Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Ther ; 32(5): 1425-1444, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38504518

ABSTRACT

Pathological ocular angiogenesis has long been associated with myeloid cell activation. However, the precise cellular and molecular mechanisms governing the intricate crosstalk between the immune system and vascular changes during ocular neovascularization formation remain elusive. In this study, we demonstrated that the absence of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells led to a substantial accumulation of microglia and macrophage subsets during the neovascularization process. Our single-cell RNA sequencing data analysis revealed a remarkable increase in the expression of the secreted phosphoprotein 1 (Spp1) gene within these microglia and macrophages, identifying subsets of Spp1-expressing microglia and macrophages during neovascularization formation in angiogenesis mouse models. Notably, the number of Spp1-expressing microglia and macrophages exhibited further elevation during neovascularization in mice lacking myeloid SOCS3. Moreover, our investigation unveiled the Spp1 gene as a direct transcriptional target gene of signal transducer and activator of transcription 3. Importantly, pharmaceutical activation of SOCS3 or blocking of SPP1 resulted in a significant reduction in pathological neovascularization. In conclusion, our study highlights the pivotal role of the SOCS3/STAT3/SPP1 axis in the regulation of pathological retinal angiogenesis.


Subject(s)
Disease Models, Animal , Macrophages , Microglia , Osteopontin , Retinal Neovascularization , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Animals , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Macrophages/metabolism , Mice , Microglia/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/genetics , Retinal Neovascularization/etiology , Osteopontin/metabolism , Osteopontin/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Gene Expression Regulation , Signal Transduction , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Angiogenesis
2.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L770-L785, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38563994

ABSTRACT

Novel therapies are needed for bronchopulmonary dysplasia (BPD) because no effective treatment exists. Mesenchymal stromal cell extracellular vesicles (MSC-sEVs) have therapeutic efficacy in a mouse pup neonatal hyperoxia BPD model. We tested the hypothesis that MSC-sEVs will improve lung functional and structural development in mechanically ventilated preterm lambs. Preterm lambs (∼129 days; equivalent to human lung development at ∼28 wk gestation) were exposed to antenatal steroids, surfactant, caffeine, and supported by mechanical ventilation for 6-7 days. Lambs were randomized to blinded treatment with either MSC-sEVs (human bone marrow MSC-derived; 2 × 1011 particles iv; n = 8; 4 F/4 M) or vehicle control (saline iv; 4 F/4 M) at 6 and 78 h post delivery. Physiological targets were pulse oximetry O2 saturation 90-94% ([Formula: see text] 60-90 mmHg), [Formula: see text] 45-60 mmHg (pH 7.25-7.35), and tidal volume 5-7 mL/kg. MSC-sEVs-treated preterm lambs tolerated enteral feedings compared with vehicle control preterm lambs. Differences in weight patterns were statistically significant. Respiratory severity score, oxygenation index, A-a gradient, distal airspace wall thickness, and smooth muscle thickness around terminal bronchioles and pulmonary arterioles were significantly lower for the MSC-sEVs group. S/F ratio, radial alveolar count, secondary septal volume density, alveolar capillary surface density, and protein abundance of VEGF-R2 were significantly higher for the MSC-sEVs group. MSC-sEVs improved respiratory system physiology and alveolar formation in mechanically ventilated preterm lambs. MSC-sEVs may be an effective and safe therapy for appropriate functional and structural development of the lung in preterm infants who require mechanical ventilation and are at risk of developing BPD.NEW & NOTEWORTHY This study focused on potential treatment of preterm infants at risk of developing bronchopulmonary dysplasia (BPD), for which no effective treatment exists. We tested treatment of mechanically ventilated preterm lambs with human mesenchymal stromal cell extracellular vesicles (MSC-sEVs). The results show improved respiratory gas exchange and parenchymal growth of capillaries and epithelium that are necessary for alveolar formation. Our study provides new mechanistic insight into potential efficacy of MSC-sEVs for preterm infants at risk of developing BPD.


Subject(s)
Animals, Newborn , Bronchopulmonary Dysplasia , Extracellular Vesicles , Lung , Mesenchymal Stem Cells , Respiration, Artificial , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/metabolism , Lung/metabolism , Lung/pathology , Respiration, Artificial/adverse effects , Respiration, Artificial/methods , Sheep , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/therapy , Bronchopulmonary Dysplasia/metabolism , Humans , Female
3.
Angiogenesis ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38483712

ABSTRACT

Pathological retinal angiogenesis profoundly impacts visual function in vascular eye diseases, such as retinopathy of prematurity (ROP) in preterm infants and age-related macular degeneration in the elderly. While the involvement of photoreceptors in these diseases is recognized, the underlying mechanisms remain unclear. This study delved into the pivotal role of photoreceptors in regulating abnormal retinal blood vessel growth using an oxygen-induced retinopathy (OIR) mouse model through the c-Fos/A disintegrin and metalloprotease 17 (Adam17) axis. Our findings revealed a significant induction of c-Fos expression in rod photoreceptors, and c-Fos depletion in these cells inhibited pathological neovascularization and reduced blood vessel leakage in the OIR mouse model. Mechanistically, c-Fos directly regulated the transcription of Adam17 a shedding protease responsible for the production of bioactive molecules involved in inflammation, angiogenesis, and cell adhesion and migration. Furthermore, we demonstrated the therapeutic potential by using an adeno-associated virus carrying a rod photoreceptor-specific short hairpin RNA against c-fos which effectively mitigated abnormal retinal blood vessel overgrowth, restored retinal thickness, and improved electroretinographic (ERG) responses. In conclusion, this study highlights the significance of photoreceptor c-Fos in ROP pathology, offering a novel perspective for the treatment of this disease.

4.
Adv Neonatal Care ; 24(1): 46-57, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38215025

ABSTRACT

BACKGROUND: Boston Children's Hospital's Level IV Neonatal Intensive Care Unit (NICU) discharges about a third of its medically complex infants home. Parental feedback indicated a need for more education and training in discharge preparation. PURPOSE: The NICU to Nursery (N2N) program was created to better prepare parents to care for their medically complex infants following Level IV NICU discharge. The goals were to (1) mitigate safety risks, (2) assess parent satisfaction, (3) assess pediatric primary care providers' (PCPs') satisfaction, (4) assess community visiting nurses' and PCPs' knowledge deficits, and (5) develop educational materials. METHODS: The N2N program provided parents with pre- and postdischarge assessments with an experienced nurse. Parents completed a survey following assessments to measure satisfaction. To enhance PCPs' knowledge, they were sent summary reports and asked for feedback. PCP feedback, along with a needs assessment of community visiting nurses, guided the development of free Web-based educational videos. RESULTS: One hundred and fifty-five parents participated in the N2N program. Parents' educational needs included medication education, safe sleep, and well-infant care, with some requiring significant nursing interventions for safety risk mitigation. Most PCPs found the home visit reports helpful. Knowledge deficits identified among PCPs and community visiting nurses included management of tubes and drains, growth and nutrition, and emergency response. More than 100,000 providers viewed the 3 Web-based educational videos developed. IMPLICATIONS FOR PRACTICE AND RESEARCH: The N2N program fills a crucial gap in the transition of medically complex infants discharged home. The next steps are developing best practices for virtual in-home assessments.


Subject(s)
Aftercare , Intensive Care Units, Neonatal , Infant, Newborn , Infant , Humans , Child , Patient Discharge , Infant Care , Needs Assessment , Parents
5.
Stem Cells ; 40(11): 991-1007, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36044737

ABSTRACT

Over the past decades, substantial advances in neonatal medical care have increased the survival of extremely premature infants. However, there continues to be significant morbidity associated with preterm birth with common complications including bronchopulmonary dysplasia (BPD), necrotizing enterocolitis (NEC), neuronal injury such as intraventricular hemorrhage (IVH) or hypoxic ischemic encephalopathy (HIE), as well as retinopathy of prematurity (ROP). Common developmental immune and inflammatory pathways underlie the pathophysiology of such complications providing the opportunity for multisystem therapeutic approaches. To date, no single therapy has proven to be effective enough to prevent or treat the sequelae of prematurity. In the past decade mesenchymal stem/stromal cell (MSC)-based therapeutic approaches have shown promising results in numerous experimental models of neonatal diseases. It is now accepted that the therapeutic potential of MSCs is comprised of their secretome, and several studies have recognized the small extracellular vesicles (sEVs) as the paracrine vector. Herein, we review the current literature on the MSC-EVs as potential therapeutic agents in neonatal diseases and comment on the progress and challenges of their translation to the clinical setting.


Subject(s)
Bronchopulmonary Dysplasia , Enterocolitis, Necrotizing , Extracellular Vesicles , Infant, Newborn, Diseases , Mesenchymal Stem Cells , Premature Birth , Infant , Pregnancy , Female , Infant, Newborn , Humans , Premature Birth/metabolism , Bronchopulmonary Dysplasia/therapy , Bronchopulmonary Dysplasia/metabolism , Infant, Newborn, Diseases/metabolism , Infant, Newborn, Diseases/therapy , Enterocolitis, Necrotizing/metabolism , Extracellular Vesicles/metabolism
6.
Am J Respir Cell Mol Biol ; 66(1): 86-95, 2022 01.
Article in English | MEDLINE | ID: mdl-34614384

ABSTRACT

In preeclamptic pregnancies, a variety of intrauterine alterations lead to abnormal placentation, release of inflammatory and/or antiangiogenic factors, and subsequent fetal growth restriction with significant potential to cause a primary insult to the developing fetal lung. Thus, modulation of the maternal intrauterine environment may be a key therapeutic avenue to prevent preeclampsia-associated developmental lung injury. A biologic therapy of interest is mesenchymal stromal cell-derived extracellular vesicles (MEx), which we have previously shown to ameliorate preeclamptic physiology through intrauterine immunomodulation. To evaluate the therapeutic potential of MEx to improve developmental lung injury in experimental preeclampsia, using the heme oxygenase-1-null mouse (Hmox1-/-) model, preeclamptic pregnant dams were administered intravenous antenatal MEx treatment during each week of pregnancy followed by analysis of fetal and postnatal lung tissues, amniotic fluid protein profiles, and lung explant and amniotic fluid cocultures in comparison with control and untreated preeclamptic pregnancies. We first identified that a preeclamptic intrauterine environment had a significant adverse impact on fetal lung development, including alterations in fetal lung developmental gene profiles in addition to postnatal alveolar and bronchial changes. Amniotic fluid proteomic analysis and fetal lung explant and amniotic fluid cocultures further demonstrated that maternally administered MEx altered the expression of multiple inflammatory mediators in the preeclamptic intrauterine compartment, resulting in the normalization of fetal lung branching morphogenesis and developmental gene expression. Our evaluation of fetal and postnatal parameters overall suggests that antenatal MEx treatment may provide a highly valuable preventative therapeutic modality for amelioration of lung development in preeclamptic disease.


Subject(s)
Extracellular Vesicles/metabolism , Lung Injury/prevention & control , Lung Injury/therapy , Mesenchymal Stem Cells/metabolism , Pre-Eclampsia/pathology , Amniotic Fluid/metabolism , Animals , Female , Fetus/embryology , Humans , Lung/embryology , Lung Injury/etiology , Mice , Pregnancy , Secretome/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 322(2): L179-L190, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34878940

ABSTRACT

Antenatal stressors such as chorioamnionitis (CA) increase the risk for bronchopulmonary dysplasia (BPD). Studies have shown that experimental BPD can be ameliorated by postnatal treatment with mesenchymal stromal cell-derived extracellular vesicles (MEx). However, the antenatal efficacy of MEx to prevent BPD is unknown. To determine whether antenatal MEx therapy attenuates intrauterine inflammation and preserves lung growth in a rat model of CA-induced BPD. At embryonic day (E)20, rat litters were treated with intra-amniotic injections of saline, endotoxin (ETX) to model chorioamnionitis, MEx, or ETX plus MEx followed by cesarean section delivery with placental harvest at E22. Placental and lung evaluations were conducted at day 0 and day 14, respectively. To assess the effects of ETX and MEx on lung growth in vitro, E15 lung explants were imaged for distal branching. Placental tissues from ETX-exposed pregnancies showed increased expression of inflammatory markers NLRP-3 and IL-1ß and altered spiral artery morphology. In addition, infant rats exposed to intrauterine ETX had reduced alveolarization and pulmonary vessel density (PVD), increased right ventricular hypertrophy (RVH), and decreased lung mechanics. Intrauterine MEx therapy of ETX-exposed pups reduced inflammatory cytokines, normalized spiral artery architecture, and preserved distal lung growth and mechanics. In vitro studies showed that MEx treatment enhanced distal lung branching and increased VEGF and SPC gene expression. Antenatal MEx treatment preserved distal lung growth and reduced intrauterine inflammation in a model of CA-induced BPD. We speculate that MEx may provide a novel therapeutic strategy to prevent BPD due to antenatal inflammation.


Subject(s)
Bronchopulmonary Dysplasia/etiology , Chorioamnionitis/pathology , Extracellular Vesicles/metabolism , Lung/growth & development , Mesenchymal Stem Cells/metabolism , Animals , Disease Models, Animal , Endotoxins , Female , Inflammation/pathology , Lung/blood supply , Lung/pathology , Placenta/pathology , Pregnancy , Rats, Sprague-Dawley , Signal Transduction
8.
Am J Respir Crit Care Med ; 204(12): 1418-1432, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34699335

ABSTRACT

Rationale: Mesenchymal stem/stromal cell (MSC)-small extracellular vesicle (MEx) treatment has shown promise in experimental models of neonatal lung injury. The molecular mechanisms by which MEx afford beneficial effects remain incompletely understood. Objectives: To investigate the therapeutic mechanism of action through assessment of MEx biodistribution and impact on immune cell phenotypic heterogeneity. Methods: MEx were isolated from the conditioned medium of human umbilical cord Wharton's jelly-derived MSCs. Newborn mice were exposed to hyperoxia (HYRX, 75% O2) from birth and returned to room air at Postnatal Day 14 (PN14). Mice received either a bolus intravenous MEx dose at PN4 or bone marrow-derived myeloid cells (BMDMy) pretreated with MEx. Animals were killed at PN4, PN7, PN14, or PN28 to characterize MEx biodistribution or for assessment of pulmonary parameters. The therapeutic role of MEx-educated BMDMy was determined in vitro and in vivo. Measurements and Main Results: MEx therapy ameliorated core histological features of HYRX-induced neonatal lung injury. Biodistribution and mass cytometry studies demonstrated that MEx localize in the lung and interact with myeloid cells. MEx restored the apportion of alveolar macrophages in the HYRX-injured lung and concomitantly suppressed inflammatory cytokine production. In vitro and ex vivo studies revealed that MEx promoted an immunosuppressive BMDMy phenotype. Functional assays demonstrated that the immunosuppressive actions of BMDMy are driven by phenotypically and epigenetically reprogrammed monocytes. Adoptive transfer of MEx-educated BMDMy, but not naive BMDMy, restored alveolar architecture, blunted fibrosis and pulmonary vascular remodeling, and improved exercise capacity. Conclusions: MEx ameliorate hyperoxia-induced neonatal lung injury though epigenetic and phenotypic reprogramming of myeloid cells.


Subject(s)
Bronchopulmonary Dysplasia/prevention & control , Cord Blood Stem Cell Transplantation/methods , Epigenesis, Genetic , Extracellular Vesicles/transplantation , Hyperoxia/complications , Myeloid Cells/metabolism , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Humans , Mice , Phenotype , Treatment Outcome
9.
Biol Reprod ; 104(2): 457-467, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33112369

ABSTRACT

Human umbilical cord-derived mesenchymal stromal cells (MSCs) are a widely recognized treatment modality for a variety of preclinical disease models and have been transitioned to human clinical trials. We have previously shown in neonatal lung disease that the therapeutic capacity of MSCs is conferred by their secreted extracellular vesicles (MEx), which function primarily through immunomodulation. We hypothesize that MEx have significant therapeutic potential pertinent to immune-mediated gestational diseases. Of particular interest is early-onset preeclampsia, which can be caused by alterations of the maternal intrauterine immune environment. Using a heme-oxygenase-1 null mouse model of pregnancy loss with preeclampsia-like features, we examined the preventative effects of maternal MEx treatment early in pregnancy. Heme oxygenase-1 null females (Hmox1-/-) or wild-type control females were bred in homozygous matings followed by evaluation of maternal and fetal parameters. A single dose of MEx was administered intravenously on gestational day (GD)1 to Hmox1-/- females (Hmox1-/- MEx). Compared with untreated Hmox1-/- females, Hmox1-/- MEx-treated pregnancies showed significant improvement in fetal loss, intrauterine growth restriction, placental spiral artery modification, and maternal preeclamptic stigmata. Biodistribution studies demonstrated that MEx localize to a subset of cells in the preimplantation uterus. Further, mass cytometric (CyTOF) evaluation of utero-placental leukocytes in Hmox1-/- MEx versus untreated pregnancies showed alteration in the abundance, surface marker repertoire, and cytokine profiles of multiple immune populations. Our data demonstrate the therapeutic potential of MEx to optimize the intrauterine immune environment and prevent maternal and fetal sequelae of preeclamptic disease.


Subject(s)
Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Pre-Eclampsia/prevention & control , Animals , Extracellular Vesicles , Female , Fetal Growth Retardation , Gene Expression Regulation , Heme Oxygenase-1/genetics , Humans , Immunomodulation , Membrane Proteins/genetics , Mesenchymal Stem Cells , Mice , Mice, Knockout , Pregnancy , Umbilical Cord , Uterus
10.
Pediatr Res ; 89(3): 446-455, 2021 02.
Article in English | MEDLINE | ID: mdl-32521539

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a major complication in prematurely born infants. Pulmonary hypertension (PH) associated with BPD (BPD-PH) is characterized by alveolar diffusion impairment, abnormal vascular remodeling, and rarefication of pulmonary vessels (vascular growth arrest), which lead to increased pulmonary vascular resistance and right heart failure. About 25% of infants with moderate to severe BPD develop BPD-PH that is associated with high morbidity and mortality. The recent evolution of broader PH-targeted pharmacotherapy in adults has opened up new treatment options for infants with BPD-PH. Sildenafil became the mainstay of contemporary BPD-PH therapy. Additional medications, such as endothelin receptor antagonists and prostacyclin analogs/mimetics, are increasingly being investigated in infants with PH. However, pediatric data from prospective or randomized controlled trials are still sparse. We discuss comprehensive diagnostic and therapeutic strategies for BPD-PH and briefly review the relevant differential diagnoses of parenchymal and interstitial developmental lung diseases. In addition, we provide a practical framework for the management of children with BPD-PH, incorporating the modified definition and classification of pediatric PH from the 2018 World Symposium on Pulmonary Hypertension, and the 2019 EPPVDN consensus recommendations on established and newly developed therapeutic strategies. Finally, current gaps of knowledge and future research directions are discussed. IMPACT: PH in BPD substantially increases mortality. Treatment of BPD-PH should be conducted by an interdisciplinary team and follow our new treatment algorithm while still kept tailored to the individual patient. We discuss recent developments in BPD-PH, make recommendations on diagnosis, monitoring and treatment of PH in BPD, and address current gaps of knowledge and potential research directions. We provide a practical framework, including a new treatment algorithm, for the management of children with BPD-PH, incorporating the modified definition and classification of pediatric PH (2018 WSPH) and the 2019 EPPVDN consensus recommendations on established and newly developed therapeutic strategies for BPD-PH.


Subject(s)
Bronchopulmonary Dysplasia/complications , Hypertension, Pulmonary/etiology , Infant, Premature, Diseases/physiopathology , Biomarkers/blood , Bronchopulmonary Dysplasia/diagnosis , Bronchopulmonary Dysplasia/physiopathology , Bronchopulmonary Dysplasia/therapy , Cardiac Catheterization , Cardiac Surgical Procedures , Echocardiography , Endothelin Receptor Antagonists/therapeutic use , Heart Failure/etiology , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Infant, Newborn , Infant, Premature , Magnetic Resonance Imaging , Nitric Oxide/metabolism , Oxygen Inhalation Therapy , Prostaglandins I/therapeutic use , Sildenafil Citrate/therapeutic use , Tomography, X-Ray Computed , Tricuspid Valve Insufficiency/diagnostic imaging , Tricuspid Valve Insufficiency/etiology , Vascular Resistance , Vasodilator Agents/therapeutic use
11.
Am J Respir Crit Care Med ; 202(8): 1146-1158, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32551816

ABSTRACT

Rationale: Antenatal inflammation with placental dysfunction is strongly associated with high bronchopulmonary dysplasia (BPD) risk in preterm infants. Whether antenatal or postnatal HIF (hypoxia-inducible factor) augmentation can preserve lung structure and function and prevent pulmonary hypertension after intrauterine inflammation is controversial.Objectives: To determine whether antenatal or postnatal prolyl-hydroxylase inhibitor (PHi) therapy increases lung HIF expression, preserves lung growth and function, and prevents pulmonary hypertension in a rat model of chorioamnionitis-induced BPD caused by antenatal inflammation.Methods: Endotoxin (ETX) was administered to pregnant rats by intraamniotic injection at Embryonic Day 20, and pups were delivered by cesarean section at Embryonic Day 22. Selective PHi drugs, dimethyloxalylglycine or GSK360A, were administered into the amniotic space at Embryonic Day 20 or after birth by intraperitoneal injection for 2 weeks. Placentas and lung tissue were collected at birth for morphometric and Western blot measurements of HIF-1a, HIF-2a, VEGF (vascular endothelial growth factor), and eNOS (endothelial nitric oxide synthase) protein contents. At Day 14, lung function was assessed, and tissues were harvested to determine alveolarization by radial alveolar counts, pulmonary vessel density, and right ventricle hypertrophy (RVH).Measurements and Main Results: Antenatal PHi therapy preserves lung alveolar and vascular growth and lung function and prevents RVH after intrauterine ETX exposure. Antenatal administration of PHi markedly upregulates lung HIF-1a, HIF-2a, VEGF, and eNOS expression after ETX exposure.Conclusions: HIF augmentation improves lung structure and function, prevents RVH, and improves placental structure following antenatal ETX exposure. We speculate that antenatal or postnatal PHi therapy may provide novel strategies to prevent BPD due to antenatal inflammation.


Subject(s)
Bronchopulmonary Dysplasia/drug therapy , Hypoxia-Inducible Factor 1/metabolism , Lung/drug effects , Peptide PHI/pharmacology , Pregnancy, Animal , Amino Acids, Dicarboxylic/pharmacology , Animals , Animals, Newborn , Blotting, Western , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Endotoxins/adverse effects , Endotoxins/pharmacology , Female , Hypoxia-Inducible Factor 1/drug effects , Immunohistochemistry , In Vitro Techniques , Injections, Intralesional , Lung/embryology , Pregnancy , Prenatal Care , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/growth & development , Pulmonary Circulation/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Reference Values , Respiratory Function Tests , Tissue Culture Techniques
12.
Am J Physiol Lung Cell Mol Physiol ; 318(1): L125-L134, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31664855

ABSTRACT

Exposure to hypoxia causes an inflammatory reaction in the mouse lung, and this response can be modulated by overexpressing the hypoxia-inducible stress-response enzyme, heme oxygenase-1 (HO-1). We hypothesized that the inflammasome activity may be a central pathway by which HO-1 controls pulmonary inflammation following alveolar hypoxia. Therefore, we investigated whether HO-1 controls inflammasome activation by altering its expression in macrophages primed with classic NOD-like receptor containing a pyrin domain 3 (NLRP3) inducers, and in murine lungs lacking HO-1 and exposed to acute hypoxia. We found that lack of HO-1 activated lipopolysaccharide (LPS) and ATP-treated bone marrow-derived macrophages, causing an increase in secreted levels of cleaved interleukin (IL)-1B, IL-18, and caspase-1, markers of increased inflammasome activity, whereas HO-1 overexpression suppressed IL-1B, NLRP3, and IL-18. The production of cleaved IL-1B and the activation of caspase-1 in LPS- and ATP-primed macrophages were inhibited by hemin, an HO-1 inducer, and two HO-1 enzymatic products [bilirubin and carbon monoxide (CO)]. Exposure of mice to hypoxia induced the expression of several inflammasome mRNA components (IL-1B, Nlrp3, and caspase-1), and this was further augmented by HO-1 deficiency. This pronounced inflammasome activation was detected as increased protein levels of apoptosis-associated speck-like protein containing a COOH-terminal caspase recruitment domain, IL-18, procaspase-1, and cleaved caspase-1 in the lungs of hypoxic mice. Systemically, Hmox1-deficient mice showed increased basal levels of IL-18 that were further increased after 48 h of hypoxic exposure. Taken together, these finding point to a pivotal role for HO-1 in the control of baseline and hypoxic inflammasome signaling, perhaps through the antioxidant properties of bilirubin and CO's pleiotropic effects.


Subject(s)
Heme Oxygenase-1/metabolism , Hypoxia/metabolism , Inflammasomes/metabolism , Lung/metabolism , Membrane Proteins/metabolism , Animals , Caspase 1/metabolism , Inflammation/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/physiology
13.
Am J Respir Cell Mol Biol ; 61(4): 512-524, 2019 10.
Article in English | MEDLINE | ID: mdl-30951642

ABSTRACT

Inflammation and vascular smooth muscle cell (VSMC) phenotypic switching are causally linked to pulmonary arterial hypertension (PAH) pathogenesis. Carbonic anhydrase inhibition induces mild metabolic acidosis and exerts protective effects in hypoxic pulmonary hypertension. Carbonic anhydrases and metabolic acidosis are further known to modulate immune cell activation. To evaluate if carbonic anhydrase inhibition modulates macrophage activation, inflammation, and VSMC phenotypic switching in severe experimental pulmonary hypertension, pulmonary hypertension was assessed in Sugen 5416/hypoxia (SU/Hx) rats after treatment with acetazolamide or ammonium chloride (NH4Cl). We evaluated pulmonary and systemic inflammation and characterized the effect of carbonic anhydrase inhibition and metabolic acidosis in alveolar macrophages and bone marrow-derived macrophages (BMDMs). We further evaluated the treatment effects on VSMC phenotypic switching in pulmonary arteries and pulmonary artery smooth muscle cells (PASMCs) and corroborated some of our findings in lungs and pulmonary arteries of patients with PAH. Both patients with idiopathic PAH and SU/Hx rats had increased expression of lung inflammatory markers and signs of PASMC dedifferentiation in pulmonary arteries. Acetazolamide and NH4Cl ameliorated SU/Hx-induced pulmonary hypertension and blunted pulmonary and systemic inflammation. Expression of carbonic anhydrase isoform 2 was increased in alveolar macrophages from SU/Hx animals, classically (M1) and alternatively (M2) activated BMDMs, and lungs of patients with PAH. Carbonic anhydrase inhibition and acidosis had distinct effects on M1 and M2 markers in BMDMs. Inflammatory cytokines drove PASMC dedifferentiation, and this was inhibited by acetazolamide and acidosis. The protective antiinflammatory effect of acetazolamide in pulmonary hypertension is mediated by a dual mechanism of macrophage carbonic anhydrase inhibition and systemic metabolic acidosis.


Subject(s)
Acetazolamide/therapeutic use , Ammonium Chloride/therapeutic use , Carbonic Anhydrase Inhibitors/therapeutic use , Carbonic Anhydrases/physiology , Hypertension, Pulmonary/drug therapy , Acidosis/chemically induced , Acidosis/complications , Acidosis/immunology , Animals , Cell Differentiation/drug effects , Contractile Proteins/biosynthesis , Contractile Proteins/genetics , Drug Evaluation, Preclinical , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypoxia/complications , Inflammation , Macrophages/drug effects , Macrophages/enzymology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/enzymology , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Protein Isoforms/antagonists & inhibitors , Pulmonary Artery/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
14.
Am J Respir Crit Care Med ; 197(1): 104-116, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28853608

ABSTRACT

RATIONALE: Mesenchymal stem/stromal cell (MSC) therapies have shown promise in preclinical models of pathologies relevant to newborn medicine, such as bronchopulmonary dysplasia (BPD). We have reported that the therapeutic capacity of MSCs is comprised in their secretome, and demonstrated that the therapeutic vectors are exosomes produced by MSCs (MSC-exos). OBJECTIVES: To assess efficacy of MSC-exo treatment in a preclinical model of BPD and to investigate mechanisms underlying MSC-exo therapeutic action. METHODS: Exosomes were isolated from media conditioned by human MSC cultures. Newborn mice were exposed to hyperoxia (HYRX; 75% O2), treated with exosomes on Postnatal Day (PN) 4 and returned to room air on PN7. Treated animals and appropriate controls were harvested on PN7, -14, or -42 for assessment of pulmonary parameters. MEASUREMENTS AND MAIN RESULTS: HYRX-exposed mice presented with pronounced alveolar simplification, fibrosis, and pulmonary vascular remodeling, which was effectively ameliorated by MSC-exo treatment. Pulmonary function tests and assessment of pulmonary hypertension showed functional improvements after MSC-exo treatment. Lung mRNA sequencing demonstrated that MSC-exo treatment induced pleiotropic effects on gene expression associated with HYRX-induced inflammation and immune responses. MSC-exos modulate the macrophage phenotype fulcrum, suppressing the proinflammatory "M1" state and augmenting an antiinflammatory "M2-like" state, both in vitro and in vivo. CONCLUSIONS: MSC-exo treatment blunts HYRX-associated inflammation and alters the hyperoxic lung transcriptome. This results in alleviation of HYRX-induced BPD, improvement of lung function, decrease in fibrosis and pulmonary vascular remodeling, and amelioration of pulmonary hypertension. The MSC-exo mechanism of action is associated with modulation of lung macrophage phenotype.


Subject(s)
Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/therapy , Exosomes/transplantation , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Pulmonary Fibrosis/prevention & control , Animals , Animals, Newborn , Biopsy, Needle , Disease Models, Animal , Humans , Hyperoxia , Immunohistochemistry , Immunomodulation , Macrophages/immunology , Mice , Pulmonary Fibrosis/therapy , Random Allocation , Recovery of Function , Respiratory Function Tests , Sensitivity and Specificity , Treatment Outcome
15.
Annu Rev Physiol ; 77: 13-27, 2015.
Article in English | MEDLINE | ID: mdl-25293529

ABSTRACT

Mesenchymal stem cells (MSCs), whose mechanism of action is predominantly paracrine, are being widely tested for the treatment of a variety of human diseases. No one factor has been proven sufficient to mediate the therapeutic effects of MSCs. However, exosomes--membrane vesicles secreted by many cells, including MSCs--are appealing candidates as vectors of their efficacy. Exosomes can transport and deliver a large cargo of proteins, lipids, and nucleic acids and can modify cell and organ function. In addition to their key role as vehicles of intercellular communication, exosomes are increasingly recognized as biomarkers and prognosticators of disease. Moreover, they have the potential to be used as vehicles of gene and drug delivery for clinical application. This article reviews the biogenesis of exosomes, their molecular composition, and their role as messengers of intercellular communication, focusing on their potential as therapeutic vectors for stem cell therapy.


Subject(s)
Cell Communication/physiology , Cell- and Tissue-Based Therapy , Exosomes/physiology , Signal Transduction/physiology , Animals , Biomarkers , Drug Delivery Systems , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Regenerative Medicine
17.
J Pharmacol Exp Ther ; 364(2): 258-274, 2018 02.
Article in English | MEDLINE | ID: mdl-29212831

ABSTRACT

Pulmonary vasoreactivity could determine the responsiveness to vasodilators and, in turn, the prognosis of pulmonary hypertension (PH). We hypothesized that pulmonary vasoreactivity is impaired, and we examined the underlying mechanisms in the Sugen-hypoxia rat model of severe PH. Male Sprague-Dawley rats were injected with Sugen (20 mg/kg s.c.) and exposed to hypoxia (9% O2) for 3 weeks, followed by 4 weeks in normoxia (Su/Hx), or treated with Sugen alone (Su) or hypoxia alone (Hx) or neither (Nx). After hemodynamic measurements, the heart was assessed for right ventricular hypertrophy (Fulton's index); the pulmonary artery, aorta, and mesenteric arteries were isolated for vascular function studies; and contractile markers were measured in pulmonary arteries using quantitative polymerase chain reaction (PCR). Other rats were used for morphometric analysis of pulmonary vascular remodeling. Right ventricular systolic pressure and Fulton's index were higher in Su/Hx versus Su, Hx, and Nx rats. Pulmonary vascular remodeling was more prominent in Su/Hx versus Nx rats. In pulmonary artery rings, contraction to high KCl (96 mM) was less in Su/Hx versus Nx and Su, and phenylephrine-induced contraction was reduced in Su/Hx versus Nx, Hx, and Su. Acetylcholine (ACh)-induced relaxation was less in Su/Hx versus Nx and Hx, suggesting reduced endothelium-dependent vasodilation. ACh relaxation was inhibited by nitric oxide synthase (NOS) and guanylate cyclase blockade in all groups, suggesting a role of the NO-cGMP pathway. Nitrate/nitrite production in response to ACh was less in Su/Hx versus Nx, supporting reduced endothelial NO production. Sodium nitroprusside (10-8 M) caused less relaxation in Su/Hx versus Nx, Hx, and Su, suggesting a decreased responsiveness of vascular smooth muscle (VSM) to vasodilators. Neither contraction nor relaxation differed in the aorta or mesenteric arteries of all groups. PCR analysis showed decreased expression of contractile markers in pulmonary artery of Su/Hx versus Nx. The reduced responsiveness to vasoconstrictors and NO-mediated vasodilation in the pulmonary, but not systemic, vessels may be an underlying mechanism of severe PH in Su/Hx rats and appears to involve attenuation of the NO relaxation pathway and a switch of pulmonary VSM cells to a synthetic less reactive phenotype.


Subject(s)
Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Nitric Oxide/metabolism , Pulmonary Artery/physiopathology , Vasodilation , Animals , Cell Hypoxia , Cyclic GMP/metabolism , Disease Models, Animal , Hemodynamics , Hypertension, Pulmonary/metabolism , Male , Muscle, Smooth, Vascular/pathology , Phenotype , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Remodeling
18.
Pediatr Res ; 83(1-2): 298-307, 2018 01.
Article in English | MEDLINE | ID: mdl-28985201

ABSTRACT

Infants born at very low gestational age contribute disproportionately to neonatal morbidity and mortality. Advancements in antenatal steroid therapies and surfactant replacement have favored the survival of infants with ever-more immature lungs. Despite such advances in medical care, cardiopulmonary and neurological impairment prevail in constituting the major adverse outcomes for neonatal intensive care unit survivors. With no single effective therapy for either the prevention or treatment of such neonatal disorders, the need for new tools to treat and reduce risk of further complications associated with extreme preterm birth is urgent. Mesenchymal stem/stromal cell (MSC)-based approaches have shown promise in numerous experimental models of lung injury relevant to neonatology. Recent studies have highlighted that the therapeutic potential of MSCs is harnessed in their secretome, and that the therapeutic vector therein is represented by the exosomes released by MSCs. In this review, we summarize the development and significance of stem cell-based therapies for neonatal diseases, focusing on preclinical models of neonatal lung injury. We emphasize the development of MSC exosome-based therapeutics and comment on the challenges in bringing these promising interventions to clinic.


Subject(s)
Bronchopulmonary Dysplasia/therapy , Exosomes/metabolism , Infant, Newborn, Diseases/therapy , Lung Injury/therapy , Mesenchymal Stem Cells/cytology , Stem Cell Transplantation , Animals , Drug Delivery Systems , Humans , Hypoxia , Infant, Newborn , Infant, Premature , Inflammation , Intensive Care Units, Neonatal , Mice , Surface-Active Agents , Treatment Outcome
19.
Int J Mol Sci ; 19(9)2018 Aug 27.
Article in English | MEDLINE | ID: mdl-30150544

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by remodeling of the pulmonary arteries, increased pulmonary infiltrates, loss of vascular cross-sectional area, and elevated pulmonary vascular resistance. Despite recent advances in the management of PAH, there is a pressing need for the development of new tools to effectively treat and reduce the risk of further complications. Dysregulated immunity underlies the development of PAH, and macrophages orchestrate both the initiation and resolution of pulmonary inflammation, thus, manipulation of lung macrophage function represents an attractive target for emerging immunomodulatory therapies, including cell-based approaches. Indeed, mesenchymal stem cell (MSC)-based therapies have shown promise, effectively modulating the macrophage fulcrum to favor an anti-inflammatory, pro-resolving phenotype, which is associated with both histological and functional benefits in preclinical models of pulmonary hypertension (PH). The complex interplay between immune system homeostasis and MSCs remains incompletely understood. Here, we highlight the importance of macrophage function in models of PH and summarize the development of MSC-based therapies, focusing on the significance of MSC exosomes (MEx) and the immunomodulatory and homeostatic mechanisms by which such therapies may afford their beneficial effects.


Subject(s)
Cell-Derived Microparticles/immunology , Exosomes/immunology , Hypertension, Pulmonary/immunology , Immunomodulation/immunology , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Animals , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Exosomes/metabolism , Exosomes/transplantation , Humans , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Pulmonary Artery/immunology , Pulmonary Artery/physiopathology
20.
Biochim Biophys Acta ; 1843(11): 2528-42, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24983771

ABSTRACT

Complex organisms may coordinate molecular responses to hypoxia by specialized avenues of communication across multiple tissues, but these mechanisms are poorly understood. Plasma-based, extracellular microRNAs have been described, yet their regulation and biological functions in hypoxia remain enigmatic. We found a unique pattern of release of the hypoxia-inducible microRNA-210 (miR-210) from hypoxic and reoxygenated cells. This microRNA is also elevated in human plasma in physiologic and pathologic conditions of altered oxygen demand and delivery. Released miR-210 can be delivered to recipient cells, and the suppression of its direct target ISCU and mitochondrial metabolism is primarily evident in hypoxia. To regulate these hypoxia-specific actions, prolyl-hydroxylation of Argonaute 2 acts as a molecular switch that reciprocally modulates miR-210 release and intracellular activity in source cells as well as regulates intracellular activity in recipient cells after miR-210 delivery. Therefore, Argonaute 2-dependent control of released miR-210 represents a unique communication system that integrates the hypoxic response across anatomically distinct cells, preventing unnecessary activity of delivered miR-210 in normoxia while still preparing recipient tissues for incipient hypoxic stress and accelerating adaptation.

SELECTION OF CITATIONS
SEARCH DETAIL