Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Angeles Fernandez-Gonzalez is active.

Publication


Featured researches published by Angeles Fernandez-Gonzalez.


Circulation | 2012

Exosomes Mediate the Cytoprotective Action of Mesenchymal Stromal Cells on Hypoxia-Induced Pulmonary Hypertension

Changjin Lee; S. Alex Mitsialis; Muhammad Aslam; Sally H. Vitali; Eleni Vergadi; Georgios Konstantinou; Konstantinos Sdrimas; Angeles Fernandez-Gonzalez; Stella Kourembanas

Background— Hypoxia induces an inflammatory response in the lung manifested by alternative activation of macrophages with elevation of proinflammatory mediators that are critical for the later development of hypoxic pulmonary hypertension. Mesenchymal stromal cell transplantation inhibits lung inflammation, vascular remodeling, and right heart failure and reverses hypoxic pulmonary hypertension in experimental models of disease. In this study, we aimed to investigate the paracrine mechanisms by which mesenchymal stromal cells are protective in hypoxic pulmonary hypertension. Methods and Results— We fractionated mouse mesenchymal stromal cell–conditioned media to identify the biologically active component affecting in vivo hypoxic signaling and determined that exosomes, secreted membrane microvesicles, suppressed the hypoxic pulmonary influx of macrophages and the induction of proinflammatory and proproliferative mediators, including monocyte chemoattractant protein-1 and hypoxia-inducible mitogenic factor, in the murine model of hypoxic pulmonary hypertension. Intravenous delivery of mesenchymal stromal cell–derived exosomes (MEX) inhibited vascular remodeling and hypoxic pulmonary hypertension, whereas MEX-depleted media or fibroblast-derived exosomes had no effect. MEX suppressed the hypoxic activation of signal transducer and activator of transcription 3 (STAT3) and the upregulation of the miR-17 superfamily of microRNA clusters, whereas it increased lung levels of miR-204, a key microRNA, the expression of which is decreased in human pulmonary hypertension. MEX produced by human umbilical cord mesenchymal stromal cells inhibited STAT3 signaling in isolated human pulmonary artery endothelial cells, demonstrating a direct effect of MEX on hypoxic vascular cells. Conclusion— This study indicates that MEX exert a pleiotropic protective effect on the lung and inhibit pulmonary hypertension through suppression of hyperproliferative pathways, including STAT3-mediated signaling induced by hypoxia.


American Journal of Respiratory and Critical Care Medicine | 2009

Bone Marrow Stromal Cells Attenuate Lung Injury in a Murine Model of Neonatal Chronic Lung Disease

Muhammad Aslam; Rajiv Baveja; Olin D. Liang; Angeles Fernandez-Gonzalez; Changjin Lee; S. Alex Mitsialis; Stella Kourembanas

RATIONALE Neonatal chronic lung disease, known as bronchopulmonary dysplasia (BPD), remains a serious complication of prematurity despite advances in the treatment of extremely low birth weight infants. OBJECTIVES Given the reported protective actions of bone marrow stromal cells (BMSCs; mesenchymal stem cells) in models of lung and cardiovascular injury, we tested their therapeutic potential in a murine model of BPD. METHODS Neonatal mice exposed to hyperoxia (75% O(2)) were injected intravenously on Day 4 with either BMSCs or BMSC-conditioned media (CM) and assessed on Day 14 for lung morphometry, vascular changes associated with pulmonary hypertension, and lung cytokine profile. MEASUREMENTS AND MAIN RESULTS Injection of BMSCs but not pulmonary artery smooth muscle cells (PASMCs) reduced alveolar loss and lung inflammation, and prevented pulmonary hypertension. Although more donor BMSCs engrafted in hyperoxic lungs compared with normoxic controls, the overall low numbers suggest protective mechanisms other than direct tissue repair. Injection of BMSC-CM had a more pronounced effect than BMSCs, preventing both vessel remodeling and alveolar injury. Treated animals had normal alveolar numbers at Day 14 of hyperoxia and a drastically reduced lung neutrophil and macrophage accumulation compared with PASMC-CM-treated controls. Macrophage stimulating factor 1 and osteopontin, both present at high levels in BMSC-CM, may be involved in this immunomodulation. CONCLUSIONS BMSCs act in a paracrine manner via the release of immunomodulatory factors to ameliorate the parenchymal and vascular injury of BPD in vivo. Our study suggests that BMSCs and factor(s) they secrete offer new therapeutic approaches for lung diseases currently lacking effective treatment.


Circulation | 2011

Early Macrophage Recruitment and Alternative Activation Are Critical for the Later Development of Hypoxia-Induced Pulmonary Hypertension

Eleni Vergadi; Mun Seog Chang; Changjin Lee; Olin D. Liang; Xianlan Liu; Angeles Fernandez-Gonzalez; S. Alex Mitsialis; Stella Kourembanas

Background— Lung inflammation precedes the development of hypoxia-induced pulmonary hypertension (HPH); however, its role in the pathogenesis of HPH is poorly understood. We sought to characterize the hypoxic inflammatory response and to elucidate its role in the development of HPH. We also aimed to investigate the mechanisms by which heme oxygenase-1, an anti-inflammatory enzyme, is protective in HPH. Methods and Results— We generated bitransgenic mice that overexpress human heme oxygenase-1 under doxycycline control in an inducible, lung-specific manner. Hypoxic exposure of mice in the absence of doxycycline resulted in early transient accumulation of monocytes/macrophages in the bronchoalveolar lavage. Alveolar macrophages acquired an alternatively activated phenotype (M2) in response to hypoxia, characterized by the expression of found in inflammatory zone-1, arginase-1, and chitinase-3-like-3. A brief 2-day pulse of doxycycline delayed, but did not prevent, the peak of hypoxic inflammation, and could not protect against HPH. In contrast, a 7-day doxycycline treatment sustained high heme oxygenase-1 levels during the entire period of hypoxic inflammation, inhibited macrophage accumulation and activation, induced macrophage interleukin-10 expression, and prevented the development of HPH. Supernatants from hypoxic M2 macrophages promoted the proliferation of pulmonary artery smooth muscle cells, whereas treatment with carbon monoxide, a heme oxygenase-1 enzymatic product, abrogated this effect. Conclusions— Early recruitment and alternative activation of macrophages in hypoxic lungs are critical for the later development of HPH. Heme oxygenase-1 may confer protection from HPH by effectively modifying the macrophage activation state in hypoxia.


Pulmonary circulation | 2012

Mesenchymal stem cell-mediated reversal of bronchopulmonary dysplasia and associated pulmonary hypertension.

Georg Hansmann; Angeles Fernandez-Gonzalez; Muhammad Aslam; Sally H. Vitali; Thomas R. Martin; S. Alex Mitsialis; Stella Kourembanas

Clinical trials have failed to demonstrate an effective preventative or therapeutic strategy for bronchopulmonary dysplasia (BPD), a multifactorial chronic lung disease in preterm infants frequently complicated by pulmonary hypertension (PH). Mesenchymal stem cells (MSCs) and their secreted components have been shown to prevent BPD and pulmonary fibrosis in rodent models. We hypothesized that treatment with conditioned media (CM) from cultured mouse bone marrow-derived MSCs could reverse hyperoxia-induced BPD and PH. Newborn mice were exposed to hyperoxia (FiO2=0.75) for two weeks, were then treated with one intravenous dose of CM from either MSCs or primary mouse lung fibroblasts (MLFs), and placed in room air for two to four weeks. Histological analysis of lungs harvested at four weeks of age was performed to determine the degree of alveolar injury, blood vessel number, and vascular remodeling. At age six weeks, pulmonary artery pressure (PA acceleration time) and right ventricular hypertrophy (RVH; RV wall thickness) were assessed by echocardiography, and pulmonary function tests were conducted. When compared to MLF-CM, a single dose of MSC-CM-treatment (1) reversed the hyperoxia-induced parenchymal fibrosis and peripheral PA devascularization (pruning), (2) partially reversed alveolar injury, (3) normalized lung function (airway resistance, dynamic lung compliance), (4) fully reversed the moderate PH and RVH, and (5) attenuated peripheral PA muscularization associated with hyperoxia-induced BPD. Reversal of key features of hyperoxia-induced BPD and its long-term adverse effects on lung function can be achieved by a single intravenous dose of MSC-CM, thereby pointing toward a new therapeutic intervention for chronic lung diseases.


Stem Cells | 2011

Mesenchymal Stromal Cells Expressing Heme Oxygenase‐1 Reverse Pulmonary Hypertension

Olin D. Liang; S. Alex Mitsialis; Mun Seog Chang; Eleni Vergadi; Changjin Lee; Muhammad Aslam; Angeles Fernandez-Gonzalez; Xianlan Liu; Rajiv Baveja; Stella Kourembanas

Pulmonary arterial hypertension (PAH) remains a serious disease, and although current treatments may prolong and improve quality of life, search for novel and effective therapies is warranted. Using genetically modified mouse lines, we tested the ability of bone marrow‐derived stromal cells (mesenchymal stem cells [MSCs]) to treat chronic hypoxia‐induced PAH. Recipient mice were exposed for 5 weeks to normobaric hypoxia (8%–10% O2), MSC preparations were delivered through jugular vein injection and their effect on PAH was assessed after two additional weeks in hypoxia. Donor MSCs derived from wild‐type (WT) mice or heme oxygenase‐1 (HO‐1) null mice (Hmox1KO) conferred partial protection from PAH when transplanted into WT or Hmox1KO recipients, whereas treatment with MSCs isolated from transgenic mice harboring a human HO‐1 transgene under the control of surfactant protein C promoter (SH01 line) reversed established disease in WT recipients. SH01‐MSC treatment of Hmox1KO animals, which develop right ventricular (RV) infarction under prolonged hypoxia, resulted in normal RV systolic pressure, significant reduction of RV hypertrophy and prevention of RV infarction. Donor MSCs isolated from a bitransgenic mouse line with doxycycline‐inducible, lung‐specific expression of HO‐1 exhibited similar therapeutic efficacy only on doxycycline treatment of the recipients. In vitro experiments indicate that potential mechanisms of MSC action include modulation of hypoxia‐induced lung inflammation and inhibition of smooth muscle cell proliferation. Cumulatively, our results demonstrate that MSCs ameliorate chronic hypoxia‐induced PAH and their efficacy is highly augmented by lung‐specific HO‐1 expression in the transplanted cells, suggesting an interplay between HO‐1‐dependent and HO‐1‐independent protective pathways. STEM CELLS 2011;29:99–107


American Journal of Respiratory Cell and Molecular Biology | 2009

Mutation of Murine Adenylate Kinase 7 Underlies a Primary Ciliary Dyskinesia Phenotype

Angeles Fernandez-Gonzalez; Stella Kourembanas; Todd A. Wyatt; S. Alex Mitsialis

Primary ciliary dyskinesia (PCD) is a genetically and phenotypically heterogeneous disorder, characterized by progressive development of bronchiectasis, inflammation, and features characteristic of chronic obstructive pulmonary disease. We report here that a murine mutation of the evolutionarily conserved adenylate kinase 7 (Ak7) gene results in animals presenting with pathological signs characteristic of PCD, including ultrastructural ciliary defects and decreased ciliary beat frequency in respiratory epithelium. The mutation is associated with hydrocephalus, abnormal spermatogenesis, mucus accumulation in paranasal passages, and a dramatic respiratory pathology upon allergen challenge. Ak7 appears to be a marker for cilia with (9 + 2) microtubular organization. This is suggested by its tissue specificity of expression and also the stringent conservation of Ak7 ortholog structure only in protozoans and metazoans possessing motile (9 + 2) cilia. Collectively, our results indicate an ancestral and crucial role of Ak7 in maintaining ciliary structure and function, and suggest that mutations of the human ortholog may underlie a subset of genetically uncharacterized PCD cases.


European Journal of Neuroscience | 2000

MPTP selectively induces haem oxygenase‐1 expression in striatal astrocytes

Angeles Fernandez-Gonzalez; Isabel Pérez-Otaño; James I. Morgan

Parkinsons disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta with accompanying evidence of increased oxidative damage, deficits in mitochondrial function and iron deposition. Recently, haem oxygenase‐1 levels were reported to be elevated in PD brains. Because this enzyme is involved in the response to oxidative stress and is critical for cellular haem and iron homeostasis, it could play a role in the pathogenesis of PD. Therefore, we investigated the expression of haem oxygenase isoform 1 (HO‐1) in the 1‐methyl‐4‐phenyl‐1,2,3,6‐tetrahydropyridine (MPTP) mouse model of PD. MPTP triggered a relatively rapid and persistent increase in HO‐1 mRNA exclusively in the mouse striatum. In situ hybridization and immunohistochemistry showed HO‐1 to be localized to striatal astrocytes. The induction of HO‐1 by MPTP was blocked by selegiline and GBR‐12909, indicating the protoxin had to be metabolized by monoamine oxidase B and taken up by dopaminergic neurons to exert its action in astrocytes. MPTP did not alter the expression of other enzymes of haem synthesis or degradation nor were the levels of mRNA for haem or iron‐binding proteins changed. Thus, expression of HO‐1 was not part of a cellular program involving haem biosynthesis or homeostasis. In addition, heat shock proteins were not induced by MPTP. Thus, MPTP elicited a selective transcriptional response in striatal astrocytes. This response appears to be mediated by molecules released from affected dopaminergic nerve terminals in the striatum acting upon neighbouring astrocytes. This signalling pathway and its potential relevance to PD are discussed.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2012

Vasculoprotective effects of heme oxygenase-1 in a murine model of hyperoxia-induced bronchopulmonary dysplasia

Angeles Fernandez-Gonzalez; S. Alex Mitsialis; Xianlan Liu; Stella Kourembanas

Bronchopulmonary dysplasia (BPD) is characterized by simplified alveolarization and arrested vascular development of the lung with associated evidence of endothelial dysfunction, inflammation, increased oxidative damage, and iron deposition. Heme oxygenase-1 (HO-1) has been reported to be protective in the pathogenesis of diseases of inflammatory and oxidative etiology. Because HO-1 is involved in the response to oxidative stress produced by hyperoxia and is critical for cellular heme and iron homeostasis, it could play a protective role in BPD. Therefore, we investigated the effect of HO-1 in hyperoxia-induced lung injury using a neonatal transgenic mouse model with constitutive lung-specific HO-1 overexpression. Hyperoxia triggered an increase in pulmonary inflammation, arterial remodeling, and right ventricular hypertrophy that was attenuated by HO-1 overexpression. In addition, hyperoxia led to pulmonary edema, hemosiderosis, and a decrease in blood vessel number, all of which were markedly improved in HO-1 overexpressing mice. The protective vascular response may be mediated at least in part by carbon monoxide, due to its anti-inflammatory, antiproliferative, and antiapoptotic properties. HO-1 overexpression, however, did not prevent alveolar simplification nor altered the levels of ferritin and lactoferrin, proteins involved in iron binding and transport. Thus the protective mechanisms elicited by HO-1 overexpression primarily preserve vascular growth and barrier function through iron-independent, antioxidant, and anti-inflammatory pathways.


American Journal of Respiratory and Critical Care Medicine | 2018

Mesenchymal Stromal Cell Exosomes Ameliorate Experimental Bronchopulmonary Dysplasia and Restore Lung Function through Macrophage Immunomodulation

Gareth Willis; Angeles Fernandez-Gonzalez; Jamie Anastas; Sally H. Vitali; Xianlan Liu; Maria Ericsson; April Kwong; S. Alex Mitsialis; Stella Kourembanas

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.


Pulmonary circulation | 2014

The Sugen 5416/hypoxia mouse model of pulmonary hypertension revisited: long-term follow-up.

Sally H. Vitali; Georg Hansmann; Chase Rose; Angeles Fernandez-Gonzalez; Annette Scheid; S. Alex Mitsialis; Stella Kourembanas

The combination of a vascular endothelial growth factor receptor antagonist, Sugen 5416 (SU5416), and chronic hypoxia is known to cause pronounced pulmonary hypertension (PH) with angioobliterative lesions in rats and leads to exaggerated PH in mice as well. We sought to determine whether weekly SU5416 injections during 3 weeks of hypoxia leads to long-term development of angioobliterative lesions and sustained or progressive PH in mice. Male C57BL/6J mice were injected with SU5416 (SuHx) or vehicle (VehHx) weekly during 3 weeks of exposure to 10% oxygen. Echocardiographic and invasive measures of hemodynamics and pulmonary vascular morphometry were performed after the 3-week hypoxic exposure and after 10 weeks of recovery in normoxia. SuHx led to higher right ventricular (RV) systolic pressure and RV hypertrophy than VehHx after 3 weeks of hypoxia. Ten weeks after hypoxic exposure, RV systolic pressure decreased but remained elevated in SuHx mice compared with VehHx or normoxic control mice, but RV hypertrophy had resolved. After 3 weeks of hypoxia and 10 weeks of follow-up in normoxia, tricuspid annular plane systolic excursion was significantly decreased, indicating decreased systolic RV function. Very few angioobliterative lesions were found at the 10-week follow-up time point in SuHx mouse lungs. In conclusion, SU5416 combined with 3 weeks of hypoxia causes a more profound PH phenotype in mice than hypoxia alone. PH persists over 10 weeks of normoxic follow-up in SuHx mice, but significant angioobliterative lesions do not occur, and neither PH nor RV dysfunction worsens. The SuHx mouse model is a useful adjunct to other PH models, but the search will continue for a mouse model that better recapitulates the human phenotype.

Collaboration


Dive into the Angeles Fernandez-Gonzalez's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

S. Alex Mitsialis

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Sally H. Vitali

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Changjin Lee

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Muhammad Aslam

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Xianlan Liu

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Olin D. Liang

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Eleni Vergadi

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Helen Christou

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

James I. Morgan

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge