Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 902
Filtrar
Más filtros

Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 210(4): 401-423, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38573068

RESUMEN

Recent genetic and genomic advancements have elucidated the complex etiology of idiopathic pulmonary fibrosis (IPF) and other progressive fibrotic interstitial lung diseases (ILDs), emphasizing the contribution of heritable factors. This state-of-the-art review synthesizes evidence on significant genetic contributors to pulmonary fibrosis (PF), including rare genetic variants and common SNPs. The MUC5B promoter variant is unusual, a common SNP that markedly elevates the risk of early and established PF. We address the utility of genetic variation in enhancing understanding of disease pathogenesis and clinical phenotypes, improving disease definitions, and informing prognosis and treatment response. Critical research gaps are highlighted, particularly the underrepresentation of non-European ancestries in PF genetic studies and the exploration of PF phenotypes beyond usual interstitial pneumonia/IPF. We discuss the role of telomere length, often critically short in PF, and its link to progression and mortality, underscoring the genetic complexity involving telomere biology genes (TERT, TERC) and others like SFTPC and MUC5B. In addition, we address the potential of gene-by-environment interactions to modulate disease manifestation, advocating for precision medicine in PF. Insights from gene expression profiling studies and multiomic analyses highlight the promise for understanding disease pathogenesis and offer new approaches to clinical care, therapeutic drug development, and biomarker discovery. Finally, we discuss the ethical, legal, and social implications of genomic research and therapies in PF, stressing the need for sound practices and informed clinical genetic discussions. Looking forward, we advocate for comprehensive genetic testing panels and polygenic risk scores to improve the management of PF and related ILDs across diverse populations.


Asunto(s)
Genómica , Fibrosis Pulmonar Idiopática , Mucina 5B , Medicina de Precisión , Humanos , Medicina de Precisión/métodos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/terapia , Mucina 5B/genética , Predisposición Genética a la Enfermedad/genética , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/terapia , Polimorfismo de Nucleótido Simple/genética
2.
Proc Natl Acad Sci U S A ; 119(15): e2121098119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35377803

RESUMEN

The pathogenesis of lung fibrosis involves hyperactivation of innate and adaptive immune pathways that release inflammatory cytokines and growth factors such as tumor growth factor (TGF)ß1 and induce aberrant extracellular matrix protein production. During the genesis of pulmonary fibrosis, resident alveolar macrophages are replaced by a population of newly arrived monocyte-derived interstitial macrophages that subsequently transition into alveolar macrophages (Mo-AMs). These transitioning cells initiate fibrosis by releasing profibrotic cytokines and remodeling the matrix. Here, we describe a strategy for leveraging the up-regulation of the mannose receptor CD206 in interstitial macrophages and Mo-AM to treat lung fibrosis. We engineered mannosylated albumin nanoparticles, which were found to be internalized by fibrogenic CD206+ monocyte derived macrophages (Mo-Macs). Mannosylated albumin nanoparticles incorporating TGFß1 small-interfering RNA (siRNA) targeted the profibrotic subpopulation of CD206+ macrophages and prevented lung fibrosis. The findings point to the potential utility of mannosylated albumin nanoparticles in delivering TGFß-siRNA into CD206+ profibrotic macrophages as an antilung fibrosis strategy.


Asunto(s)
Linfotoxina-alfa , Macrófagos Alveolares , Nanopartículas , Fibrosis Pulmonar , ARN Interferente Pequeño , Animales , Bleomicina/farmacología , Modelos Animales de Enfermedad , Linfotoxina-alfa/genética , Macrófagos Alveolares/inmunología , Receptor de Manosa , Ratones , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/terapia , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética
3.
Small ; 20(22): e2308775, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38126895

RESUMEN

RNA interference (RNAi) is an efficient strategy to post-transcriptionally silence gene expression. While all siRNA drugs on the market target the liver, the lung offers a variety of currently undruggable targets, which can potentially be treated with RNA therapeutics. To achieve this goal, the synthesis of poly(spermine acrylamides) (P(SpAA) is reported herein. Polymers are prepared via polymerization of N-acryloxysuccinimide (NAS) and afterward this active ester is converted into spermine-based pendant groups. Copolymerizations with decylacrylamide are employed to increase the hydrophobicity of the polymers. After deprotection, polymers show excellent siRNA encapsulation to obtain perfectly sized polyplexes at very low polymer/RNA ratios. In vitro 2D and 3D cell culture, ex vivo and in vivo experiments reveal superior properties of amphiphilic spermine-copolymers with respect to delivery of siRNA to lung cells in comparison to commonly used lipid-based transfection agents. In line with the in vitro results, siRNA delivery to human lung explants confirm more efficient gene silencing of protease-activated receptor 2 (PAR2), a G protein-coupled receptor involved in fibrosis. This study reveals the importance of the balance between efficient polyplex formation, cellular uptake, gene knockdown, and toxicity for efficient siRNA delivery in vitro, in vivo, and in fibrotic human lung tissue ex vivo.


Asunto(s)
Fibrosis Pulmonar , ARN Interferente Pequeño , Espermina , Espermina/química , Espermina/farmacología , Humanos , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/terapia , Animales , Pulmón/patología , Pulmón/metabolismo , Polímeros/química , Acrilamidas/química
4.
Stem Cells ; 41(10): 928-943, 2023 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-37419489

RESUMEN

This study was performed to determine the effect of human umbilical cord mesenchymal stem cells (hucMSCs) treatment on pulmonary fibrosis and investigate the circFOXP1-mediated autophagic mechanism of hucMSCs treatment. Pulmonary fibrosis models were established by spraying bleomycin in mice and TGF-ß1 treatment of MRC-5 cells. Results showed that hucMSCs were retained in lung and hucMSCs treatment alleviated pulmonary fibrosis. Morphological staining indicated that hucMSCs-treated mice had thinner alveolar walls, effectively improved alveolar structure, significantly reduced alveolar inflammation, and decreased collagen deposition than control mice. Fibrotic proteins, including vimentin, α-SMA, collagens I and III, and the differentiation-related protein S100 calcium-binding protein A4 was reduced considerably in the hucMSCs-treated group. The mechanistic study revealed that the inhibition of hucMSCs treatment on pulmonary fibrogenesis depended on downregulating circFOXP1, in which hucMSCs treatment promoted circFOXP1-mediated autophagy process via blocking the nuclear human antigen R (HuR) translocation and promoting the HuR degradation, leading to a marked decrease in autophagy negative regulators EZH2, STAT1, and FOXK1. In conclusion, hucMSCs treatment significantly improved pulmonary fibrosis by downregulating the circFOXP1-HuR-EZH2/STAT1/FOXK1 autophagic axis. hucMSCs can act as an effective treatment for pulmonary fibrosis.


Asunto(s)
Células Madre Mesenquimatosas , Fibrosis Pulmonar , Ratones , Humanos , Animales , Fibrosis Pulmonar/terapia , Fibrosis , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Autofagia , Cordón Umbilical , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Factor de Transcripción STAT1 , Factores de Transcripción Forkhead/metabolismo
5.
Respir Res ; 25(1): 395, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39487454

RESUMEN

Comprehensive care integrates individual patient needs and is highly valued for patients with pulmonary fibrosis (PF). The importance of a patient-centered care approach is rooted in the unpredictable progressiveness of the disease course in PF. The respiratory impairment associated with PF has a major impact on the quality of life for both patients and their caregivers. We believe that prioritizing patient preferences could improve the shared decision making process and may ultimately lead to better health outcomes. Despite the growing emphasis for this approach, it remains challenging to adopt it in clinical practice. In this review, we propose the comprehensive Triple A Care Model, consisting of the domains Access, Anticipate, and Act, which emphasizes core elements of patient-centered care for patients with PF. We will provide an overview of the unmet needs in care for patients with PF and elaborate on the current methods for delivering patient-centered care. The latest insights into symptom management and supportive measures and several approaches to improving access to care are discussed, in line with the most recent guidelines.


Asunto(s)
Accesibilidad a los Servicios de Salud , Atención Dirigida al Paciente , Fibrosis Pulmonar , Humanos , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/diagnóstico , Accesibilidad a los Servicios de Salud/tendencias , Calidad de Vida , Prioridad del Paciente
6.
Respir Res ; 25(1): 170, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637860

RESUMEN

While the COVID-19 outbreak and its complications are still under investigation, post-inflammatory pulmonary fibrosis (PF) has already been described as a long-term sequela of acute respiratory distress syndrome (ARDS) secondary to SARS-CoV2 infection. However, therapeutical strategies for patients with ARDS and PF are still limited and do not significantly extend lifespan. So far, lung transplantation remains the only definitive treatment for end-stage PF. Over the last years, numerous preclinical and clinical studies have shown that allogeneic mesenchymal stromal cells (MSCs) might represent a promising therapeutical approach in several lung disorders, and their potential for ARDS treatment and PF prevention has been investigated during the COVID-19 pandemic. From April 2020 to April 2022, we treated six adult patients with moderate COVID-19-related ARDS in a late proliferative stage with up to two same-dose infusions of third-party allogeneic bone marrow-derived MSCs (BM-MSCs), administered intravenously 15 days apart. No major adverse events were registered. Four patients completed the treatment and reached ICU discharge, while two received only one dose of MSCs due to multiorgan dysfunction syndrome (MODS) and subsequent death. All four survivors showed improved gas exchanges (PaO2/FiO2 ratio > 200), contrary to the others. Furthermore, LDH trends after MSCs significantly differed between survivors and the deceased. Although further investigations and shared protocols are still needed, the safety of MSC therapy has been recurrently shown, and its potential in treating ARDS and preventing PF might represent a new therapeutic strategy.


Asunto(s)
COVID-19 , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Fibrosis Pulmonar , Síndrome de Dificultad Respiratoria , Adulto , Humanos , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/etiología , Pandemias , ARN Viral , Síndrome de Dificultad Respiratoria/terapia , Síndrome de Dificultad Respiratoria/etiología , COVID-19/terapia , Trasplante de Células Madre Mesenquimatosas/métodos
7.
Cytotherapy ; 26(8): 930-938, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38520411

RESUMEN

BACKGROUND AIMS: Rheumatoid arthritis (RA) is characterized by an overactive immune system, with limited treatment options beyond immunosuppressive drugs or biological response modifiers. Human embryonic stem cell-derived mesenchymal stromal cells (hESC-MSCs) represent a novel alternative, possessing diverse immunomodulatory effects. In this study, we aimed to elucidate the therapeutic effects and underlying mechanisms of hESC-MSCs in treating RA. METHODS: MSC-like cells were differentiated from hESC (hESC-MSCs) and cultured in vitro. Cell proliferation was assessed using Cell Counting Kit-8 assay and Ki-67 staining. Flow cytometry was used to analyze cell surface markers, T-cell proliferation and immune cell infiltration. The collagen-induced arthritis (CIA) mouse model and bleomycin-induced model of lung fibrosis (BLE) were established and treated with hESC-MSCs intravenously for in vivo assessment. Pathological analyses, reverse transcription-quantitative polymerase chain reaction and Western blotting were conducted to evaluate the efficacy of hESC-MSCs treatment. RESULTS: Intravenous transplantation of hESC-MSCs effectively reduced inflammation in CIA mice in this study. Furthermore, hESC-MSC administration enhanced regulatory T cell infiltration and activation. Additional findings suggest that hESC-MSCs may reduce lung fibrosis in BLE mouse models, indicating their potential to mitigate complications associated with RA progression. In vitro experiments revealed a significant inhibition of T-cell activation and proliferation during co-culture with hESC-MSCs. In addition, hESC-MSCs demonstrated enhanced proliferative capacity compared with traditional primary MSCs. CONCLUSIONS: Transplantation of hESC-MSCs represents a promising therapeutic strategy for RA, potentially regulating T-cell proliferation and differentiation.


Asunto(s)
Artritis Reumatoide , Diferenciación Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias Humanas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Fibrosis Pulmonar , Animales , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Artritis Reumatoide/terapia , Artritis Reumatoide/inmunología , Trasplante de Células Madre Mesenquimatosas/métodos , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/patología , Células Madre Embrionarias Humanas/citología , Proliferación Celular , Inflamación/terapia , Inflamación/patología , Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Artritis Experimental/terapia , Artritis Experimental/patología , Artritis Experimental/inmunología
8.
Cytotherapy ; 26(10): 1236-1244, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38852093

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is characterized by progressive lung interstitial lesions with the disease pathophysiology incompletely understood, which is a serious and fatal disorder with limited treatment options. Mesenchymal stem cells (MSCs) have exhibited promising therapeutic capability for IPF. While most types of MSCs are obtained invasively, urine-derived stem cells (USCs) can be gained in a safe, noninvasive, and inexpensive procedure, which are readily available and reported to exhibit no risk of teratoma formation or oncogenic potential in vivo, sounding alternative to other MSCs. This study aims to investigate the therapeutic effect and mechanism of USCs on IPF, using a bleomycin (BLM)-induced IPF model in mice. METHODS: Cell surface marker examination by flow cytometry analysis and cell differentiation culture were used to characterize USCs obtained from healthy individuals. BLM was instilled endotracheally in adult C57BL/6 mice, followed by USCs or human bone marrow-derived mesenchymal stem cells (BMSCs) treatment by tail vein injection on day 14. Mice were euthanized on day 14 before administration or day 21 for the evaluation of pulmonary histopathology and hydroxyproline (HYP) content. Inflammatory factors of the lung, including transforming growth factor (TGF)-ß1, TNF-α, IL-6, MMP2 were analyzed by quantitative real-time PCR (qRT-PCR). Additionally, immunohistochemistry (IHC) and western blotting (WB) were applied to evaluate the expression of α-SMA and activation of TGF-ß1-Smad2/3 in lung. RESULTS: USCs highly expressed CD29 and CD90, showing negative expression of hematopoietic stem cell markers (CD45, CD34) and could differentiate into, at least, bone and fat in vitro. In mice challenged with BLM, septal thickening and prominent fibrosis were observed on day 14, with higher HYP content and mRNA levels of TGF-ß1, TNF-α and IL-6 exhibited, compared to untreated mice. USCs could migrate to lung and accumulate there in mouse model after intravenous injection. Transplantation of USCs into BLM-induced mice improved their pulmonary histopathology, decreasing Ashcroft score, Szapiel score, HYP content and mRNA levels of TGF-ß1 and MMP2 of lung, similar to the effects of BMSCs. IHC and WB further revealed that USCs could inhibit activation of the TGF­ß1-Smad2/3 pathway of lung in vivo. CONCLUSIONS: Transplantation of USCs effectively reverses pulmonary fibrotic phenotype in an experimental IPF model, inhibiting the TGF-ß1-Smad2/3 pathway, a key driver of fibrosis. These results suggest the therapeutic application of USCs for IPF, instead of other types of MSCs obtained invasively.


Asunto(s)
Bleomicina , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Factor de Crecimiento Transformador beta1 , Animales , Ratones , Factor de Crecimiento Transformador beta1/metabolismo , Proteína Smad2/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Proteína smad3/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Transducción de Señal/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Diferenciación Celular/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Masculino , Fibrosis Pulmonar Idiopática/terapia , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología
9.
Pediatr Res ; 95(6): 1543-1552, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38245633

RESUMEN

BACKGROUND: Pulmonary fibrosis (PF) is a progressive lung disorder with a high mortality rate; its therapy remains limited due to the inefficiency of drug delivery. In this study, the system of drug delivery of nintedanib (Nin) by exosomes derived from adipose-derived stem cells (ADSCs-Exo, Exo) was developed to effectively deliver Nin to lung lesion tissue to ensure enhanced anti-fibrosis therapy. METHODS: The bleomycin (BLM)-induced PF model was constructed in vivo and in vitro. The effects of Exo-Nin on BLM-induced PF and its regulatory mechanism were examined using RT-qPCR, Western blotting, immunofluorescence, and H&E staining. RESULTS: We found Exo-Nin significantly improved BLM-induced PF in vivo and in vitro compared to Nin and Exo groups alone. Mechanistically, Exo-Nin alleviated fibrogenesis by suppressing endothelial-mesenchymal transition through the down-regulation of the TGF-ß/Smad pathway and the attenuation of oxidative stress in vivo and in vitro. CONCLUSIONS: Utilizing adipose stem cell-derived exosomes as carriers for Nin exhibited a notable enhancement in therapeutic efficacy. This improvement can be attributed to the regenerative properties of exosomes, indicating promising prospects for adipose-derived exosomes in cell-free therapies for PF. IMPACT: The system of drug delivery of nintedanib (Nin) by exosomes derived from adipose-derived stem cells was developed to effectively deliver Nin to lung lesion tissue to ensure enhanced anti-fibrosis therapy. The use of adipose stem cell-derived exosomes as the carrier of Nin may increase the therapeutic effect of Nin, which can be due to the regenerative properties of the exosomes and indicate promising prospects for adipose-derived exosomes in cell-free therapies for PF.


Asunto(s)
Bleomicina , Exosomas , Indoles , Fibrosis Pulmonar , Exosomas/metabolismo , Exosomas/trasplante , Animales , Indoles/farmacología , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/metabolismo , Ratones , Tejido Adiposo/citología , Células Madre/citología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Pulmón/patología , Pulmón/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Humanos , Transducción de Señal , Masculino , Sistemas de Liberación de Medicamentos
10.
Curr Opin Pulm Med ; 30(5): 508-515, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38946547

RESUMEN

PURPOSE OF REVIEW: Pulmonary fibrosis (PF) negatively influences health-related quality of life (HRQOL). Patients living with PF have voiced the desire for a focus on symptoms and HRQOL in both disease monitoring and treatment decisions. RECENT FINDINGS: Currently available disease modifying treatments do little to impact HRQOL. Newer studies evaluating pharmacologic and nonpharmacologic therapies targeting symptoms and HRQOL in PF have been conducted with some promising results. There is increasing recognition of the importance of incorporating HRQOL as a higher tier endpoint in clinical trials. Disease-specific measure of HRQOL have been developed for those living with PF, and there is ongoing work to better understand the validity and reliability characteristics of these tools. In addition to research, there is recognition of the potential benefits of measuring HRQOL and symptoms in clinical practice in facilitate integrating patient perspective into care and allow for more personalized treatment approaches. SUMMARY: There is increased momentum to discover treatments that impact HRQOL in PF. More work is desperately needed to identify better treatment targets, and to incorporate HRQOL and symptoms as higher tier endpoints in clinical trials. Further work is also needed to address the practicalities of integrating HRQOL measurement into clinical care.


Asunto(s)
Fibrosis Pulmonar , Calidad de Vida , Humanos , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/psicología , Reproducibilidad de los Resultados
11.
Curr Opin Pulm Med ; 30(5): 530-539, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38963163

RESUMEN

PURPOSE OF REVIEW: Updated clinical practice guidelines for pulmonary fibrosis recommend early integrated palliative care using a primary palliative approach. Clinicians need to be aware of the various palliative interventions in order to implement guidelines' recommendations. This review provides an update on evidence-based palliative therapies. RECENT FINDINGS: Literature review indicates early integration of palliative care in pulmonary fibrosis is feasible and meets patient needs. Key components of a primary palliative approach include comprehensive symptoms and needs screening, systematic symptom management using nonpharmacologic interventions, supplemental oxygen and opioids for dyspnea and cough. Patient-centered communication is essential for successful integration of palliative care. Early, iterative advance care planning in clinic to understand patient goals, values and preferences for current and future care, improves patient care and satisfaction. Prioritizing caregiver inclusion in clinics can address their needs. Collaborating with a multidisciplinary allied team facilitates integration of palliative care and supports patients throughout the disease course. Different models of palliative care delivery exist and can be adapted for local use. The use of artificial intelligence algorithms and tools may improve care and continuity. SUMMARY: Clinicians must develop competency in palliative care. Organizational and policy support is essential to enable seamless integration of palliative care throughout the care continuum.


Asunto(s)
Cuidados Paliativos , Fibrosis Pulmonar , Humanos , Cuidados Paliativos/métodos , Fibrosis Pulmonar/terapia , Planificación Anticipada de Atención , Guías de Práctica Clínica como Asunto , Atención Dirigida al Paciente
12.
Curr Opin Pulm Med ; 30(5): 523-529, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38913018

RESUMEN

PURPOSE OF REVIEW: To review the current understanding of the impact, mechanisms and treatments for cough in patients with interstitial lung disease (ILD). Evidence suggests that cough is a prevalent symptom in patients with ILD and has a significant impact on patients. RECENT FINDINGS: There is increasing interest in the role of cough hypersensitivity as seen in chronic refractory cough in patients with ILD, and encouraging recent results suggest that ILD-associated cough responds to opiate therapy. SUMMARY: Understanding the aetiology of cough in patients with ILD is crucial to continue to develop therapies which might be effective in reducing cough and increasing quality of life.


Asunto(s)
Tos , Enfermedades Pulmonares Intersticiales , Calidad de Vida , Humanos , Tos/etiología , Tos/terapia , Enfermedades Pulmonares Intersticiales/terapia , Enfermedades Pulmonares Intersticiales/complicaciones , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/terapia , Enfermedad Crónica
13.
Cell Biol Int ; 48(11): 1714-1730, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39023281

RESUMEN

Pulmonary fibrosis, a debilitating lung disorder characterised by excessive fibrous tissue accumulation in lung parenchyma, compromises respiratory function leading to a life-threatening respiratory failure. While its origins are multifaceted and poorly understood, the urokinase system, including urokinase-type plasminogen activator (uPA) and its receptor (uPAR), plays a significant role in regulating fibrotic response, extracellular matrix remodelling, and tissue repair. Mesenchymal stem/stromal cells (MSCs) hold promise in regenerative medicine for treating pulmonary fibrosis. Our study aimed to investigate the potential of MSCs to inhibit pulmonary fibrosis as well as the contribution of uPAR expression to this effect. We found that intravenous MSC administration significantly reduced lung fibrosis in the bleomycin-induced pulmonary fibrosis model in mice as revealed by MRI and histological evaluations. Notably, administering the MSCs isolated from adipose tissue of uPAR knockout mice (Plaur-/- MSCs) attenuated lung fibrosis to a lesser extent as compared to WT MSCs. Collagen deposition, a hallmark of fibrosis, was markedly reduced in lungs treated with WT MSCs versus Plaur-/- MSCs. Along with that, endogenous uPA levels were affected differently; after Plaur-/- MSCs were administered, the uPA content was specifically decreased within the blood vessels. Our findings support the potential of MSC treatment in attenuating pulmonary fibrosis. We provide evidence that the observed anti-fibrotic effect depends on uPAR expression in MSCs, suggesting that uPAR might counteract the uPA accumulation in lungs.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Fibrosis Pulmonar , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Animales , Masculino , Ratones , Bleomicina , Modelos Animales de Enfermedad , Pulmón/patología , Pulmón/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética
14.
J Nanobiotechnology ; 22(1): 662, 2024 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-39462403

RESUMEN

BACKGROUND: Radiation-induced lung injury (RILI) is associated with alveolar epithelial cell death and secondary fibrosis in injured lung. Mesenchymal stem cell (MSC)-derived exosomes have regenerative effect against lung injury and the potential to intervene of RILI. However, their intervention efficacy is limited because they lack lung targeting characters and do not carry sufficient specific effectors. SARS-CoV-2 spike glycoprotein (SARS-CoV-2-S-RBD) binds angiotensin-converting enzyme 2 (ACE2) receptor and mediates interaction with host cells. MiR-486-5p is a multifunctional miRNA with angiogenic and antifibrotic potential and acts as an effector in MSC-derived exosomes. Ferroptosis is a form of cell death associated with radiation injury, its roles and mechanisms in RILI remain unclear. In this study, we developed an engineered MSC-derived exosomes with SARS-CoV-2-S-RBD- and miR-486-5p- modification and investigated their intervention effects on RIPF and action mechanisms via suppression of epithelial cell ferroptosis. RESULTS: Adenovirus-mediated gene modification led to miR-486-5p overexpression in human umbilical cord MSC exosomes (p < 0.05), thereby constructing miR-486-5p engineered MSC exosomes (miR-486-MSC-Exo). MiR-486-MSC-Exo promoted the proliferation and migration of irradiated mouse lung epithelial (MLE-12) cells in vitro and inhibited RILI in vivo (all p < 0.05). MiR-486-MSC-Exo suppressed ferroptosis in MLE-12 cells, and an in vitro assay revealed that the expression of fibrosis-related genes is up-regulated following ferroptosis (both p < 0.05). MiR-486-MSC-Exo reversed the up-regulated expression of fibrosis-related genes induced by TGF-ß1 in vitro and improved pathological fibrosis in RIPF mice in vivo (all p < 0.05). SARS-CoV-2-S-RBD-modified and miR-486-5p-engineered MSC exosomes (miR-486-RBD-MSC-Exo) were also constructed, and the distribution of DiR dye-labeled miR-486-RBD-MSC-Exo in hACE2CKI/CKI Sftpc-Cre+ mice demonstrated long-term retention in the lung (p < 0.05). MiR-486-RBD-MSC-Exo significantly improved the survival rate and pathological changes in hACE2CKI/CKI Sftpc-Cre+ RIPF mice (all p < 0.05). Furthermore, miR-486-MSC-Exo exerted anti-fibrotic effects via targeted SMAD2 inhibition and Akt phosphorylation activation (p < 0.05). CONCLUSIONS: Engineered MSC exosomes with SARS-CoV-2-S-RBD- and miR-486-5p-modification were developed. MiR-486-RBD-MSC-Exo suppressed ferroptosis and fibrosis of MLE-12 cells in vitro, and alleviated RILI and long-term RIPF in ACE2 humanized mice in vivo. MiR-486-MSC-Exo exerted anti-fibrotic effects via SMAD2 inhibition and Akt activation. This study provides a potential approach for RIPF intervention.


Asunto(s)
Exosomas , Ferroptosis , Células Madre Mesenquimatosas , MicroARNs , Fibrosis Pulmonar , SARS-CoV-2 , Exosomas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Animales , Células Madre Mesenquimatosas/metabolismo , Humanos , Ratones , Fibrosis Pulmonar/terapia , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Lesión Pulmonar/terapia , COVID-19/terapia , Pulmón/patología , Traumatismos por Radiación/terapia , Traumatismos por Radiación/metabolismo , Masculino
15.
BMC Pulm Med ; 24(1): 14, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38178092

RESUMEN

BACKGROUND: Pulmonary fibrosis (PF) is a progressive fibrosing interstitial pneumonia that leads to respiratory failure and other complications, which is ultimately fatal. Mesenchymal stem cells (MSCs) transplant is a promising strategy to solve this problem, while the procurement of MSCs from the patient for autotransplant remains a challenge. METHODS: Here, we presented olfactory mucosa mesenchymal stem cells (OM-MSCs) from mouse turbinate and determined the preventing efficacy of allotransplant for PF. We demonstrated the antiinflammation and immunomodulatory effects of OM-MSCs. Flow cytometric analysis was used to verify the effect of OM-MSCs on monocyte-derived macrophage populations in the lung. RESULTS: Administration of OM-MSCs reduces inflammation, attenuates the matrix metallopeptidase 13 (MMP13) expression level and restores the bleomycin (BLM)-induced pulmonary fibrosis by assessing the architecture of lung, collagen type I; (COL1A1), actin alpha 2, smooth muscle, aorta (ACTA2/α-SMA) and hydroxyproline. This therapeutic effect of OM-MSCs was related to the increase in the ratio of nonclassical monocytes to proinflammatory monocytes in the lung. CONCLUSIONS: This study suggests that transplant of OM-MSCs represents an effective and safe treatment for PF.


Asunto(s)
Células Madre Mesenquimatosas , Fibrosis Pulmonar , Humanos , Ratones , Animales , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/metabolismo , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Inmunomodulación , Mucosa Olfatoria/metabolismo
16.
Allergol Immunopathol (Madr) ; 52(4): 53-59, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38970265

RESUMEN

BACKGROUND: Pulmonary fibrosis is a pathological hallmark of lung injury. It is an aggressive disease that replaces normal lung parenchyma by fibrotic tissue. The transforming growth factor-beta-mothers against decapentaplegic homolog 3 (TGF-ß1-Smad3) signaling pathway plays a key role in regulating lung fibrosis. Decorin (DCN), a small leucine-rich proteoglycan, has a modulatory effect on the immune system by reversibly binding with TGF-ß and reducing its bioavailability. Mesenchymal stem cell (MSC) therapy is a new strategy that has an immune-modulatory capacity. OBJECTIVE: The aim of this study was to introduce a new therapeutic approach to harness remodeling in injured lung. MATERIAL AND METHODS: Bone marrow MSCs were isolated and transduced by decorin gene. Lung injury was induced by bleomycin and mice were treated with MSCs, MSCs-decorin, and decorin. Then, oxidative stress biomarkers, remodeling biomarkers, bronchoalveolar lavage cells, and histopathology study were conducted. RESULTS: Reduced catalase and superoxide dismutase increased due to treatments. Elevated malondialdehyde, hydroxyproline, TGF-ß levels, and polymorphonuclear cells count decreased in the treated groups. Additionally, the histopathology of lung tissues showed controlled inflammation and fibrosis. CONCLUSION: Transfected decorin gene to MSCs and used cell therapy could control remodeling and bleomycin-induced lung injury.


Asunto(s)
Bleomicina , Decorina , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Fibrosis Pulmonar , Decorina/genética , Decorina/metabolismo , Animales , Ratones , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/terapia , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/terapia , Lesión Pulmonar/inmunología , Lesión Pulmonar/genética , Transducción Genética , Estrés Oxidativo , Células Cultivadas , Modelos Animales de Enfermedad , Masculino , Humanos
17.
Microsc Microanal ; 30(3): 539-551, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38758132

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease of unknown origin with limited treatment options and poor prognosis. The encouraging findings from preclinical investigations utilizing mesenchymal stem cells (MSCs) indicated that they could serve as a promising therapeutic alternative for managing chronic lung conditions, such as IPF. The objective of this study was to compare the efficiency of bone marrow-derived MSCs (BM-MSCs) versus prednisolone, the standard anti-inflammatory medication, in rats with bleomycin (BLM)-induced lung fibrosis. Four groups were created: a control group, a BLM group, a prednisolone-treated group, and a BM-MSCs-treated group. To induce lung fibrosis, 5 mg/kg of BLM was administered intratracheally. BLM significantly increased serum levels of pro-inflammatory cytokines and oxidative stress markers. The disturbed lung structure was also revealed by light and transmission electron microscopic studies. Upregulation in the immune expression of alpha-smooth muscle actin, transforming growth factor beta-1, and Bax was demonstrated. Interestingly, all findings significantly regressed on treatment with prednisolone and BM-MSCs. However, treatment with BM-MSCs showed better results than with prednisolone. In conclusion, BM-MSCs could be a promising approach for managing lung fibrosis.


Asunto(s)
Bleomicina , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas , Prednisolona , Fibrosis Pulmonar , Animales , Prednisolona/uso terapéutico , Prednisolona/farmacología , Ratas , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/patología , Pulmón/patología , Inmunohistoquímica , Masculino , Citocinas/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Trasplante de Células Madre Mesenquimatosas/métodos , Histocitoquímica , Células de la Médula Ósea , Microscopía Electrónica de Transmisión
18.
Int J Mol Sci ; 25(20)2024 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-39457053

RESUMEN

Hermansky-Pudlak syndrome (HPS)-associated pulmonary fibrosis (HPS-PF) is a progressive lung disease that is a major cause of morbidity and mortality in HPS patients. Previous studies have demonstrated that the HPS proteins play an essential role in the biogenesis and function of lysosome-related organelles (LROs) in alveolar epithelial type II (AT2) cells and found that HPS-PF is associated with dysfunction of AT2 cells and abnormal immune reactions. Despite recent advances in research on HPS and the pathology of HPS-PF, the pathological mechanisms underlying HPS-PF remain poorly understood, and no effective treatment has been established. Therefore, it is necessary to refresh the progress in the pathogenesis of HPS-PF to increase our understanding of the pathogenic mechanism of HPS-PF and develop targeted therapeutic strategies. This review summarizes the recent progress in the pathogenesis of HPS-PF provides information about the current treatment strategies for HPS-PF, and hopefully increases our understanding of the pathogenesis of HPS-PF and offers thoughts for new therapeutic interventions.


Asunto(s)
Síndrome de Hermanski-Pudlak , Fibrosis Pulmonar , Síndrome de Hermanski-Pudlak/terapia , Síndrome de Hermanski-Pudlak/genética , Síndrome de Hermanski-Pudlak/complicaciones , Humanos , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/patología , Animales , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología
19.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673961

RESUMEN

Mesenchymal stem cell-derived exosomes (MSC-Exos) are nano-sized extracellular vesicles which contain various MSC-sourced anti-fibrotic, immunoregulatory and angio-modulatory proteins (growth factors, immunoregulatory cytokines, chemokines), lipids, and nucleic acids (messenger RNA and microRNAs). Due to their lipid envelope, MSC-Exos easily by-pass all barriers in the body and deliver their cargo directly in target cells, modulating their viability, proliferation, phenotype and function. The results obtained in recently published experimental studies demonstrated beneficial effects of MSC-Exos in the treatment of lung fibrosis. MSC-Exos reduced activation of fibroblasts and prevented their differentiation in myofibroblasts. By delivering MSC-sourced immunoregulatory factors in lung-infiltrated monocytes and T cells, MSC-Exos modulate their function, alleviating on-going inflammation and fibrosis. MSC-Exos may also serve as vehicles for the target delivery of anti-fibrotic and immunomodulatory agents, enabling enhanced attenuation of lung fibrosis. Although numerous pre-clinical studies have demonstrated the therapeutic potential of MSC-Exos in the treatment of pulmonary fibrosis, there are several challenges that currently hinder their clinical implementation. Therefore, in this review article, we summarized current knowledge and we discussed future perspectives regarding molecular and cellular mechanisms which were responsible for the anti-fibrotic, anti-inflammatory and immunoregulatory properties of MSC-Exos, paving the way for their clinical use in the treatment of lung fibrosis.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Fibrosis Pulmonar , Exosomas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Fibrosis Pulmonar/terapia , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Animales
20.
Zhonghua Jie He He Hu Xi Za Zhi ; 47(6): 571-575, 2024 Jun 12.
Artículo en Zh | MEDLINE | ID: mdl-38858210

RESUMEN

COVID-19 is caused by the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and manifests primarily as acute lung injury with diffuse interstitial lung disease evident in imaging. Patients often present with clinical features similar to those of autoimmune diseases and share imaging, treatment and serological similarities with autoimmune-related interstitial lung diseases. The association between autoimmune abnormalities and post-COVID-19 pulmonary fibrosis is also recognized. This article provided a comprehensive review of the pathogenic mechanisms, clinical manifestations, and therapeutic interventions associated with autoimmune abnormalities induced by SARS-CoV-2 infection.


Asunto(s)
Enfermedades Autoinmunes , COVID-19 , Enfermedades Pulmonares Intersticiales , SARS-CoV-2 , Humanos , COVID-19/complicaciones , COVID-19/inmunología , COVID-19/terapia , Enfermedades Pulmonares Intersticiales/etiología , Enfermedades Pulmonares Intersticiales/inmunología , Enfermedades Pulmonares Intersticiales/terapia , Enfermedades Autoinmunes/terapia , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA