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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38564376

RESUMEN

RATIONALE: The chronic lung disease bronchopulmonary dysplasia (BPD) is the most severe complication of extreme prematurity. BPD results in impaired lung alveolar and vascular development and long-term respiratory morbidity, for which only supportive therapies exist. Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) improve lung structure and function in experimental BPD. Results of clinical trials with MSCs for many disorders do not yet match the promising preclinical studies. A lack of specific criteria to define functionally distinct MSCs persists. OBJECTIVES: To determine and correlate single-cell UC-MSC transcriptomic profile with therapeutic potential. METHODS: UC-MSCs from five term donors and human neonatal dermal fibroblasts (HNDFs, control cells of mesenchymal origin) transcriptomes were investigated by single-cell RNA sequencing analysis (scRNA-seq). The lung-protective effect of UC-MSCs with a distinct transcriptome and control HNDFs was tested in vivo in hyperoxia-induced neonatal lung injury in rats. MEASUREMENTS AND MAIN RESULTS: UC-MSCs showed limited transcriptomic heterogeneity, but were different from HNDFs. Gene ontology enrichment analysis revealed distinct - progenitor-like and fibroblast-like - UC-MSC subpopulations. Only the treatment with progenitor-like UC-MSCs improved lung function and structure and attenuated pulmonary hypertension in hyperoxia-exposed rat pups. Moreover, scRNA-seq identified major histocompatibility complex class I as a molecular marker of non-therapeutic cells and associated with decreased lung retention. CONCLUSIONS: UC-MSCs with a progenitor-like transcriptome, but not with a fibroblast-like transcriptome, provide lung protection in experimental BPD. High expression of major histocompatibility complex class I is associated with reduced therapeutic benefit. scRNA-seq may be useful to identify subsets of MSCs with superior repair capacity for clinical application.

2.
Stem Cells ; 40(5): 479-492, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35445270

RESUMEN

Late lung development is a period of alveolar and microvascular formation, which is pivotal in ensuring sufficient and effective gas exchange. Defects in late lung development manifest in premature infants as a chronic lung disease named bronchopulmonary dysplasia (BPD). Numerous studies demonstrated the therapeutic properties of exogenous bone marrow and umbilical cord-derived mesenchymal stromal cells (MSCs) in experimental BPD. However, very little is known regarding the regenerative capacity of resident lung MSCs (L-MSCs) during normal development and in BPD. In this study we aimed to characterize the L-MSC population in homeostasis and upon injury. We used single-cell RNA sequencing (scRNA-seq) to profile in situ Ly6a+ L-MSCs in the lungs of normal and O2-exposed neonatal mice (a well-established model to mimic BPD) at 3 developmental timepoints (postnatal days 3, 7, and 14). Hyperoxia exposure increased the number and altered the expression profile of L-MSCs, particularly by increasing the expression of multiple pro-inflammatory, pro-fibrotic, and anti-angiogenic genes. In order to identify potential changes induced in the L-MSCs transcriptome by storage and culture, we profiled 15 000 Ly6a+ L-MSCs after in vitro culture. We observed great differences in expression profiles of in situ and cultured L-MSCs, particularly those derived from healthy lungs. Additionally, we have identified the location of Ly6a+/Col14a1+ L-MSCs in the developing lung and propose Serpinf1 as a novel, culture-stable marker of L-MSCs. Finally, cell communication analysis suggests inflammatory signals from immune and endothelial cells as main drivers of hyperoxia-induced changes in L-MSCs transcriptome.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Células Madre Mesenquimatosas , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/terapia , Células Endoteliales , Humanos , Hiperoxia/genética , Hiperoxia/metabolismo , Recién Nacido , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Análisis de Secuencia de ARN
3.
Ann Surg ; 275(3): e586-e595, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33055583

RESUMEN

OBJECTIVE: To evaluate the effect of combining antenatal sildenafil with fetal tracheal occlusion (TO) in fetal rabbits with surgically induced congenital diaphragmatic hernia (CDH). BACKGROUND: Although antenatal sildenafil administration rescues vascular abnormalities in lungs of fetal rabbits with CDH, it only partially improves airway morphometry. We hypothesized that we could additionally stimulate lung growth by combining this medical treatment with fetal TO. METHODS: CDH was created on gestational day (GD)23 (n=54). Does were randomized to receive either sildenafil 10 mg/kg/d or placebo by subcutaneous injection from GD24 to GD30. On GD28, fetuses were randomly assigned to TO or sham neck dissection. At term (GD30) fetuses were delivered, ventilated, and finally harvested for histological and molecular analyses. Unoperated littermates served as controls. RESULTS: The lung-to-body-weight ratio was significantly reduced in sham-CDH fetuses either (1.2 ±â€Š0.3% vs 2.3 ±â€Š0.3% in controls, P=0.0003). Sildenafil had no effect on this parameter, while CDH fetuses undergoing TO had a lung-to-body-weight ratio comparable to that of controls (2.5 ±â€Š0.8%, P<0.0001). Sildenafil alone induced an improvement in the mean terminal bronchiolar density (2.5 ±â€Š0.8 br/mm2 vs 3.5 ±â€Š0.9 br/mm2, P=0.043) and lung mechanics (static elastance 61 ±â€Š36 cmH2O /mL vs 113 ±â€Š40 cmH2O/mL, P=0.008), but both effects were more pronounced in fetuses undergoing additional TO (2.1 ±â€Š0.8 br/mm2, P=0.001 and 31 ±â€Š9 cmH2O/mL, P<0.0001 respectively). Both CDH-sham and CDH-TO fetuses treated with placebo had an increased medial wall thickness of peripheral pulmonary vessels (41.9 ±â€Š2.9% and 41.8 ±â€Š3.2%, vs 24.0 ±â€Š2.9% in controls, P<0.0001). CDH fetuses treated with sildenafil, either with or without TO, had a medial thickness in the normal range (29.4% ±â€Š2.6%). Finally, TO reduced gene expression of vascular endothelial growth factor and surfactant protein A and B, but this effect was counteracted by sildenafil. CONCLUSION: In the rabbit model for CDH, the combination of maternal sildenafil and TO has a complementary effect on vascular and parenchymal lung development.


Asunto(s)
Hernias Diafragmáticas Congénitas , Pulmón/crecimiento & desarrollo , Citrato de Sildenafil/administración & dosificación , Tráquea/cirugía , Animales , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Feto , Embarazo , Conejos , Distribución Aleatoria
4.
Pediatr Res ; 89(4): 803-813, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32434214

RESUMEN

BACKGROUND: Bronchopulmonary dysplasia (BPD), the most common complication of prematurity, arises from various factors that compromise lung development, including oxygen and inflammation. Hyperoxia has been used to mimic the disease in newborn rodents. The use of a second hit to induce systemic inflammation has been suggested as an added strategy to better mimic the inflammatory aspect of BPD. Here we report a novel 2 hit (2HIT) BPD model with in-depth characterization of the innate immune response, enabling mechanistic studies of therapies with an immunomodulatory component. METHODS: C57BL/6N mice were exposed to 85% O2 from postnatal day (P)1 to P7, and received postnatally (P3) Escherichia coli LPS. At various timepoints, immune activation in the lung and at the systemic level was analyzed by fluorescence-activated cell sorting (FACS), and gene and protein expressions. RESULTS: 2HIT mice showed fewer alveoli, increased lung compliance, and right ventricular hypertrophy. A transient proinflammatory cytokine response was observed locally and systemically. Type 2 anti-inflammatory cytokine expression was decreased in the lung together with the number of mature alveolar macrophages. Simultaneously, a Siglec-F intermediate macrophage population emerged. CONCLUSION: This study provides long-term analysis of the 2HIT model, suggesting impairment of type 2 cytokine environment and altered alveolar macrophage profile in the lung. IMPACT: We have developed a novel 2HIT mouse BPD model with postnatal LPS and hyperoxia exposure, which enables mechanistic studies of potential therapeutic strategies with an immunomodulatory component. This is the first report of in-depth characterization of the lung injury and recovery describing the evolution of the innate immune response in a standardized mouse model for experimental BPD with postnatal LPS and hyperoxia exposure. The 2HIT model has the potential to help understand the link between inflammation and impaired lung development, and will enable testing of new therapies in a short and more robust manner.


Asunto(s)
Displasia Broncopulmonar/inmunología , Inmunidad Innata , Inflamación/inmunología , Oxígeno/metabolismo , Animales , Peso Corporal , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Hiperoxia , Lipopolisacáridos/metabolismo , Pulmón/efectos de los fármacos , Pulmón/fisiología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
Pediatr Res ; 87(7): 1193-1200, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31816623

RESUMEN

BACKGROUND: Bronchopulmonary dysplasia (BPD) remains a frequent complication following preterm birth, affecting respiratory health throughout life. Transcriptome analysis in a preterm rabbit model for BPD revealed dysregulation of key genes for inflammation, vascular growth and lung development in animals exposed to hyperoxia, which could be prevented by simvastatin. METHODS: Preterm rabbits were randomized to either normoxia (21% O2) or hyperoxia (95% O2) and within each condition to treatment with 5 mg/kg simvastatin daily or control. Lung function, structure and mRNA-expression was assessed on day 7. RESULTS: Simvastatin partially prevented the effect of hyperoxia on lung function, without altering alveolar structure or inflammation. A trend towards a less fibrotic phenotype was noted in simvastatin-treated pups, and airways were less muscularized. Most importantly, simvastatin completely prevented hyperoxia-induced arterial remodeling, in association with partial restoration of VEGFA and VEGF receptor 2 (VEGFR2) expression. Simvastatin however decreased survival in pups exposed to normoxia, but not to hyperoxia. CONCLUSION: Repurposing of simvastatin could be an advantageous therapeutic strategy for bronchopulmonary dysplasia and other developmental lung diseases with pulmonary vascular disease. The increased mortality in the treated normoxia group however limits the translational value at this dose and administration route.


Asunto(s)
Displasia Broncopulmonar/prevención & control , Hiperoxia/prevención & control , Simvastatina/uso terapéutico , Animales , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/fisiopatología , Femenino , Perfilación de la Expresión Génica , Hiperoxia/patología , Hiperoxia/fisiopatología , Embarazo , Nacimiento Prematuro , Conejos , Distribución Aleatoria , Pruebas de Función Respiratoria , Análisis de Supervivencia
6.
Prenat Diagn ; 37(11): 1093-1099, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28842991

RESUMEN

The amniotic fluid is a clinically accessible source of mesenchymal stem cells (AF-MSC) during gestation, which enables autologous cellular therapy for perinatal disorders. The origin of AF-MSC remains elusive: renal and neuronal progenitors have been isolated from the AF-MSC pool, yet no cells with pulmonary characteristics. We analyzed gene expression of pulmonary and renal markers of 212 clonal lines of AF-MSC isolated from amniocentesis samples. AF-MSC were cultured on dishes coated with extracellular matrix (ECM) proteins from decellularized fetal rabbit lungs. In vivo differentiation potential of AF-MSC that expressed markers suggestive of lung fate was tested by renal subcapsular injections in immunodeficient mice. Of all the isolated AF-MSC lines, 26% were positive for lung endodermal markers FOXA2 and NKX2.1 and lacked expression of renal markers (KSP). This AF-MSC subpopulation expressed other lung-specific factors, including IRX1, P63, FOXP2, LGR6, SFTC, and PDPN. Pulmonary marker expression decreased over passages when AF-MSC were cultured under conventional conditions, yet remained more stable when culturing the cells on lung ECM-coated dishes. Renal subcapsular injection of AF-MSC expressing lung-specific markers resulted in engrafted cells that were SPTB positive. These data suggest that FOXA2+/NKX2.1+/KSP- AF-MSC lines have lung characteristics which are supported by culture on lung ECM-coated dishes.


Asunto(s)
Líquido Amniótico/citología , Pulmón/citología , Células Madre Mesenquimatosas/citología , Técnicas de Cultivo , Femenino , Humanos , Embarazo
8.
J Transl Med ; 14(1): 247, 2016 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-27567616

RESUMEN

BACKGROUND: The administration of supplemental oxygen to treat ventilatory insufficiency may lead to the formation of reactive oxygen species and subsequent tissue damage. Cytochrome P4501A1 (CYP1A1) can modulate hyperoxic lung injury by a currently unknown mechanism. Our objective was to evaluate the effect of administration of omeprazole on the induction of CYP1A1 and its influence on hyperoxic lung injury in an established preterm rabbit model. METHODS: Omeprazole was administered either (1) directly to the fetus, (2) to the mother or (3) after birth to the pups in different doses (2-10 or 20 mg/kg). Controls were injected with the same amount of saline. Pups were housed in normoxia (21 %) or hyperoxia (>95 %) for 5 days. Outcome parameters were induction of CYP1A1 measured by real-time polymerase chain reaction (RT-PCR) immediately after delivery, at day 3 and day 5 as well as lung function, morphometry and immunohistochemistry assessed at day 5 of life. Transcriptome analysis was used to define the targeted pathways. RESULTS: Daily neonatal injections demonstrated a dose-dependent increase in CYP1A1. Lung function tests showed a significant improvement in tissue damping, tissue elasticity, total lung capacity, static compliance and elastance. Morphometry revealed a more developed lung architecture with thinned septae in animals treated with the highest dose (20 mg/kg) of omeprazole. Surfactant protein B, vascular endothelial growth factor and its receptor were significantly increased on immunohistochemical stainings after omeprazole treatment. CONCLUSIONS: Neonatal administration of omeprazole induces CYP1A1 in a dose-dependent matter and combined pre- and postnatal administration attenuates hyperoxic lung injury in preterm rabbits, even with the lowest dose of omeprazole without clear CYP1A1 induction.


Asunto(s)
Hiperoxia/complicaciones , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/etiología , Omeprazol/uso terapéutico , Inhibidores de la Bomba de Protones/uso terapéutico , Animales , Animales Recién Nacidos , Citocromo P-450 CYP1A1/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Perfilación de la Expresión Génica , Inmunohistoquímica , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/crecimiento & desarrollo , Pulmón/patología , Lesión Pulmonar/genética , Lesión Pulmonar/fisiopatología , Omeprazol/farmacología , Inhibidores de la Bomba de Protones/farmacología , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Pruebas de Función Respiratoria , Análisis de Supervivencia , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
9.
J Urol ; 196(1): 261-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26880411

RESUMEN

PURPOSE: Serious complications can develop with the mesh implants used for stress urinary incontinence and pelvic organ prolapse surgery. We evaluated 2 materials currently in clinical use and 2 alternative materials using a rabbit abdominal model to assess host response and biomechanical properties of the materials before and after implantation. MATERIALS AND METHODS: Poly-L-lactic acid and polyurethane meshes were electrospun to be compared to commercially available polypropylene and polyvinylidene fluoride meshes. A total of 40 immunocompetent full-thickness abdominal wall defect rabbit models were used, including 8 in each of the poly-L-lactic acid, polyurethane, polyvinylidene fluoride and polypropylene experimental groups, and sham controls. Two 20 mm defects were created per animal and primarily repaired. The experimental groups then underwent onlay of each repair material while sham controls did not. Four rabbits per group were sacrificed at days 30 and 90. Abdominal wall specimens containing the defect with or without repair material were explanted to be assessed by histology (hematoxylin and eosin staining, and immunohistochemistry) and biomechanical testing at 30 and 90 days. RESULTS: At 90 days of implantation tissues repaired with all 4 materials showed biomechanical properties without significant differences. However, polypropylene and polyvinylidene fluoride meshes demonstrated a sustained chronic inflammatory response profile by 90 days. In contrast, poly-L-lactic acid and polyurethane meshes integrated well into host tissues with a decreased inflammatory response, indicative of constructive remodeling. CONCLUSIONS: Poly-L-lactic acid and polyurethane alternative materials achieved better host integration in rabbit models than current synthetic repair materials.


Asunto(s)
Prolapso de Órgano Pélvico/cirugía , Implantación de Prótesis/instrumentación , Mallas Quirúrgicas , Incontinencia Urinaria de Esfuerzo/cirugía , Animales , Fenómenos Biomecánicos , Masculino , Ensayo de Materiales , Microscopía Electrónica de Rastreo , Poliésteres , Polipropilenos , Poliuretanos , Polivinilos , Conejos
10.
Prenat Diagn ; 35(5): 456-62, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25641322

RESUMEN

OBJECTIVES: Stem cells (SCs) can be isolated from amniotic fluid (AF) for a variety of perinatal applications. In view of this, we compared different cryopreservation protocols for these AFSCs. METHODS: We screened seven freezing and thawing protocols using two well-established human AFSC lines: freezing protocol 1 (FP1), 10% dimethyl sulfoxide (DMSO); FP2, 2.5% DMSO, caspase inhibitor, and catalase; FP3, 5% glycerol, caspase inhibitor, and catalase; FP4, sperm freezing medium; FP5, slow-freezing solution; FP6, ethylene glycol, sucrose, and Ficoll 70; and FP7, vitrification solution. Outcome measures were post-thawing cell viability, recovery, doubling time and mesenchymal SC markers. The three best performing protocols were subsequently tested on cells isolated from clinical consecutive freshly harvested AF samples from two fetal medicine units. RESULTS: Protocols 1, 5, and 6 performed significantly better on well-characterized cell lines. They performed equally well on cell pellets from freshly harvested AF (n = 28). CONCLUSIONS: We identified three suitable cryopreservation protocols because of high cell recovery and unchanged SC characteristics. Given one of these, the slow-freezing solution, is compatible with current good manufacturing practice legislation, it may be ultimately clinically used.


Asunto(s)
Líquido Amniótico/citología , Criopreservación/métodos , Células Madre/citología , Antígenos CD/metabolismo , Inhibidores de Caspasas , Catalasa , Línea Celular , Proliferación Celular , Supervivencia Celular , Dimetilsulfóxido , Glicol de Etileno , Femenino , Ficoll , Glicerol , Humanos , Embarazo , Soluciones , Células Madre/metabolismo , Células Madre/fisiología , Sacarosa , Vitrificación
11.
Cells ; 11(7)2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35406742

RESUMEN

Extreme preterm birth disrupts late lung development and puts newborns at risk of developing chronic lung disease, known as bronchopulmonary dysplasia (BPD). BPD can be associated with life-long complications, and currently no effective treatment is available. Cell therapies are entering the clinics to curb complications of extreme preterm birth with several clinical trials testing the feasibility, safety and efficacy of mesenchymal stromal cells (MSCs). The therapeutic effect of MSCs is contained in their secretome, and nanosized membranous structures released by the MSCs, known as extracellular vesicles (EVs), have been shown to be the therapeutic vectors. Driven by this discovery, the efficacy of EV-based therapy is currently being explored in models of BPD. EVs derived from MSCs, contain a rich cargo of anti-inflammatory and pro-angiogenic molecules, making them suitable candidates to treat multifactorial diseases such as BPD. Here, we review the state-of-the-art of preclinical studies involving MSC-derived EVs in models of BPD and highlight technical and regulatory challenges that need to be addressed before clinical translation. In addition, we aim at increasing awareness regarding the importance of rigorous reporting of experimental details of EV experiments and to increase the outreach of the current established guidelines amongst researchers in the BPD field.


Asunto(s)
Displasia Broncopulmonar , Vesículas Extracelulares , Enfermedades del Recién Nacido , Células Madre Mesenquimatosas , Nacimiento Prematuro , Displasia Broncopulmonar/terapia , Humanos , Recién Nacido , Pulmón
12.
Nat Commun ; 12(1): 1565, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692365

RESUMEN

During late lung development, alveolar and microvascular development is finalized to enable sufficient gas exchange. Impaired late lung development manifests as bronchopulmonary dysplasia (BPD) in preterm infants. Single-cell RNA sequencing (scRNA-seq) allows for assessment of complex cellular dynamics during biological processes, such as development. Here, we use MULTI-seq to generate scRNA-seq profiles of over 66,000 cells from 36 mice during normal or impaired lung development secondary to hyperoxia with validation of some of the findings in lungs from BPD patients. We observe dynamic populations of cells, including several rare cell types and putative progenitors. Hyperoxia exposure, which mimics the BPD phenotype, alters the composition of all cellular compartments, particularly alveolar epithelium, stromal fibroblasts, capillary endothelium and macrophage populations. Pathway analysis and predicted dynamic cellular crosstalk suggest inflammatory signaling as the main driver of hyperoxia-induced changes. Our data provides a single-cell view of cellular changes associated with late lung development in health and disease.


Asunto(s)
Hiperoxia/genética , Hiperoxia/fisiopatología , Pulmón/metabolismo , Pulmón/patología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/patología , Genotipo , Masculino , Ratones
13.
Semin Perinatol ; 42(7): 453-458, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30376986

RESUMEN

Improved survival of extreme preterm infants has made the task of protecting the ever more immature lung from injury more challenging. As a consequence, the incidence of bronchopulmonary dysplasia (BPD), the chronic lung disease of prematurity, has remained unchanged. The multifactorial disease pathogenesis of BPD - including amongst others inflammation, oxidative stress and excessive lung stretch - adds further complexity to finding effective therapies that would prevent lung injury and promote lung growth. Mesenchymal stromal cells and the discovery of their pleiotropic effects represent an appealing approach for the prevention of BPD. Mesenchymal stromal cells do not engraft but exert their therapeutic benefit through paracrine effects. These paracrine effects seem to be mediated through the release of nanosized extra-cellular vesicles used for cell-cell communication. This review will summarize our current knowledge on these potential nanotherapies for micropreemies.


Asunto(s)
Displasia Broncopulmonar/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inflamación/terapia , Pulmón/crecimiento & desarrollo , Trasplante de Células Madre Mesenquimatosas , Nanomedicina/tendencias , Cordón Umbilical/citología , Displasia Broncopulmonar/fisiopatología , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Humanos , Recién Nacido , Recien Nacido Prematuro , Recién Nacido de muy Bajo Peso , Inflamación/fisiopatología , Pulmón/fisiopatología , Neonatología
14.
J Matern Fetal Neonatal Med ; 31(7): 958-966, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28277906

RESUMEN

Bronchopulmonary dysplasia (BPD) remains the most common complication of extreme prematurity as no effective treatment is available to date. This calls for the exploration of new therapeutic options like cell therapy, which is already effective for various human (lung) disorders. We systematically searched the MEDLINE, Embase, and Web of Science databases from the earliest date till January 2017 and included original studies on the perinatal use of cell-based therapies (i.e. cells and/or cell-derivatives) to treat BDP in animal models. Fourth publications describing 47 interventions were retrieved. Newborn mice/rats raised in a hyperoxic environment were studied in most interventions. Different cell types - either intact cells or their conditioned medium - were administered, but bone marrow and umbilical cord blood derived mesenchymal stem cells were most prevalent. All studies reported positive effects on outcome parameters including alveolar and vascular morphometry, lung function, and inflammation. Cell homing to the lungs was demonstrated in some studies, but the therapeutic effects seemed to be mostly mediated via paracrine modulation of inflammation, fibrosis and angiogenesis. CONCLUSION: Multiple rat/mouse studies show promise for cell therapy for BPD. Yet careful study of action mechanisms and side effects in large animal models is imperative before clinical translation can be achieved.


Asunto(s)
Displasia Broncopulmonar/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Displasia Broncopulmonar/prevención & control , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Humanos , Recien Nacido Extremadamente Prematuro , Recién Nacido , Ratones , Ratas , Resultado del Tratamiento
15.
Eur J Pediatr Surg ; 28(3): 285-292, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28564708

RESUMEN

PURPOSE: The reconstruction of congenital diaphragmatic hernia or other congenital soft tissue defects often requires implants. These can be either degradable or permanent, each having their advantages. Whatever type is being used, the host response induced by implants plays a crucial role to determine the outcome. Macrophages are pivotal during implant remodeling; they are plastic and acquire in response to environmental stimuli either an inflammatory status and mediate subsequent fibrosis or a regulatory status and facilitate functional remodeling. Matrices engineered with mesenchymal stem cells (MSCs) have the capacity to modulate the host immune reaction. MSCs are believed to promote constructive remodeling of the implant through a regulatory macrophage response among others. Herein, we evaluate this potential of MSC derived from the amniotic fluid (AF-MSC), an interesting MSC type for neonatal reconstruction, on electrospun polylactic acid (PLA) scaffolds. METHODS: We seeded AF-MSC at a density of 1.105/cm2 on electrospun PLA matrices and determined cell viability. In vivo, we used cell-seeded or cell-free PLA matrices for subcutaneous implantation in immune competent rats. The host immune response was evaluated by histomorphometry at 14 days postoperatively. RESULTS: The PLA matrix supported adherence and proliferation of AF-MSC. Fourteen days after implantation, PLA matrices were well penetrated by inflammatory cells, new blood vessels, and collagen fibers. AF-MSC-seeded scaffolds were associated with a similar response yet with a decreased number of eosinophils, increased matrix degradation and collagen fiber deposition compared with controls. The amount of total macrophages and of M2-subtype was similar for all animals. CONCLUSION: Electrospun PLA matrices are a suitable substrate for short-term culture of AF-MSC. In rats, addition of AF-MSC to PLA matrices modulates the host response after subcutaneous implantation, yet without a difference in macrophage profile compared with control.


Asunto(s)
Líquido Amniótico/citología , Regeneración Tisular Dirigida/métodos , Hernias Diafragmáticas Congénitas/cirugía , Herniorrafia/métodos , Trasplante de Células Madre Mesenquimatosas , Poliésteres/uso terapéutico , Andamios del Tejido , Animales , Hernias Diafragmáticas Congénitas/inmunología , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar
16.
Neonatology ; 113(3): 275-285, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29393249

RESUMEN

BACKGROUND: Bronchopulmonary dysplasia (BPD) is a chronic lung disease that affects extremely preterm infants and remains - despite improvements in neonatal intensive care - a major cause of neonatal mortality and morbidity. Cell-therapeutic strategies employing mesenchymal stem cells (MSC) have been shown to modulate lung development in BPD models. OBJECTIVE: Herein, we evaluate the potential of human amniotic fluid (hAF)-SC and hAF-SC with upregulated expression of vascular endothelial growth factor (VEGF) as cell-therapeutic agents for BPD. METHODS: Preterm rabbit pups were raised in normoxia (21% O2) or hyperoxia (≥95% O2). Hyperoxia-exposed pups randomly received an intraperitoneal injection of fibroblasts, naïve hAF-SC, or hAF-SC-VEGF on postnatal day (PN) 0. On PN7, surviving pups were tested for pulmonary (forced oscillation technique) and vascular (pulmonary artery Doppler ultrasound) function, and lungs were processed for morphometric measurements of parenchymal and vascular structure and inflammation. RESULTS: Intraperitoneal injection of cells resulted in homing to the lungs. The lungs of hyperoxia-exposed animals displayed parenchymal and vascular structural and functional damage reminiscent of BPD, which was significantly improved after treatment with hAF-SC-VEGF. Treating hyperoxia-exposed animals with naïve AF-SC attenuated only the lung inflammation and the vascular structural defect. Treatment with fibroblasts, which were used as a cellular control, did not lead to any improvements. CONCLUSION: hAF-SC with upregulated VEGF expression display enhanced potential to prevent/reverse lung injury in preterm rabbits, whereas naïve hAF-SC only show a moderate therapeutic potential. These results point towards an added value of VEGF delivered by hAF-SC in the treatment of BPD.


Asunto(s)
Displasia Broncopulmonar/terapia , Hiperoxia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Líquido Amniótico/citología , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/etiología , Hipertensión Pulmonar/complicaciones , Pulmón/patología , Células Madre Mesenquimatosas/metabolismo , Alveolos Pulmonares/fisiología , Conejos , Distribución Aleatoria , Regulación hacia Arriba
17.
Semin Pediatr Surg ; 25(3): 186-90, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27301606

RESUMEN

Regenerative medicine offers hope of a sustainable solution for severe airway disease by the creation of functional, immunocompatible organ replacements. When considering fetuses and newborns, there is a specific spectrum of airway pathologies that could benefit from cell therapy and tissue engineering applications. While hypoplastic lungs associated with congenital diaphragmatic hernia (CDH) could benefit from cellular based treatments aimed at ameliorating lung function, patients with upper airway obstruction could take advantage from a de novo tissue engineering approach. Moreover, the international acceptance of the EXIT procedure as a means of securing the precarious neonatal airway, together with the advent of fetal surgery as a method of heading off postnatal co-morbidities, offers the revolutionary possibility of extending the clinical indication for tissue-engineered airway transplantation to infants affected by diverse severe congenital laryngotracheal malformations. This article outlines the necessary basic components for regenerative medicine solutions in this potential clinical niche.


Asunto(s)
Enfermedades de la Laringe/congénito , Enfermedades de la Laringe/cirugía , Anomalías del Sistema Respiratorio/cirugía , Ingeniería de Tejidos/métodos , Enfermedades de la Tráquea/congénito , Enfermedades de la Tráquea/cirugía , Terapias Fetales/métodos , Regeneración Tisular Dirigida , Humanos , Recién Nacido , Laringe/anomalías , Laringe/cirugía , Trasplante de Tejidos , Tráquea/anomalías , Tráquea/cirugía
18.
Eur J Pediatr Surg ; 24(3): 270-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24937193

RESUMEN

In the EU-27, 2,100 babies with congenital diaphragmatic hernia (CDH) are born annually. CDH is fatal in 30% of them. Experimental fetal surgery in severe cases results in a survival rate of 50 to 60% at its best. Failure is due to insufficient lung growth, persistent pulmonary hypertension or prematurity induced by the procedure. For nonsurvivors alternative strategies are required. Survivors undergo anatomical repair, but large diaphragmatic defects are closed using a patch. At present the used materials are less than ideal, mainly because of recurrence and chest deformation. To overcome the above limitations, alternative medical therapies (pharmacologic or cell therapy) that are more potent and less invasive are needed. Also a more functional postnatal repair may be possible when using novel scaffolds or engineered constructs. We see a prominent place for autologous amniotic fluid-derived stem cells for these novel strategies, which could be prenatally harvested following appropriate patient selection by noninvasive imaging.


Asunto(s)
Hernias Diafragmáticas Congénitas/terapia , Trasplante de Células Madre , Ingeniería de Tejidos/métodos , Líquido Amniótico/citología , Humanos , Pulmón/anomalías , Pulmón/cirugía , Andamios del Tejido , Trasplante Autólogo
19.
Neuron ; 77(1): 70-82, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23312517

RESUMEN

GABAergic interneurons mainly originate in the medial ganglionic eminence (MGE) of the embryonic ventral telencephalon (VT) and migrate tangentially to the cortex, guided by membrane-bound and secreted factors. We found that Sip1 (Zfhx1b, Zeb2), a transcription factor enriched in migrating cortical interneurons, is required for their proper differentiation and correct guidance. The majority of Sip1 knockout interneurons fail to migrate to the neocortex and stall in the VT. RNA sequencing reveals that Sip1 knockout interneurons do not acquire a fully mature cortical interneuron identity and contain increased levels of the repulsive receptor Unc5b. Focal electroporation of Unc5b-encoding vectors in the MGE of wild-type brain slices disturbs migration to the neocortex, whereas reducing Unc5b levels in Sip1 knockout slices and brains rescues the migration defect. Our results reveal that Sip1, through tuning of Unc5b levels, is essential for cortical interneuron guidance.


Asunto(s)
Movimiento Celular/fisiología , Corteza Cerebral/crecimiento & desarrollo , Interneuronas/fisiología , Neocórtex/crecimiento & desarrollo , Proteínas del Tejido Nervioso/deficiencia , Receptores de Superficie Celular/deficiencia , Animales , Corteza Cerebral/citología , Técnicas de Inactivación de Genes , Ratones , Ratones Transgénicos , Neocórtex/citología , Proteínas del Tejido Nervioso/genética , Receptores de Netrina , Técnicas de Cultivo de Órganos , Receptores de Superficie Celular/genética , Telencéfalo/citología , Telencéfalo/crecimiento & desarrollo
20.
Cytokine Growth Factor Rev ; 22(5-6): 287-300, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22119658

RESUMEN

Signaling by the many ligands of the TGFß family strongly converges towards only five receptor-activated, intracellular Smad proteins, which fall into two classes i.e. Smad2/3 and Smad1/5/8, respectively. These Smads bind to a surprisingly high number of Smad-interacting proteins (SIPs), many of which are transcription factors (TFs) that co-operate in Smad-controlled target gene transcription in a cell type and context specific manner. A combination of functional analyses in vivo as well as in cell cultures and biochemical studies has revealed the enormous versatility of the Smad proteins. Smads and their SIPs regulate diverse molecular and cellular processes and are also directly relevant to development and disease. In this survey, we selected appropriate examples on the BMP-Smads, with emphasis on Smad1 and Smad5, and on a number of SIPs, i.e. the CPSF subunit Smicl, Ttrap (Tdp2) and Sip1 (Zeb2, Zfhx1b) from our own research carried out in three different vertebrate models.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Humanos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA