Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Indian J Med Res ; 159(1): 71-77, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38293841

RESUMEN

BACKGROUND OBJECTIVES: This study aimed to compare the admission characteristics and outcomes of tribal and non-tribal neonates admitted to a level II special newborn care unit (SNCU) in rural Gujarat. METHODS: This was a retrospective observational study that looked at all neonates admitted to a high-volume SNCU between 2013 and 2021. A series of quality improvement measures were introduced over the study period. Admission characteristics, such as birth weight, gestational age, gender and outcomes for tribal and non-tribal neonates, were compared. RESULTS: Six thousand nine hundred and ninety neonates [4829 tribal (69.1%) and 2161 (30.9%) non-tribal] were admitted to the SNCU. Tribal neonates had lower mean birth weight (2047 vs . 2311 g, P <0.01) and gestational week at birth (35.8 vs . 36.7 weeks, P <0.01) compared to non-tribal neonates. Common causes of admissions were neonatal jaundice (1990, 28.4%), low birth weight (1308, 18.7%) and neonatal sepsis (843, 12%). Six hundred and thirty-eight (9.1%) neonates died during the treatment in the SNCU. The odds of death among tribal neonates was similar to non-tribal neonates [adjusted odds ratio: 1.12 (95% confidence interval [CI]: 0.89, 1.42)]. The tribal neonates had significantly higher cause-specific case fatality rate from sepsis [relative risk (RR): 2.18 (95% CI: 1.41, 3.37)], prematurity [RR: 1.98 (95% CI: 1.23, 3.17)] and low birth weight [RR: 1.83 (95% CI: 1.17, 2.85)]. The overall case fatality rate in the SNCU decreased from 18.2 per cent during the year 2013-2014 to 2.1 per cent in the year 2020-2021. INTERPRETATION CONCLUSIONS: There was a reduction in the case fatality rate over the study period. Tribal and non-tribal neonates had similar risk of death. Sepsis prevention and management, mechanical respiratory support and timely referral to a higher centre might help further reduction in mortality for these neonates.


Asunto(s)
Hospitalización , Sepsis , Recién Nacido , Humanos , Peso al Nacer , Recien Nacido Prematuro , Recién Nacido de Bajo Peso , Sepsis/epidemiología
2.
J Cell Biol ; 222(12)2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37843535

RESUMEN

With each breath, oxygen diffuses across remarkably thin alveolar type I (AT1) cells into underlying capillaries. Interspersed cuboidal AT2 cells produce surfactant and act as stem cells. Even transient disruption of this delicate barrier can promote capillary leak. Here, we selectively ablated AT1 cells, which uncovered rapid AT2 cell flattening with near-continuous barrier preservation, culminating in AT1 differentiation. Proliferation subsequently restored depleted AT2 cells in two phases, mitosis of binucleated AT2 cells followed by replication of mononucleated AT2 cells. M phase entry of binucleated and S phase entry of mononucleated cells were both triggered by AT1-produced hbEGF signaling via EGFR to Wnt-active AT2 cells. Repeated AT1 cell killing elicited exuberant AT2 proliferation, generating aberrant daughter cells that ceased surfactant function yet failed to achieve AT1 differentiation. This hyperplasia eventually resolved, yielding normal-appearing alveoli. Overall, this specialized regenerative program confers a delicate simple epithelium with functional resiliency on par with the physical durability of thicker, pseudostratified, or stratified epithelia.


Asunto(s)
Pulmón , Células Madre , Tensoactivos , Diferenciación Celular , División Celular , Células Cultivadas , Células Madre/citología
3.
Exp Mol Med ; 55(8): 1831-1842, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37582976

RESUMEN

We present an in-depth single-cell atlas of in vitro multiculture systems on human primary airway epithelium derived from normal and diseased lungs of 27 individual donors. Our large-scale single-cell profiling identified new cell states and differentiation trajectories of rare airway epithelial cell types in human distal lungs. By integrating single-cell datasets of human lung tissues, we discovered immune-primed subsets enriched in lungs and organoids derived from patients with chronic respiratory disease. To demonstrate the full potential of our platform, we further illustrate transcriptomic responses to various respiratory virus infections in vitro airway models. Our work constitutes a single-cell roadmap for the cellular and molecular characteristics of human primary lung cells in vitro and their relevance to human tissues in vivo.


Asunto(s)
Células Epiteliales , Pulmón , Humanos , Células Epiteliales/metabolismo , Epitelio , Diferenciación Celular/fisiología , Organoides
4.
Nature ; 619(7971): 860-867, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37468622

RESUMEN

Many cancers originate from stem or progenitor cells hijacked by somatic mutations that drive replication, exemplified by adenomatous transformation of pulmonary alveolar epithelial type II (AT2) cells1. Here we demonstrate a different scenario: expression of KRAS(G12D) in differentiated AT1 cells reprograms them slowly and asynchronously back into AT2 stem cells that go on to generate indolent tumours. Like human lepidic adenocarcinoma, the tumour cells slowly spread along alveolar walls in a non-destructive manner and have low ERK activity. We find that AT1 and AT2 cells act as distinct cells of origin and manifest divergent responses to concomitant WNT activation and KRAS(G12D) induction, which accelerates AT2-derived but inhibits AT1-derived adenoma proliferation. Augmentation of ERK activity in KRAS(G12D)-induced AT1 cells increases transformation efficiency, proliferation and progression from lepidic to mixed tumour histology. Overall, we have identified a new cell of origin for lung adenocarcinoma, the AT1 cell, which recapitulates features of human lepidic cancer. In so doing, we also uncover a capacity for oncogenic KRAS to reprogram a differentiated and quiescent cell back into its parent stem cell en route to adenomatous transformation. Our work further reveals that irrespective of a given cancer's current molecular profile and driver oncogene, the cell of origin exerts a pervasive and perduring influence on its subsequent behaviour.


Asunto(s)
Adenocarcinoma del Pulmón , Reprogramación Celular , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas p21(ras) , Células Madre , Humanos , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Reprogramación Celular/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células Madre/metabolismo , Células Madre/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo
5.
Environ Toxicol ; 38(1): 39-48, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36124540

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder associated with striatal dopaminergic neuronal loss in the Substantia nigra. Oxidative stress plays a significant role in several neurodegenerative diseases. Paraquat (PQ) is considered a potential neurotoxin that affects the brain leading to the death of dopaminergic neurons mimicking the PD phenotype. Various scientific reports have proven that cryptotanshinone possesses antioxidant and anti-inflammatory properties. We hypothesized that cryptotanshinone could extend its neuroprotective activity by exerting antioxidant effects. This study was designed to evaluate the effects of cryptotanshinone in both cellular and animal models of PQ-induced PD. Annexin V-PI double staining and immunoblotting were used to detect apoptosis and oxidative stress proteins, respectively. Reactive oxygen species kits were used to evaluate oxidative stress in cells. For in vivo studies, 18 B6 mice were divided into three groups. The rotarod data revealed the motor function and immunostaining showed the survival of TH+ neurons in SNpc region. Our study showed that cryptotanshinone attenuated paraquat-induced oxidative stress by upregulating anti-oxidant markers in vitro, and restored behavioral deficits and survival of dopaminergic neurons in vivo, demonstrating its therapeutic potential.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Animales , Ratones , Paraquat/toxicidad , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Neuronas Dopaminérgicas/metabolismo , Estrés Oxidativo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Modelos Animales de Enfermedad
6.
Antioxidants (Basel) ; 11(12)2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36552630

RESUMEN

Supplementing with vitamin B3 has been reported to protect against retinal ganglion cell (RGC) damage events and exhibit multiple neuroprotective properties in a mouse model of optic nerve injury. In this study, a rat model of anterior ischemic optic neuropathy was used to assess the neuroprotective benefits of vitamin B3 (rAION). Vitamin B3 (500 mg/kg/day) or phosphate-buffered saline (PBS) was administered to the rAION-induced rats every day for 28 days. The vitamin B3-treated group had significantly higher first positive and second negative peak (P1-N2) amplitudes of flash visual-evoked potentials and RGC densities than the PBS-treated group (p < 0.05). A terminal deoxynucleotidyl transferase dUTP nick end labeling assay conducted on vitamin B3-treated rats revealed a significant reduction in apoptotic cells (p < 0.05). Superoxide dismutase and thiobarbituric acid reactive substance activity showed that vitamin B3 treatment decreased reactive oxygen species (p < 0.05). Therefore, vitamin B3 supplementation preserves vision in rAION-induced rats by reducing oxidative stress, neuroinflammation, and mitochondrial apoptosis.

7.
Nat Commun ; 13(1): 7137, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-36414616

RESUMEN

The lung's gas exchange surface is comprised of alveolar AT1 and AT2 cells that are corrupted in several common and deadly diseases. They arise from a bipotent progenitor whose differentiation is thought to be dictated by differential mechanical forces. Here we show the critical determinant is FGF signaling. Fgfr2 is expressed in the developing progenitors in mouse then restricts to nascent AT2 cells and remains on throughout life. Its ligands are expressed in surrounding mesenchyme and can, in the absence of exogenous mechanical cues, induce progenitors to form alveolospheres with intermingled AT2 and AT1 cells. FGF signaling directly and cell autonomously specifies AT2 fate; progenitors lacking Fgfr2 in vitro and in vivo exclusively acquire AT1 fate. Fgfr2 loss in AT2 cells perinatally results in reprogramming to AT1 identity, whereas loss or inhibition later in life triggers AT2 apoptosis and compensatory regeneration. We propose that Fgfr2 signaling selects AT2 fate during development, induces a cell non-autonomous AT1 differentiation signal, then continuously maintains AT2 identity and survival throughout life.


Asunto(s)
Células Epiteliales Alveolares , Mesodermo , Animales , Ratones , Diferenciación Celular , Transducción de Señal , Apoptosis
8.
Am J Respir Cell Mol Biol ; 67(3): 284-308, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35679511

RESUMEN

Clinical and molecular heterogeneity are common features of human disease. Understanding the basis for heterogeneity has led to major advances in therapy for many cancers and pulmonary diseases such as cystic fibrosis and asthma. Although heterogeneity of risk factors, disease severity, and outcomes in survivors are common features of the acute respiratory distress syndrome (ARDS), many challenges exist in understanding the clinical and molecular basis for disease heterogeneity and using heterogeneity to tailor therapy for individual patients. This report summarizes the proceedings of the 2021 Aspen Lung Conference, which was organized to review key issues related to understanding clinical and molecular heterogeneity in ARDS. The goals were to review new information about ARDS phenotypes, to explore multicellular and multisystem mechanisms responsible for heterogeneity, and to review how best to account for clinical and molecular heterogeneity in clinical trial design and assessment of outcomes. The report concludes with recommendations for future research to understand the clinical and basic mechanisms underlying heterogeneity in ARDS to advance the development of new treatments for this life-threatening critical illness.


Asunto(s)
Síndrome de Dificultad Respiratoria , Humanos , Pulmón , Factores de Riesgo , Índice de Severidad de la Enfermedad , Tórax
9.
Bioeng Transl Med ; 7(2): e10289, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35600664

RESUMEN

An ischemic insult at optic nerve (ON) is followed by detrimental neuroinflammation that results in progressive and long-lasting retinal ganglion cell (RGC) death and vision loss. Icariin was reported to be a safe and effective natural anti-inflammatory drug. Herein, we evaluated the long-term therapeutic effects of a single intravitreal injection of poly(lactide-co-glycolide) PLGA-icariin in a rat model of anterior ischemic optic neuropathy (rAION). Treatment with PLGA microspheres of icariin preserved the visual function and RGC density for 1 month in the rAION model. In addition, ON edema and macrophage infiltration were inhibited by treating PLGA microspheres of icariin. We found that the binding complex of icariin and CCAAT enhancer binding protein beta (CEBP-ß) significantly induced endogenous granulocyte colony-stimulating factor (G-CSF) expression to activate noncanonical nuclear factor kappa B (NF-κB) signaling pathway by promoting an alternative phosphorylation reaction of IKK-ß. Activation of noncanonical NF-κB signaling pathway promoted the M2 microglia/macrophage polarization and AKT1 activation, which prevented neuroinflammation and RGC apoptosis after ON infarct. This study concluded that protective mechanism of icariin is a CEBP-ß/G-CSF axis-induced noncanonical NF-κB activation, which provides the long-term neuroprotective effects via anti-inflammatory and antiapoptotic actions after ON ischemia.

10.
Thorax ; 77(12): 1176-1186, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35580897

RESUMEN

INTRODUCTION: Chronic lung disease, that is, bronchopulmonary dysplasia (BPD) is the most common complication in preterm infants and develops as a consequence of the misguided formation of the gas-exchange area undergoing prenatal and postnatal injury. Subsequent vascular disease and its progression into pulmonary arterial hypertension critically determines long-term outcome in the BPD infant but lacks identification of early, disease-defining changes. METHODS: We link impaired bone morphogenetic protein (BMP) signalling to the earliest onset of vascular pathology in the human preterm lung and delineate the specific effects of the most prevalent prenatal and postnatal clinical risk factors for lung injury mimicking clinically relevant conditions in a multilayered animal model using wild-type and transgenic neonatal mice. RESULTS: We demonstrate (1) the significant reduction in BMP receptor 2 (BMPR2) expression at the onset of vascular pathology in the lung of preterm infants, later mirrored by reduced plasma BMP protein levels in infants with developing BPD, (2) the rapid impairment (and persistent change) of BMPR2 signalling on postnatal exposure to hyperoxia and mechanical ventilation, aggravated by prenatal cigarette smoke in a preclinical mouse model and (3) a link to defective alveolar septation and matrix remodelling through platelet derived growth factor-receptor alpha deficiency. In a treatment approach, we partially reversed vascular pathology by BMPR2-targeted treatment with FK506 in vitro and in vivo. CONCLUSION: We identified impaired BMP signalling as a hallmark of early vascular disease in the injured neonatal lung while outlining its promising potential as a future biomarker or therapeutic target in this growing, high-risk patient population.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Lesiones del Sistema Vascular , Lactante , Recién Nacido , Humanos , Ratones , Animales , Recien Nacido Prematuro , Lesiones del Sistema Vascular/complicaciones , Lesiones del Sistema Vascular/patología , Displasia Broncopulmonar/etiología , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Hiperoxia/patología , Pulmón , Ratones Transgénicos , Factores de Riesgo , Animales Recién Nacidos
11.
Cytotherapy ; 24(8): 774-788, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35613962

RESUMEN

The ISCT Scientific Signature Series Symposium "Advances in Cell and Gene Therapies for Lung Diseases and Critical Illnesses" was held as an independent symposium in conjunction with the biennial meeting, "Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases," which took place July 12-15, 2021, at the University of Vermont. This is the third Respiratory System-based Signature Series event; the first 2, "Tracheal Bioengineering, the Next Steps" and "Cellular Therapies for Pulmonary Diseases and Critical Illnesses: State of the Art of European Science," took place in 2014 and 2015, respectively. Cell- and gene-based therapies for respiratory diseases and critical illnesses continue to be a source of great promise and opportunity. This reflects ongoing advancements in understanding of the mechanisms by which cell-based therapies, particularly those using mesenchymal stromal cells (MSCs), can mitigate different lung injuries and the increasing sophistication with which preclinical data is translated into clinical investigations. This also reflects continuing evolution in gene transfer vectors, including those designed for in situ gene editing in parallel with those targeting gene or cell replacement. Therefore, this symposium convened global thought leaders in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value of cell- and gene-based therapies for patients with respiratory diseases and critical illnesses.


Asunto(s)
Enfermedad Crítica , Enfermedades Pulmonares , Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedad Crítica/terapia , Terapia Genética , Humanos , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/terapia , Células Madre
12.
Mol Ther ; 30(1): 223-237, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33794364

RESUMEN

Cystic fibrosis (CF) is a monogenic disease caused by impaired production and/or function of the CF transmembrane conductance regulator (CFTR) protein. Although we have previously shown correction of the most common pathogenic mutation, there are many other pathogenic mutations throughout the CF gene. An autologous airway stem cell therapy in which the CFTR cDNA is precisely inserted into the CFTR locus may enable the development of a durable cure for almost all CF patients, irrespective of the causal mutation. Here, we use CRISPR-Cas9 and two adeno-associated viruses (AAVs) carrying the two halves of the CFTR cDNA to sequentially insert the full CFTR cDNA along with a truncated CD19 (tCD19) enrichment tag in upper airway basal stem cells (UABCs) and human bronchial epithelial cells (HBECs). The modified cells were enriched to obtain 60%-80% tCD19+ UABCs and HBECs from 11 different CF donors with a variety of mutations. Differentiated epithelial monolayers cultured at air-liquid interface showed restored CFTR function that was >70% of the CFTR function in non-CF controls. Thus, our study enables the development of a therapy for almost all CF patients, including patients who cannot be treated using recently approved modulator therapies.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Sistemas CRISPR-Cas , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/terapia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Humanos , Mutación , Células Madre/metabolismo
13.
Antioxidants (Basel) ; 10(12)2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34943037

RESUMEN

Traumatic optic neuropathy (TON) may cause severe visual loss following direct or indirect head trauma which may result in optic nerve injuries and therefore contribute to the subsequent loss of retinal ganglion cells by inflammatory mediators and reactive oxygen species (ROS). Granulocyte colony-stimulating factor (G-CSF) provides the anti-inflammatory and anti-oxidative actions but has a short half-life and also induces leukocytosis upon typical systemic administration. The purpose of the present study was to investigate the relationship between the anti-oxidative response and neuroprotective effects of long-acting pegylated human G-CSF (PEG-G-CSF) in a rat model of optic nerve crush (ONC). Adult male Wistar rats (150-180 g) were chosen to have a sham operation in one eye and have ONC in the other. PEG-G-CSF or phosphate-buffered saline (PBS control) was immediately administered after ONC by intravitreal injection (IVI). We found the IVI of PEG-G-CSF does not induce systemic leukocytosis, but increases survival of RGCs and preserves the visual function after ONC. TUNEL assays showed fewer apoptotic cells in the retina in the PEG-G-CSF-treated eyes. The number of sorely ED1-positive cells was attenuated at the lesion site in the PEG-G-CSF-treated eyes. Immunoblotting showed up-regulation of p-Akt1, Nrf2, Sirt3, and HO-1 in the ON of the PEG-G-CSF-treated eyes. Our results demonstrated that one IVI of long-acting PEG-G-CSF is neuroprotective in the rONC. PEG-G-CSF activates the p-Akt1/Nrf2/Sirt3 and the p-Akt1/Nrf2/HO-1 axes to provide the antioxidative action and further attenuated RGC apoptosis and neuroinflammation. This provides crucial preclinical information for the development of alternative therapy with IVI of PEG-G-CSF in TON.

14.
Respir Res ; 22(1): 265, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34666752

RESUMEN

RATIONALE: αv integrins, key regulators of transforming growth factor-ß activation and fibrogenesis in in vivo models of pulmonary fibrosis, are expressed on abnormal epithelial cells (αvß6) and fibroblasts (αvß1) in fibrotic lungs. OBJECTIVES: We evaluated multiple αv integrin inhibition strategies to assess which most effectively reduced fibrogenesis in explanted lung tissue from patients with idiopathic pulmonary fibrosis. METHODS: Selective αvß6 and αvß1, dual αvß6/αvß1, and multi-αv integrin inhibitors were characterized for potency, selectivity, and functional activity by ligand binding, cell adhesion, and transforming growth factor-ß cell activation assays. Precision-cut lung slices generated from lung explants from patients with idiopathic pulmonary fibrosis or bleomycin-challenged mouse lungs were treated with integrin inhibitors or standard-of-care drugs (nintedanib or pirfenidone) and analyzed for changes in fibrotic gene expression or TGF-ß signaling. Bleomycin-challenged mice treated with dual αvß6/αvß1 integrin inhibitor, PLN-74809, were assessed for changes in pulmonary collagen deposition and Smad3 phosphorylation. MEASUREMENTS AND MAIN RESULTS: Inhibition of integrins αvß6 and αvß1 was additive in reducing type I collagen gene expression in explanted lung tissue slices from patients with idiopathic pulmonary fibrosis. These data were replicated in fibrotic mouse lung tissue, with no added benefit observed from inhibition of additional αv integrins. Antifibrotic efficacy of dual αvß6/αvß1 integrin inhibitor PLN-74809 was confirmed in vivo, where dose-dependent inhibition of pulmonary Smad3 phosphorylation and collagen deposition was observed. PLN-74809 also, more potently, reduced collagen gene expression in fibrotic human and mouse lung slices than clinically relevant concentrations of nintedanib or pirfenidone. CONCLUSIONS: In the fibrotic lung, dual inhibition of integrins αvß6 and αvß1 offers the optimal approach for blocking fibrogenesis resulting from integrin-mediated activation of transforming growth factor-ß.


Asunto(s)
Antifibróticos/farmacología , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Integrina alfa6beta1/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Receptores de Vitronectina/antagonistas & inhibidores , Animales , Bleomicina , Línea Celular , Técnicas de Cocultivo , Cadena alfa 1 del Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Integrina alfa6beta1/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Fosforilación , Receptores de Vitronectina/metabolismo , Transducción de Señal , Proteína smad3/metabolismo
15.
J Heart Lung Transplant ; 40(8): 856-859, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34059432

RESUMEN

As the world responds to the global crisis of the COVID-19 pandemic an increasing number of patients are experiencing increased morbidity as a result of multi-organ involvement. Of these, a small proportion will progress to end-stage lung disease, become dialysis dependent, or both. Herein, we describe the first reported case of a successful combined lung and kidney transplantation in a patient with COVID-19. Lung transplantation, isolated or combined with other organs, is feasible and should be considered for select patients impacted by this deadly disease.


Asunto(s)
Lesión Renal Aguda/etiología , Lesión Renal Aguda/cirugía , COVID-19/complicaciones , COVID-19/cirugía , Trasplante de Riñón , Trasplante de Pulmón , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/cirugía , Humanos , Masculino , Persona de Mediana Edad
16.
AJOG Glob Rep ; 1(4): 100026, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36277462

RESUMEN

BACKGROUND: Although risk factors of preterm deliveries across the world have been extensively studied, the trends and risk factors of preterm deliveries for the population of rural India, and specifically tribal women, remain unexplored. OBJECTIVE: The aim of this study was to assess and compare the preterm delivery rates among women from a rural area in Gujarat, India, based on socioeconomic and clinical factors. The second aim of the study was to assess and identify predictors or risk factors for preterm deliveries. STUDY DESIGN: This was a retrospective medical record review study investigating deliveries that took place at the Kasturba Maternity Hospital in Jhagadia, Gujarat, from January 2012 to June 2019 (N=32,557). We performed odds ratio and adjusted odds ratio analyses of preterm delivery risk factors. Lastly, we also considered the neonatal outcomes of preterm deliveries, both overall and comparing tribal and nontribal mothers. RESULTS: For the study period, the tribal preterm delivery rate was 19.7% and the nontribal preterm delivery rate was 13.9%; the rate remained consistent for both groups over the 7-year study period. Adjusted odds ratios indicated that tribal status (adjusted odds ratio, 1.16; 95% confidence interval, 1.08-1.24), maternal illiteracy ((adjusted odds ratio, 1.29, 95% confidence interval, 1.18-1.42), paternal illiteracy (adjusted odds ratio, 1.27; 95% confidence interval, 1.15-1.410), hemoglobin <10 g/dL (adjusted odds ratio, 1.41; 95% confidence interval, 1.32-1.51), and a lack of antenatal care (adjusted odds ratio, 2.15; 95% confidence interval, 1.94-2.37) are significantly associated with higher odds of preterm delivery. The overall stillbirth rate among tribal women was 3.06% and 1.73% among nontribal women; among preterm deliveries, tribal women have a higher proportion of stillbirth outcomes (11.77%) than nontribal women (8.86%). CONCLUSION: Consistent with existing literature, risk factors for preterm deliveries in rural India include clinical factors such as a lack of antenatal care and low hemoglobin. In addition, sociodemographic factors, such as tribal status, are independently associated with higher odds of delivering preterm. The higher rates of preterm deliveries among tribal women need to be studied further to detail the underlying reasons of how it can influence a woman's delivery outcome.

17.
Nature ; 588(7839): 670-675, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33238290

RESUMEN

The distal lung contains terminal bronchioles and alveoli that facilitate gas exchange. Three-dimensional in vitro human distal lung culture systems would strongly facilitate the investigation of pathologies such as interstitial lung disease, cancer and coronavirus disease 2019 (COVID-19) pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we describe the development of a long-term feeder-free, chemically defined culture system for distal lung progenitors as organoids derived from single adult human alveolar epithelial type II (AT2) or KRT5+ basal cells. AT2 organoids were able to differentiate into AT1 cells, and basal cell organoids developed lumens lined with differentiated club and ciliated cells. Single-cell analysis of KRT5+ cells in basal organoids revealed a distinct population of ITGA6+ITGB4+ mitotic cells, whose offspring further segregated into a TNFRSF12Ahi subfraction that comprised about ten per cent of KRT5+ basal cells. This subpopulation formed clusters within terminal bronchioles and exhibited enriched clonogenic organoid growth activity. We created distal lung organoids with apical-out polarity to present ACE2 on the exposed external surface, facilitating infection of AT2 and basal cultures with SARS-CoV-2 and identifying club cells as a target population. This long-term, feeder-free culture of human distal lung organoids, coupled with single-cell analysis, identifies functional heterogeneity among basal cells and establishes a facile in vitro organoid model of human distal lung infections, including COVID-19-associated pneumonia.


Asunto(s)
COVID-19/virología , Pulmón/citología , Modelos Biológicos , Organoides/citología , Organoides/virología , SARS-CoV-2/fisiología , Técnicas de Cultivo de Tejidos , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , COVID-19/metabolismo , COVID-19/patología , Diferenciación Celular , División Celular , Células Clonales/citología , Células Clonales/metabolismo , Células Clonales/virología , Humanos , Técnicas In Vitro , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Integrina alfa6/análisis , Integrina beta4/análisis , Queratina-5/análisis , Organoides/metabolismo , Neumonía Viral/metabolismo , Neumonía Viral/patología , Neumonía Viral/virología , SARS-CoV-2/crecimiento & desarrollo , Análisis de la Célula Individual , Receptor de TWEAK/análisis
18.
bioRxiv ; 2020 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-32743583

RESUMEN

The distal lung contains terminal bronchioles and alveoli that facilitate gas exchange and is affected by disorders including interstitial lung disease, cancer, and SARS-CoV-2-associated COVID-19 pneumonia. Investigations of these localized pathologies have been hindered by a lack of 3D in vitro human distal lung culture systems. Further, human distal lung stem cell identification has been impaired by quiescence, anatomic divergence from mouse and lack of lineage tracing and clonogenic culture. Here, we developed robust feeder-free, chemically-defined culture of distal human lung progenitors as organoids derived clonally from single adult human alveolar epithelial type II (AT2) or KRT5 + basal cells. AT2 organoids exhibited AT1 transdifferentiation potential, while basal cell organoids progressively developed lumens lined by differentiated club and ciliated cells. Organoids consisting solely of club cells were not observed. Upon single cell RNA-sequencing (scRNA-seq), alveolar organoids were composed of proliferative AT2 cells; however, basal organoid KRT5 + cells contained a distinct ITGA6 + ITGB4 + mitotic population whose proliferation segregated to a TNFRSF12A hi subfraction. Clonogenic organoid growth was markedly enriched within the TNFRSF12A hi subset of FACS-purified ITGA6 + ITGB4 + basal cells from human lung or derivative organoids. In vivo, TNFRSF12A + cells comprised ~10% of KRT5 + basal cells and resided in clusters within terminal bronchioles. To model COVID-19 distal lung disease, we everted the polarity of basal and alveolar organoids to rapidly relocate differentiated club and ciliated cells from the organoid lumen to the exterior surface, thus displaying the SARS-CoV-2 receptor ACE2 on the outwardly-facing apical aspect. Accordingly, basal and AT2 apical-out organoids were infected by SARS-CoV-2, identifying club cells as a novel target population. This long-term, feeder-free organoid culture of human distal lung alveolar and basal stem cells, coupled with single cell analysis, identifies unsuspected basal cell functional heterogeneity and exemplifies progenitor identification within a slowly proliferating human tissue. Further, our studies establish a facile in vitro organoid model for human distal lung infectious diseases including COVID-19-associated pneumonia.

20.
Artículo en Inglés | MEDLINE | ID: mdl-32179507

RESUMEN

The distal lung is a honeycomb-like collection of delicate gas exchange sacs called alveoli lined by two interspersed epithelial cell types: the cuboidal, surfactant-producing alveolar type II (AT2) and the flat, gas-exchanging alveolar type I (AT1) cell. During aging, a subset of AT2 cells expressing the canonical Wnt target gene, Axin2, function as stem cells, renewing themselves while generating new AT1 and AT2 cells. Wnt activity endows AT2 cells with proliferative competency, enabling them to respond to activating cues, and simultaneously blocks AT2 to AT1 cell transdifferentiation. Acute alveolar injury rapidly expands the AT2 stem cell pool by transiently inducing Wnt signaling activity in "bulk" AT2 cells, facilitating rapid epithelial repair. AT2 cell "stemness" is thus tightly regulated by access to Wnts, supplied by a specialized single-cell fibroblast niche during maintenance and by AT2 cells themselves during injury repair. Two non-AT2 "reserve" cell populations residing in the distal airways also contribute to alveolar repair, but only after widespread epithelial injury, when they rapidly proliferate, migrate, and differentiate into airway and alveolar lineages. Here, we review alveolar renewal and repair with a focus on the niches, rather than the stem cells, highlighting what is known about the cellular and molecular mechanisms by which they control stem cell activity in vivo.


Asunto(s)
Células Epiteliales Alveolares/fisiología , Regeneración , Nicho de Células Madre , Envejecimiento/fisiología , Animales , Humanos , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...