Network


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

Hotspot


Dive into the research topics where Maria E. Laucho-Contreras is active.

Publication


Featured researches published by Maria E. Laucho-Contreras.


Nature Medicine | 2016

Mitochondrial iron chelation ameliorates cigarette smoke–induced bronchitis and emphysema in mice

Suzanne M. Cloonan; Kimberly Glass; Maria E. Laucho-Contreras; Abhiram R. Bhashyam; Morgan Cervo; Maria A. Pabon; Csaba Konràd; Francesca Polverino; Ilias I. Siempos; Elizabeth Perez; Kenji Mizumura; Manik C. Ghosh; Harikrishnan Parameswaran; Niamh C Williams; Kristen T Rooney; Zhihua Chen; Monica P. Goldklang; Guo-Cheng Yuan; Stephen C. Moore; Dawn L. DeMeo; Tracey A. Rouault; Jeanine D'Armiento; Eric A. Schon; Giovanni Manfredi; John Quackenbush; Ashfaq Mahmood; Edwin K. Silverman; Caroline A. Owen; Augustine M. K. Choi

Chronic obstructive pulmonary disease (COPD) is linked to both cigarette smoking and genetic determinants. We have previously identified iron-responsive element–binding protein 2 (IRP2) as an important COPD susceptibility gene and have shown that IRP2 protein is increased in the lungs of individuals with COPD. Here we demonstrate that mice deficient in Irp2 were protected from cigarette smoke (CS)-induced experimental COPD. By integrating RNA immunoprecipitation followed by sequencing (RIP-seq), RNA sequencing (RNA-seq), and gene expression and functional enrichment clustering analysis, we identified Irp2 as a regulator of mitochondrial function in the lungs of mice. Irp2 increased mitochondrial iron loading and levels of cytochrome c oxidase (COX), which led to mitochondrial dysfunction and subsequent experimental COPD. Frataxin-deficient mice, which had higher mitochondrial iron loading, showed impaired airway mucociliary clearance (MCC) and higher pulmonary inflammation at baseline, whereas mice deficient in the synthesis of cytochrome c oxidase, which have reduced COX, were protected from CS-induced pulmonary inflammation and impairment of MCC. Mice treated with a mitochondrial iron chelator or mice fed a low-iron diet were protected from CS-induced COPD. Mitochondrial iron chelation also alleviated CS-induced impairment of MCC, CS-induced pulmonary inflammation and CS-associated lung injury in mice with established COPD, suggesting a critical functional role and potential therapeutic intervention for the mitochondrial-iron axis in COPD.


American Journal of Respiratory and Critical Care Medicine | 2016

A Chronic Obstructive Pulmonary Disease Susceptibility Gene, FAM13A, Regulates Protein Stability of β-Catenin

Zhiqiang Jiang; Taotao Lao; Weiliang Qiu; Francesca Polverino; Kushagra Gupta; Feng Guo; John D. Mancini; Zun Zar Chi Naing; Michael H. Cho; Peter J. Castaldi; Yang Sun; Jane Yu; Maria E. Laucho-Contreras; Lester Kobzik; Benjamin A. Raby; Augustine M. K. Choi; Mark A. Perrella; Caroline A. Owen; Edwin K. Silverman; Xiaobo Zhou

RATIONALE A genetic locus within the FAM13A gene has been consistently associated with chronic obstructive pulmonary disease (COPD) in genome-wide association studies. However, the mechanisms by which FAM13A contributes to COPD susceptibility are unknown. OBJECTIVES To determine the biologic function of FAM13A in human COPD and murine COPD models and discover the molecular mechanism by which FAM13A influences COPD susceptibility. METHODS Fam13a null mice (Fam13a(-/-)) were generated and exposed to cigarette smoke. The lung inflammatory response and airspace size were assessed in Fam13a(-/-) and Fam13a(+/+) littermate control mice. Cellular localization of FAM13A protein and mRNA levels of FAM13A in COPD lungs were assessed using immunofluorescence, Western blotting, and reverse transcriptase-polymerase chain reaction, respectively. Immunoprecipitation followed by mass spectrometry identified cellular proteins that interact with FAM13A to reveal insights on FAM13As function. MEASUREMENTS AND MAIN RESULTS In murine and human lungs, FAM13A is expressed in airway and alveolar type II epithelial cells and macrophages. Fam13a null mice (Fam13a(-/-)) were resistant to chronic cigarette smoke-induced emphysema compared with Fam13a(+/+) mice. In vitro, FAM13A interacts with protein phosphatase 2A and recruits protein phosphatase 2A with glycogen synthase kinase 3β and β-catenin, inducing β-catenin degradation. Fam13a(-/-) mice were also resistant to elastase-induced emphysema, and this resistance was reversed by coadministration of a β-catenin inhibitor, suggesting that FAM13A could increase the susceptibility of mice to emphysema development by inhibiting β-catenin signaling. Moreover, human COPD lungs had decreased protein levels of β-catenin and increased protein levels of FAM13A. CONCLUSIONS We show that FAM13A may influence COPD susceptibility by promoting β-catenin degradation.


European Respiratory Journal | 2015

Protective role for club cell secretory protein-16 (CC16) in the development of COPD.

Maria E. Laucho-Contreras; Francesca Polverino; Kushagra Gupta; Taylor Kl; Emer Kelly; Pinto-Plata; Miguel Divo; Ashfaq N; Hans Petersen; Stripp B; Pilon Al; Yohannes Tesfaigzi; Bartolome R. Celli; Caroline A. Owen

Club cell secretory protein-16 (CC16) is the major secreted product of airway club cells, but its role in the pathogenesis of chronic obstructive pulmonary disease (COPD) is unclear. We measured CC16 airway expression in humans with and without COPD and CC16 function in a cigarette smoke (CS)-induced COPD murine model. Airway CC16 expression was measured in COPD patients, smokers without COPD and non-smokers. We exposed wildtype (WT) and CC16−/−mice to CS or air for up to 6 months, and measured airway CC16 expression, pulmonary inflammation, alveolar septal cell apoptosis, airspace enlargement, airway mucin 5AC (MUC5AC) expression, small airway remodelling and pulmonary function. Smokers and COPD patients had reduced airway CC16 immunostaining that decreased with increasing COPD severity. Exposing mice to CS reduced airway CC16 expression. CC16−/− mice had greater CS-induced emphysema, airway remodelling, pulmonary inflammation, alveolar cell apoptosis, airway MUC5AC expression, and more compliant lungs than WT mice. These changes were associated with increased nuclear factor-κB (NF-κB) activation in CC16−/− lungs. CS-induced acute pulmonary changes were reversed by adenoviral-mediated over-expression of CC16. CC16 protects lungs from CS-induced injury by reducing lung NF-κB activation. CS-induced airway CC16 deficiency increases CS-induced pulmonary inflammation and injury and likely contributes to the pathogenesis of COPD. Cigarette smoke exposure reduces airway levels of anti-inflammatory CC16 to thereby contribute to the genesis of COPD http://ow.ly/GOMiZ


American Journal of Respiratory and Critical Care Medicine | 2015

B Cell–Activating Factor. An Orchestrator of Lymphoid Follicles in Severe Chronic Obstructive Pulmonary Disease

Francesca Polverino; Borja G. Cosío; Jaime Pons; Maria E. Laucho-Contreras; Paula Tejera; Amanda Iglesias; Angel Rios; Andreas Jahn; Jaume Sauleda; Miguel Divo; Victor Pinto-Plata; Lynette M. Sholl; Ivan O. Rosas; Alvar Agusti; Bartolome R. Celli; Caroline A. Owen

RATIONALE Patients with chronic obstructive pulmonary disease (COPD) have increased pulmonary lymphoid follicle (LF) counts. B cell-activating factor of tumor necrosis factor family (BAFF) regulates B cells in health, but its role in COPD pathogenesis is unclear. OBJECTIVES To determine whether BAFF expression in pulmonary LFs correlates with COPD severity, LF size or number, and/or readouts of B-cell function in LFs. METHODS We correlated BAFF immunostaining in LFs in lung explants or biopsies from nonsmoking control subjects (NSC), smokers without COPD (SC), and patients with COPD with the number and size of LFs, and LF B-cell apoptosis, activation, and proliferation. We analyzed serum BAFF levels and BAFF expression in B cells in blood and bronchoalveolar lavage samples from the same subject groups. We assessed whether: (1) cigarette smoke extract (CSE) increases B-cell BAFF expression and (2) recombinant BAFF (rBAFF) rescues B cells from CSE-induced apoptosis by inhibiting activation of nuclear factor-κB (NF-κB). MEASUREMENTS AND MAIN RESULTS Patients with Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage IV COPD had increased numbers and larger pulmonary LFs than patients with GOLD stages I-II COPD and SC. We identified two main types of pulmonary LFs: (1) type A, the predominant type in GOLD stages I-II COPD and SC, characterized by abundant apoptotic but few BAFF-positive cells (mostly B cells); and (2) type B, the main type in GOLD stage IV COPD, characterized by abundant BAFF-positive cells but few apoptotic cells (mostly B cells). BAFF levels were also higher in blood and bronchoalveolar lavage B cells in patients with COPD versus NSC and SC. Surprisingly, rBAFF blocked CSE-induced B-cell apoptosis by inhibiting CSE-induced NF-κB activation. CONCLUSIONS Our data support the hypothesis that B-cell BAFF expression creates a self-perpetuating loop contributing to COPD progression by promoting pulmonary B-cell survival and LF expansion.


American Journal of Respiratory and Critical Care Medicine | 2017

A Pilot Study Linking Endothelial Injury in Lungs and Kidneys in Chronic Obstructive Pulmonary Disease

Francesca Polverino; Maria E. Laucho-Contreras; Hans Petersen; Vanesa Bijol; Lynette M. Sholl; Mary E. Choi; Miguel Divo; Victor Pinto-Plata; Alfredo Chetta; Yohannes Tesfaigzi; Bartolome R. Celli; Caroline A. Owen

Rationale: Patients with chronic obstructive pulmonary disease (COPD) frequently have albuminuria (indicative of renal endothelial cell injury) associated with hypoxemia. Objectives: To determine whether (1) cigarette smoke (CS)‐induced pulmonary and renal endothelial cell injury explains the association between albuminuria and COPD, (2) CS‐induced albuminuria is linked to increases in the oxidative stress‐advanced glycation end products (AGEs) receptor for AGEs (RAGE) pathway, and (3) enalapril (which has antioxidant properties) limits the progression of pulmonary and renal injury by reducing activation of the AGEs‐RAGE pathway in endothelial cells in both organs. Methods: In 26 patients with COPD, 24 ever‐smokers without COPD, 32 nonsmokers who underwent a renal biopsy or nephrectomy, and in CS‐exposed mice, we assessed pathologic and ultrastructural renal lesions, and measured urinary albumin/creatinine ratios, tissue oxidative stress levels, and AGEs and RAGE levels in pulmonary and renal endothelial cells. The efficacy of enalapril on pulmonary and renal lesions was assessed in CS‐exposed mice. Measurements and Main Results: Patients with COPD and/or CS‐exposed mice had chronic renal injury, increased urinary albumin/creatinine ratios, and increased tissue oxidative stress and AGEs‐RAGE levels in pulmonary and renal endothelial cells. Treating mice with enalapril attenuated CS‐induced increases in urinary albumin/creatinine ratios, tissue oxidative stress levels, endothelial cell AGEs and RAGE levels, pulmonary and renal cell apoptosis, and the progression of chronic renal and pulmonary lesions. Conclusions: Patients with COPD and/or CS‐exposed mice have pulmonary and renal endothelial cell injury linked to increased endothelial cell AGEs and RAGE levels. Albuminuria could identify patients with COPD in whom angiotensin‐converting enzyme inhibitor therapy improves renal and lung function by reducing endothelial injury.


Journal of Immunology | 2014

ADAM9 Is a Novel Product of Polymorphonuclear Neutrophils: Regulation of Expression and Contributions to Extracellular Matrix Protein Degradation during Acute Lung Injury

Robin Roychaudhuri; Anja Hergrueter; Francesca Polverino; Maria E. Laucho-Contreras; Kushagra Gupta; Niels Borregaard; Caroline A. Owen

A disintegrin and a metalloproteinase domain (ADAM) 9 is known to be expressed by monocytes and macrophages. In this study, we report that ADAM9 is also a product of human and murine polymorphonuclear neutrophils (PMNs). ADAM9 is not synthesized de novo by circulating PMNs. Rather, ADAM9 protein is stored in the gelatinase and specific granules and the secretory vesicles of human PMNs. Unstimulated PMNs express minimal quantities of surface ADAM9, but activation of PMNs with degranulating agonists rapidly (within 15 min) increases PMN surface ADAM9 levels. Human PMNs produce small quantities of soluble forms of ADAM9. Surprisingly, ADAM9 degrades several extracellular matrix (ECM) proteins, including fibronectin, entactin, laminin, and insoluble elastin, as potently as matrix metalloproteinase-9. However, ADAM9 does not degrade types I, III, or IV collagen or denatured collagens in vitro. To determine whether Adam9 regulates PMN recruitment or ECM protein turnover during inflammatory responses, we compared wild-type and Adam9−/− mice in bacterial LPS- and bleomycin-mediated acute lung injury (ALI). Adam9 lung levels increase 10-fold during LPS-mediated ALI in wild-type mice (due to increases in leukocyte-derived Adam9), but Adam9 does not regulate lung PMN (or macrophage) counts during ALI. Adam9 increases mortality, promotes lung injury, reduces lung compliance, and increases degradation of lung elastin during LPS- and/or bleomycin-mediated ALI. Adam9 does not regulate collagen accumulation in the bleomycin-treated lung. Thus, ADAM9 is expressed in an inducible fashion on PMN surfaces where it degrades some ECM proteins, and it promotes alveolar–capillary barrier injury during ALI in mice.


American Journal of Pathology | 2015

A novel nonhuman primate model of cigarette smoke-induced airway disease.

Francesca Polverino; Melanie Doyle-Eisele; Jacob D. McDonald; Julie A. Wilder; Christopher Royer; Maria E. Laucho-Contreras; Emer Kelly; Miguel Divo; Victor Pinto-Plata; Joe L. Mauderly; Bartolome R. Celli; Yohannes Tesfaigzi; Caroline A. Owen

Small animal models of chronic obstructive pulmonary disease (COPD) have several limitations for identifying new therapeutic targets and biomarkers for human COPD. These include a pulmonary anatomy that differs from humans, the limited airway pathologies and lymphoid aggregates that develop in smoke-exposed mice, and the challenges associated with serial biological sampling. Thus, we assessed the utility of cigarette smoke (CS)-exposed cynomolgus macaque as a nonhuman primate (NHP) large animal model of COPD. Twenty-eight NHPs were exposed to air or CS 5 days per week for up to 12 weeks. Bronchoalveolar lavage and pulmonary function tests were performed at intervals. After 12 weeks, we measured airway pathologies, pulmonary inflammation, and airspace enlargement. CS-exposed NHPs developed robust mucus metaplasia, submucosal gland hypertrophy and hyperplasia, airway inflammation, peribronchial fibrosis, and increases in bronchial lymphoid aggregates. Although CS-exposed NHPs did not develop emphysema over the study time, they exhibited pathologies that precede emphysema development, including increases in the following: i) matrix metalloproteinase-9 and proinflammatory mediator levels in bronchoalveolar lavage fluid, ii) lung parenchymal leukocyte counts and lymphoid aggregates, iii) lung oxidative stress levels, and iv) alveolar septal cell apoptosis. CS-exposed NHPs can be used as a model of airway disease occurring in COPD patients. Unlike rodents, NHPs can safely undergo longitudinal sampling, which could be useful for assessing novel biomarkers or therapeutics for COPD.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Hhip haploinsufficiency sensitizes mice to age-related emphysema

Taotao Lao; Zhiqiang Jiang; Jeong Yun; Weiliang Qiu; Feng Guo; Chunfang Huang; John D. Mancini; Kushagra Gupta; Maria E. Laucho-Contreras; Zun Zar Chi Naing; Li Zhang; Mark A. Perrella; Caroline A. Owen; Edwin K. Silverman; Xiaobo Zhou

Significance Genome-wide association studies (GWAS) have been very successful in discovering genetic loci associated with complex traits. However, only few studies applied murine models to investigate how GWAS genes contribute to human lung diseases. Motivated by GWAS linking Hedgehog interacting protein (HHIP) to emphysema and impairments in lung function, this study demonstrated that Hhip+/− mice developed spontaneous emphysema and lung function impairment over time. Moreover, emphysema, associated with increased oxidative stress in Hhip+/− lungs, was prevented by treating the mice with the antioxidant, N-acetyl cysteine (NAC). This post-GWAS functional study connects aging-related diseases, molecular mechanisms, and potential therapy in a genetic haploinsufficient murine model, which may lead to improvements in understanding pathophysiologic concepts of alveolar loss related to aging. Genetic variants in Hedgehog interacting protein (HHIP) have consistently been associated with the susceptibility to develop chronic obstructive pulmonary disease and pulmonary function levels, including the forced expiratory volume in 1 s (FEV1), in general population samples by genome-wide association studies. However, in vivo evidence connecting Hhip to age-related FEV1 decline and emphysema development is lacking. Herein, using Hhip heterozygous mice (Hhip+/−), we observed increased lung compliance and spontaneous emphysema in Hhip+/− mice starting at 10 mo of age. This increase was preceded by increases in oxidative stress levels in the lungs of Hhip+/− vs. Hhip+/+ mice. To our knowledge, these results provide the first line of evidence that HHIP is involved in maintaining normal lung function and alveolar structures. Interestingly, antioxidant N-acetyl cysteine treatment in mice starting at age of 5 mo improved lung function and prevented emphysema development in Hhip+/− mice, suggesting that N-acetyl cysteine treatment limits the progression of age-related emphysema in Hhip+/− mice. Therefore, reduced lung function and age-related spontaneous emphysema development in Hhip+/− mice may be caused by increased oxidative stress levels in murine lungs as a result of haploinsufficiency of Hhip.


PLOS ONE | 2014

Mononuclear Phagocytes and Airway Epithelial Cells: Novel Sources of Matrix Metalloproteinase-8 (MMP-8) in Patients with Idiopathic Pulmonary Fibrosis

Vanessa J. Craig; Francesca Polverino; Maria E. Laucho-Contreras; Yuanyuan Shi; Yushi Liu; Juan C. Osorio; Yohannes Tesfaigzi; Victor Pinto-Plata; Bernadette R. Gochuico; Ivan O. Rosas; Caroline A. Owen

Objectives Matrix metalloproteinase-8 (MMP-8) promotes lung fibrotic responses to bleomycin in mice. Although prior studies reported that MMP-8 levels are increased in plasma and bronchoalveolar lavage fluid (BALF) samples from IPF patients, neither the bioactive forms nor the cellular sources of MMP-8 in idiopathic pulmonary fibrosis (IPF) patients have been identified. It is not known whether MMP-8 expression is dys-regulated in IPF leukocytes or whether MMP-8 plasma levels correlate with IPF outcomes. Our goal was to address these knowledge gaps. Methods We measured MMP-8 levels and forms in blood and lung samples from IPF patients versus controls using ELISAs, western blotting, and qPCR, and assessed whether MMP-8 plasma levels in 73 IPF patients correlate with rate of lung function decline and mortality. We used immunostaining to localize MMP-8 expression in IPF lungs. We quantified MMP-8 levels and forms in blood leukocytes from IPF patients versus controls. Results IPF patients have increased BALF, whole lung, and plasma levels of soluble MMP-8 protein. Active MMP-8 is the main form elevated in IPF lungs. MMP-8 mRNA levels are increased in monocytes from IPF patients, but IPF patients and controls have similar levels of MMP-8 in PMNs. Surprisingly, macrophages and airway epithelial cells are the main cells expressing MMP-8 in IPF lungs. Plasma and BALF MMP-8 levels do not correlate with decline in lung function and/or mortality in IPF patients. Conclusion Blood and lung MMP-8 levels are increased in IPF patients. Active MMP-8 is the main form elevated in IPF lungs. Surprisingly, blood monocytes, lung macrophages, and airway epithelial cells are the main cells in which MMP-8 is upregulated in IPF patients. Plasma and BALF MMP-8 levels are unlikely to serve as a prognostic biomarker for IPF patients. These results provide new information about the expression patterns of MMP-8 in IPF patients.


Journal of Visualized Experiments | 2015

Automated measurement of pulmonary emphysema and small airway remodeling in cigarette smoke-exposed mice.

Maria E. Laucho-Contreras; Katherine L. Taylor; Ravi Mahadeva; Steve Boukedes; Caroline A. Owen

COPD is projected to be the third most common cause of mortality world-wide by 2020((1)). Animal models of COPD are used to identify molecules that contribute to the disease process and to test the efficacy of novel therapies for COPD. Researchers use a number of models of COPD employing different species including rodents, guinea-pigs, rabbits, and dogs((2)). However, the most widely-used model is that in which mice are exposed to cigarette smoke. Mice are an especially useful species in which to model COPD because their genome can readily be manipulated to generate animals that are either deficient in, or over-express individual proteins. Studies of gene-targeted mice that have been exposed to cigarette smoke have provided valuable information about the contributions of individual molecules to different lung pathologies in COPD((3-5)). Most studies have focused on pathways involved in emphysema development which contributes to the airflow obstruction that is characteristic of COPD. However, small airway fibrosis also contributes significantly to airflow obstruction in human COPD patients((6)), but much less is known about the pathogenesis of this lesion in smoke-exposed animals. To address this knowledge gap, this protocol quantifies both emphysema development and small airway fibrosis in smoke-exposed mice. This protocol exposes mice to CS using a whole-body exposure technique, then measures respiratory mechanics in the mice, inflates the lungs of mice to a standard pressure, and fixes the lungs in formalin. The researcher then stains the lung sections with either Gills stain to measure the mean alveolar chord length (as a readout of emphysema severity) or Massons trichrome stain to measure deposition of extracellular matrix (ECM) proteins around small airways (as a readout of small airway fibrosis). Studies of the effects of molecular pathways on both of these lung pathologies will lead to a better understanding of the pathogenesis of COPD.

Collaboration


Dive into the Maria E. Laucho-Contreras's collaboration.

Top Co-Authors

Avatar

Caroline A. Owen

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Francesca Polverino

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Bartolome R. Celli

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Miguel Divo

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yohannes Tesfaigzi

Lovelace Respiratory Research Institute

View shared research outputs
Top Co-Authors

Avatar

Kushagra Gupta

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lynette M. Sholl

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Edwin K. Silverman

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge