05) and increased PaO2/FiO2 levels (P < 0.05). The therapeutic effect of integrated Chinese and western medicine was more significant than that of Western Medicine alone. No serious side effects were observed. CONCLUSIONS: Our study results show that QDHX in combination with conventional drug therapy can significantly reduce some clinical symptoms in patients with ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Síndrome de Dificultad Respiratoria , Humanos , Lesión Pulmonar Aguda/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Unidades de Cuidados Intensivos , Oxígeno
11.
Exp Gerontol ; 188: 112389, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432575

RESUMEN

Aging-related diseases (ARDs) are a major global health concern, and the development of effective therapies is urgently needed. Kaempferol, a flavonoid found in several plants, has emerged as a promising candidate for ameliorating ARDs. This comprehensive review examines Kaempferol's chemical properties, safety profile, and pharmacokinetics, and highlights its potential therapeutic utility against ARDs. Kaempferol's therapeutic potential is underpinned by its distinctive chemical structure, which confers antioxidative and anti-inflammatory properties. Kaempferol counteracts reactive oxygen species (ROS) and modulates crucial cellular pathways, thereby combating oxidative stress and inflammation, hallmarks of ARDs. Kaempferol's low toxicity and wide safety margins, as demonstrated by preclinical and clinical studies, further substantiate its therapeutic potential. Compelling evidence supports Kaempferol's substantial potential in addressing ARDs through several mechanisms, notably anti-inflammatory, antioxidant, and anti-apoptotic actions. Kaempferol exhibits a versatile neuroprotective effect by modulating various proinflammatory signaling pathways, including NF-kB, p38MAPK, AKT, and the ß-catenin cascade. Additionally, it hinders the formation and aggregation of beta-amyloid protein and regulates brain-derived neurotrophic factors. In terms of its anticancer potential, kaempferol acts through diverse pathways, inducing apoptosis, arresting the cell cycle at the G2/M phase, suppressing epithelial-mesenchymal transition (EMT)-related markers, and affecting the phosphoinositide 3-kinase/protein kinase B signaling pathways. Subsequent studies should focus on refining dosage regimens, exploring innovative delivery systems, and conducting comprehensive clinical trials to translate these findings into effective therapeutic applications.


Asunto(s)
Quempferoles , Síndrome de Dificultad Respiratoria , Humanos , Quempferoles/farmacología , Quempferoles/uso terapéutico , Quempferoles/química , Fosfatidilinositol 3-Quinasas , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Envejecimiento , Síndrome de Dificultad Respiratoria/tratamiento farmacológico
12.
Sci Rep ; 14(1): 5632, 2024 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-38453960

RESUMEN

This study aimed to investigate differences in clinical characteristics and laboratory findings between children infected with Macrolide-Sensitive Mycoplasma pneumoniae (MSMP) and Macrolide-Resistant Mycoplasma pneumoniae (MRMP). Additionally, the research sought to identify laboratory markers for rapidly distinguishing refractory Mycoplasma pneumoniae pneumonia (RMPP) from ordinary Mycoplasma pneumoniae pneumonia (OMPP). In total, 265 Mycoplasma pneumoniae (MP) patients were included, with MRMP identified by specific point mutations in domain V of the 23S rRNA gene. A retrospective analysis compared the clinical courses and laboratory data, revealing that MRMP patients experienced prolonged febrile days (P = 0.004), elevated CRP levels (P < 0.001), and higher MP DNA loads than MSMP patients (P = 0.037). Based on clinical symptoms, MRMP was divided into RMPP (n = 56) and OMPP (n = 70), with RMPP demonstrating significantly increased IL-18, community-acquired respiratory distress syndrome (CARDS) toxins in nasopharyngeal aspirate, and serum CRP levels (P < 0.001; P = 0.006; P < 0.001). In conclusion, timely recognition of RMPP is crucial for enhancing prognosis. The identification of MRMP, coupled with proinflammatory cytokines such as IL-18, CARDS toxins, and CRP, emerges as promising markers with the potential to contribute significantly to diagnostic accuracy and prognosis assessment.


Asunto(s)
Neumonía por Mycoplasma , Síndrome de Dificultad Respiratoria , Niño , Humanos , Antibacterianos/farmacología , China , Farmacorresistencia Bacteriana/genética , Interleucina-18 , Macrólidos/farmacología , Mycoplasma pneumoniae/genética , Neumonía por Mycoplasma/diagnóstico , Neumonía por Mycoplasma/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Estudios Retrospectivos
13.
Eur J Pharmacol ; 971: 176509, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38493914

RESUMEN

Acute respiratory distress syndrome (ARDS) became a focus of intensive research due to its death toll during the Covid-19 pandemic. An uncontrolled and excessive inflammatory response mediated by proinflammatory molecules such as high mobility group box protein 1 (HMGB1), IL-6, and TNF mounts as a response to infection. In this study, ethyl pyruvate (EP), a known inhibitor of HMGB1, was tested in the model of murine ARDS induced in C57BL/6 mice by intranasal administration of polyinosinic:polycytidylic acid (poly(I:C)). Intraperitoneal administration of EP ameliorated the ARDS-related histopathological changes in the lungs of poly(I:C)-induced ARDS and decreased numbers of immune cells in the lungs, broncho-alveolar lavage fluid and draining lymph nodes (DLN). Specifically, fewer CD8+ T cells and less activated CD4+ T cells were observed in DLN. Consequently, the lungs of EP-treated animals had fewer damage-inflicting CD8+ cells and macrophages. Additionally, the expression and production of proinflammatory cytokines, IL-17, IFN-γ and IL-6 were downregulated in the lungs. The expression of chemokine CCL5 which recruits immune cells into the lungs was also reduced. Finally, EP downregulated the expression of HMGB1 in the lungs. Our results imply that EP should be further evaluated as a potential candidate for ARDS therapy.


Asunto(s)
Proteína HMGB1 , Piruvatos , Síndrome de Dificultad Respiratoria , Humanos , Animales , Ratones , Linfocitos T CD8-positivos/metabolismo , Proteína HMGB1/metabolismo , Interleucina-6 , Pandemias , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Síndrome de Dificultad Respiratoria/tratamiento farmacológico
14.
Int J Mol Sci ; 25(5)2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38474059

RESUMEN

Acute Respiratory Distress Syndrome (ARDS) is characterized by lung inflammation and increased membrane permeability, which represents the leading cause of mortality in ICUs. Mechanical ventilation strategies are at the forefront of supportive approaches for ARDS. Recently, an increasing understanding of RNA biology, function, and regulation, as well as the success of RNA vaccines, has spurred enthusiasm for the emergence of novel RNA-based therapeutics. The most common types of RNA seen in development are silencing (si)RNAs, antisense oligonucleotide therapy (ASO), and messenger (m)RNAs that collectively account for 80% of the RNA therapeutics pipeline. These three RNA platforms are the most mature, with approved products and demonstrated commercial success. Most recently, miRNAs have emerged as pivotal regulators of gene expression. Their dysregulation in various clinical conditions offers insights into ARDS pathogenesis and offers the innovative possibility of using microRNAs as targeted therapy. This review synthesizes the current state of the literature to contextualize the therapeutic potential of miRNA modulation. It considers the potential for miR-based therapeutics as a nuanced approach that incorporates the complexity of ARDS pathophysiology and the multifaceted nature of miRNA interactions.


Asunto(s)
MicroARNs , Neumonía , Síndrome de Dificultad Respiratoria , Humanos , MicroARNs/genética , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Neumonía/complicaciones , Respiración Artificial/efectos adversos
15.
ACS Appl Mater Interfaces ; 16(10): 12244-12262, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38421312

RESUMEN

Acute respiratory distress syndrome (ARDS) is a serious respiratory condition characterized by a damaged pulmonary endothelial barrier that causes protein-rich lung edema, an influx of proinflammatory cells, and treatment-resistant hypoxemia. Damage to pulmonary endothelial cells and inflammation are pivotal in ARDS development with a key role played by endothelial cell pyroptosis. Disulfiram (DSF), a drug that has long been used to treat alcohol addiction, has recently been identified as a potent inhibitor of gasdermin D (GSDMD)-induced pore formation and can thus prevent pyroptosis and inflammatory cytokine release. These findings indicate that DSF is a promising treatment for inflammatory disorders. However, addressing the challenge posed by its intrinsic physicochemical properties, which hinder intravenous administration, and effective delivery to pulmonary vascular endothelial cells are crucial. Herein, we used biocompatible liposomes incorporating a lung endothelial cell-targeted peptide (CGSPGWVRC) to produce DSF-loaded nanoparticles (DTP-LET@DSF NPs) for targeted delivery and reactive oxygen species-responsive release facilitated by the inclusion of thioketal (TK) within the liposomal structure. After intravenous administration, DTP-LET@DSF NPs exhibited excellent cytocompatibility and minor systemic toxicity, effectively inhibited pyroptosis, mitigated lipopolysaccharide (LPS)-induced ARDS, and prevented cytokine storms resulting from excessive immune reactions in ARDS mice. This study presents a straightforward nanoplatform for ARDS treatment that potentially paves the way for the clinical use of this nanomedicine.


Asunto(s)
Disulfiram , Síndrome de Dificultad Respiratoria , Animales , Ratones , Disulfiram/farmacología , Células Endoteliales , Reposicionamiento de Medicamentos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Pulmón , Liposomas/farmacología , Lipopolisacáridos/farmacología
16.
Expert Opin Investig Drugs ; 33(3): 243-277, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38316432

RESUMEN

INTRODUCTION: Acute Respiratory Distress Syndrome (ARDS) is a heterogeneous form of lung injury with severe hypoxemia and bilateral infiltrates after an inciting event that results in diffuse lung inflammation with a high mortality rate. While research in COVID-related ARDS has resulted in several pharmacotherapeutic agents that have undergone successful investigation, non-COVID ARDS studies have not resulted in many widely accepted pharmacotherapeutic agents despite exhaustive research. AREAS COVERED: The aim of this review is to discuss adjuvant pharmacotherapies targeting non-COVID Acute Lung Injury (ALI)/ARDS and novel therapeutics in COVID associated ALI/ARDS. In ARDS, variable data may support selective use of neuromuscular blocking agents, corticosteroids and neutrophil elastase inhibitors, but are not yet universally used. COVID-ALI/ARDS has data supporting the use of IL-6 monoclonal antibodies, corticosteroids, and JAK inhibitor therapy. EXPERT OPINION: Although ALI/ARDS modifying pharmacological agents have been identified in COVID-related disease, the data in non-COVID ALI/ARDS has been less compelling. The increased use of more specific molecular phenotyping based on physiologic parameters and biomarkers, will ensure equipoise between groups, and will likely allow more precision in confirming pharmacological agent efficacy in future studies.


Asunto(s)
Síndrome de Dificultad Respiratoria , Humanos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Corticoesteroides/uso terapéutico , Biomarcadores
17.
Front Immunol ; 15: 1328565, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38312838

RESUMEN

The human respiratory and circulatory systems collaborate intricately to ensure oxygen delivery to all cells, which is vital for ATP production and maintaining physiological functions and structures. During limited oxygen availability, hypoxia-inducible factors (HIFs) are stabilized and play a fundamental role in maintaining cellular processes for hypoxia adaptation. First discovered during investigations of erythropoietin production regulation, HIFs influence physiological and pathological processes, including development, inflammation, wound healing, and cancer. HIFs promote extracellular adenosine signaling by enhancing adenosine generation and receptor signaling, representing an endogenous feedback mechanism that curbs excessive inflammation, supports injury resolution, and enhances hypoxia tolerance. This is especially important for conditions that involve tissue hypoxia, such as acute respiratory distress syndrome (ARDS), which globally poses significant health challenges without specific treatment options. Consequently, pharmacological strategies to amplify HIF-mediated adenosine production and receptor signaling are of great importance.


Asunto(s)
Adenosina , Síndrome de Dificultad Respiratoria , Humanos , Hipoxia/patología , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Inflamación , Oxígeno
18.
Artif Cells Nanomed Biotechnol ; 52(1): 114-121, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38423099

RESUMEN

Acute respiratory distress syndrome (ARDS) is a complication of pulmonary disease that produces life-threatening hypoxaemia. Despite ventilation and hyperoxic therapies, undetected hypoxia can manifest in capillary beds leading to multi-organ failure. Ox66™ is an ingestible, solid-state form of oxygen designed to supplement oxygen deficits. Twenty-four anaesthetized rats underwent a two-hit model of respiratory distress (ARDS), where a single dose (5 mg/kg) of lipopolysaccharide (LPS) was given intratracheally, and then the respiratory tidal volume was reduced by 40%. After 60 min, animals were randomized to receive Ox66™, or normal saline (NS; vehicle control) via gavage or supplemental inspired oxygen (40% FiO2). A second gavage was administered at 120 min. Cardiovascular function and blood oximetry/chemistry were measured alongside the peripheral spinotrapezius muscle's interstitial oxygenation (PISFO2). ARDS reduced mean arterial pressure (MAP) and PISFO2 compared to baseline (BL) for all treatment groups. Treatment with Ox66 or NS did not improve MAP, but 40% FiO2 caused a rapid return to BL. PISFO2 improved after treatment with Ox66™ and 40% FiO2 and remained elevated for both groups against NS until study conclusion. Both oxygen treatments also suppressed the inflammatory response to LPS, suggesting that Ox66™ can deliver therapeutically-impactful levels of oxygen in situations of pulmonary dysfunction.


Asunto(s)
Síndrome de Dificultad Respiratoria , Roedores , Animales , Ratas , Lipopolisacáridos/farmacología , Pulmón , Oxígeno , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Distribución Aleatoria
19.
ACS Appl Mater Interfaces ; 16(7): 8310-8320, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38343060

RESUMEN

The COVID-19 pandemic has become an unprecedented global medical emergency, resulting in more than 5 million deaths. Acute respiratory distress syndrome (ARDS) caused by COVID-19, characterized by the release of a large number of pro-inflammatory cytokines and the production of excessive toxic ROS, is the most common serious complication leading to death. To develop new strategies for treating ARDS caused by COVID-19, a mouse model of ARDS was established by using lipopolysaccharide (LPS). Subsequently, we have constructed a novel nanospray with anti-inflammatory and antioxidant capacity by loading pentoxifylline (PTX) and edaravone (Eda) on zeolite imidazolate frameworks-8 (ZIF-8). This nanospray was endowed with synergetic therapy, which could kill two birds with one stone: (1) the loaded PTX played a powerful anti-inflammatory role by inhibiting the activation of inflammatory cells and the synthesis of pro-inflammatory cytokines; (2) Eda served as a free radical scavenger in ARDS. Furthermore, compared with the traditional intravenous administration, nanosprays can be administered directly and inhaled efficiently and reduce the risk of systemic adverse reactions greatly. This nanospray could not only coload two drugs efficiently but also realize acid-responsive release on local lung tissue. Importantly, ZIF8-EP nanospray showed an excellent therapeutic effect on ARDS in vitro and in vivo, which provided a new direction for the treatment of ARDS.


Asunto(s)
COVID-19 , Pentoxifilina , Síndrome de Dificultad Respiratoria , Animales , Ratones , Humanos , Pentoxifilina/farmacología , Pentoxifilina/uso terapéutico , Edaravona/uso terapéutico , Pandemias , Pulmón , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Citocinas , Concentración de Iones de Hidrógeno , Lipopolisacáridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA