Network


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

Hotspot


Dive into the research topics where Ana Lucia Coelho is active.

Publication


Featured researches published by Ana Lucia Coelho.


Blood | 2009

Epigenetic regulation of the alternatively activated macrophage phenotype.

Haitao Wen; Callie Ann Sprunger Corsa; Tianju Liu; Ana Lucia Coelho; Ronald M. Allen; William F. Carson; Karen A. Cavassani; Xiangzhi Li; Nicholas W. Lukacs; Cory M. Hogaboam; Yali Dou; Steven L. Kunkel

Alternatively activated (M2) macrophages play critical roles in diverse chronic diseases, including parasite infections, cancer, and allergic responses. However, little is known about the acquisition and maintenance of their phenotype. We report that M2-macrophage marker genes are epigenetically regulated by reciprocal changes in histone H3 lysine-4 (H3K4) and histone H3 lysine-27 (H3K27) methylation; and the latter methylation marks are removed by the H3K27 demethylase Jumonji domain containing 3 (Jmjd3). We found that continuous interleukin-4 (IL-4) treatment leads to decreased H3K27 methylation, at the promoter of M2 marker genes, and a concomitant increase in Jmjd3 expression. Furthermore, we demonstrate that IL-4-dependent Jmjd3 expression is mediated by STAT6, a major transcription factor of IL-4-mediated signaling. After IL-4 stimulation, activated STAT6 is increased and binds to consensus sites at the Jmjd3 promoter. Increased Jmjd3 contributes to the decrease of H3K27 dimethylation and trimethylation (H3K27me2/3) marks as well as the transcriptional activation of specific M2 marker genes. The decrease in H3K27me2/3 and increase in Jmjd3 recruitment were confirmed by in vivo studies using a Schistosoma mansoni egg-challenged mouse model, a well-studied system known to support an M2 phenotype. Collectively, these data indicate that chromatin remodeling is mechanistically important in the acquisition of the M2-macrophage phenotype.


Science Translational Medicine | 2010

TLR9 Differentiates Rapidly from Slowly Progressing Forms of Idiopathic Pulmonary Fibrosis

Glenda Trujillo; Alessia Meneghin; Kevin R. Flaherty; Lynette M. Sholl; Jeffrey L. Myers; Ella A. Kazerooni; Barry H. Gross; Sameer R. Oak; Ana Lucia Coelho; Holly L. Evanoff; Elizabeth Day; Galen B. Toews; Amrita Joshi; Matthew Schaller; Beatrice Waters; Gabor Jarai; John Westwick; Steven L. Kunkel; Fernando J. Martinez; Cory M. Hogaboam

Compared to slow progressors, patients with rapidly progressive idiopathic pulmonary fibrosis express more TLR9, which recognizes unmethylated CpG DNA and stimulates the fibrotic process. Taking a Toll on Breathing Despite the incredible rate of advances being made in medical science, the exact causes of many diseases remain unknown. These diseases are classified as idiopathic—“a disease of its own kind.” But like a thief who leaves clues at a crime scene that disclose his or her identity, diseases can spur aberrant biological processes that hint at the condition’s cause. Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive form of lung disease of unknown origin characterized by the excess production of fibrous connective tissue (fibrosis) in the supporting framework (interstitium) of the lungs. These changes cause the hardening and/or scarring of lung tissue due to excess collagen, resulting in shortness of breath, a chronic dry cough, fatigue, weakness, chest discomfort, loss of appetite, and rapid weight loss. Patients with IPF have a poor prognosis and are usually expected to live only an average of 4 to 6 years after diagnosis; however, IPF displays a very heterogeneous path, with disease progressing rapidly in some patients and more slowly in others. Thus far, physicians have been unable to predict the speed of disease progression in patients newly diagnosed with IPF. Now, Trujillo et al. have identified a marker that differentiates these two patient groups and that may also mediate rapid progression of this disease. Toll-like receptor 9 (TLR9) is an innate immune molecule that recognizes a particular type of DNA frequently found in bacteria and viruses—unmethylated CpG DNA. Signaling through TLR9 promotes the differentiation of lung fibroblasts taken from IPF patients into myofibroblasts—cells that resemble both smooth muscle and fibroblasts—a key process in fibrosis. Trujillo et al. hypothesized that TLR9 may contribute to rapidly progressing IPF. Indeed, they found higher amounts of TLR9 in rapidly progressing IPF patients compared to slow progressing patients and normal controls. Moreover, in a xenograft mouse model of IPF, fibroblasts from rapid progressors induced more severe fibrosis in response to TLR9 activation than those from slow progressors. The presence of CpG also induced epithelial to mesenchymal transition—another hallmark of fibrosis—in a lung epithelial cell line in vitro. Together, these results suggest that TLR9 may serve as a marker for IPF rapid progressors and that TLR9 targeting may be a new therapeutic strategy for treating IPF. Thus, although the cause(s) of IPF remains unknown, the new data offer hope for an improvement in the prognosis and possibly treatment of this devastating disease. Idiopathic pulmonary fibrosis is characterized by diffuse alveolar damage and severe fibrosis, resulting in a steady worsening of lung function and gas exchange. Because idiopathic pulmonary fibrosis is a generally progressive disorder with highly heterogeneous disease progression, we classified affected patients as either rapid or slow progressors over the first year of follow-up and then identified differences between the two groups to investigate the mechanism governing rapid progression. Previous work from our laboratory has demonstrated that Toll-like receptor 9 (TLR9), a pathogen recognition receptor that recognizes unmethylated CpG motifs in bacterial and viral DNA, promotes myofibroblast differentiation in lung fibroblasts cultured from biopsies of patients with idiopathic pulmonary fibrosis. Therefore, we hypothesized that TLR9 functions as both a sensor of pathogenic molecules and a profibrotic signal in rapidly progressive idiopathic pulmonary fibrosis. Indeed, TLR9 was present at higher concentrations in surgical lung biopsies from rapidly progressive patients than in tissue from slowly progressing patients. Moreover, fibroblasts from rapid progressors were more responsive to the TLR9 agonist, CpG DNA, than were fibroblasts from slowly progressing patients. Using a humanized severe combined immunodeficient mouse, we then demonstrated increased fibrosis in murine lungs receiving human lung fibroblasts from rapid progressors compared with mice receiving fibroblasts from slowly progressing patients. This fibrosis was exacerbated by intranasal CpG challenges. Furthermore, CpG induced the differentiation of blood monocytes into fibrocytes and the epithelial-to-mesenchymal transition of A549 lung epithelial cells. These data suggest that TLR9 may drive the pathogenesis of rapidly progressive idiopathic pulmonary fibrosis and may serve as a potential indicator for this subset of the disease.


PLOS ONE | 2011

A micro RNA processing defect in rapidly progressing idiopathic pulmonary fibrosis

Sameer R. Oak; Lynne A. Murray; Athula Herath; Matthew A. Sleeman; Ian K. Anderson; Amrita Joshi; Ana Lucia Coelho; Kevin R. Flaherty; Galen B. Toews; Darryl A. Knight; Fernando J. Martinez; Cory M. Hogaboam

Background Idiopathic pulmonary fibrosis exhibits differential progression from the time of diagnosis but the molecular basis for varying progression rates is poorly understood. The aim of the present study was to ascertain whether differential miRNA expression might provide one explanation for rapidly versus slowly progressing forms of IPF. Methodology and Principal Findings miRNA and mRNA were isolated from surgical lung biopsies from IPF patients with a clinically documented rapid or slow course of disease over the first year after diagnosis. A quantitative PCR miRNA array containing 88 of the most abundant miRNA in the human genome was used to profile lung biopsies from 9 patients with rapidly progressing IPF, 6 patients with slowly progressing IPF, and 10 normal lung biopsies. Using this approach, 11 miRNA were significantly increased and 36 were significantly decreased in rapid biopsies compared with normal biopsies. Slowly progressive biopsies exhibited 4 significantly increased miRNA and 36 significantly decreased miRNA compared with normal lung. Among the miRNA present in IPF with validated mRNA targets were those with regulatory effects on epithelial-mesenchymal transition (EMT). Five miRNA (miR-302c, miR-423-5p, miR-210, miR-376c, and miR-185) were significantly increased in rapid compared with slow IPF lung biopsies. Additional analyses of rapid biopsies and fibroblasts grown from the same biopsies revealed that the expression of AGO1 and AGO2 (essential components of the miRNA processing RISC complex) were lower compared with either slow or normal lung biopsies and fibroblasts. Conclusion These findings suggest that the development and/or clinical progression of IPF might be the consequence of aberrant miRNA processing.


American Journal of Respiratory Cell and Molecular Biology | 2014

Targeting Interleukin-13 with Tralokinumab Attenuates Lung Fibrosis and Epithelial Damage in a Humanized SCID Idiopathic Pulmonary Fibrosis Model

Lynne A. Murray; Huilan Zhang; Sameer R. Oak; Ana Lucia Coelho; Athula Herath; Kevin R. Flaherty; Joyce S. Lee; Matthew J. Bell; Darryl A. Knight; Fernando J. Martinez; Matthew A. Sleeman; Erica L. Herzog; Cory M. Hogaboam

The aberrant fibrotic and repair responses in the lung are major hallmarks of idiopathic pulmonary fibrosis (IPF). Numerous antifibrotic strategies have been used in the clinic with limited success, raising the possibility that an effective therapeutic strategy in this disease must inhibit fibrosis and promote appropriate lung repair mechanisms. IL-13 represents an attractive target in IPF, but its disease association and mechanism of action remains unknown. In the present study, an overexpression of IL-13 and IL-13 pathway markers was associated with IPF, particularly a rapidly progressive form of this disease. Targeting IL-13 in a humanized experimental model of pulmonary fibrosis using tralokinumab (CAT354) was found to therapeutically block aberrant lung remodeling in this model. However, targeting IL-13 was also found to promote lung repair and to restore epithelial integrity. Thus, targeting IL-13 inhibits fibrotic processes and enhances repair processes in the lung.


The International Journal of Biochemistry & Cell Biology | 2012

Notch signaling mediates TGF-β1-induced epithelial-mesenchymal transition through the induction of Snai1.

Yosuke Matsuno; Ana Lucia Coelho; Gabor Jarai; John Westwick; Cory M. Hogaboam

Epithelial-mesenchymal transition (EMT) is a process by which epithelial cells undergo phenotypic transition to mesenchymal cells and thus is involved in the pathogenesis of tumor metastasis and organ fibrosis. Notch signaling is a highly conserved pathway that regulates intercellular communication and directs cell fate decisions. Here, we show the critical role of Notch signaling in TGF-β1-induced EMT. Inhibition of Notch signaling either by γ-secretase inhibitor or by knocking down of Notch signaling molecules by small interfering RNA abrogated EMT in association with decreased expression of Snai1. Constitutive activation of Notch signaling was sufficient for the induction of Snai1 as well as Notch ligand Jagged1. Notch signaling induced Snai1 expression via direct transcriptional activation. Collectively, these data show that Notch signaling activation promote TGF-β1-induced EMT through the induction of Snai1. Further studies on Notch signaling may provide diagnostic and therapeutic targets for cancer and fibrotic disease.


Journal of Leukocyte Biology | 2008

A systemic granulomatous response to Schistosoma mansoni eggs alters responsiveness of bone marrow-derived macrophages to Toll-like receptor agonists

Amrita Joshi; Tracy Raymond; Ana Lucia Coelho; Steven L. Kunkel; Cory M. Hogaboam

Macrophages play a pivotal role in innate and acquired immune responses to Schistosoma mansoni. Classical (M1) or alternative (M2) activation states of these cells further delineate their roles in tissue damage through innate immunity or fibrotic remodeling, respectively. In the present study, we addressed the following question: Does systemic Th2‐type cytokine polarization evoked by S. mansoni affect macrophage differentiation and activation? To this end, we analyzed bone marrow‐derived macrophages from mice with S. mansoni egg‐induced pulmonary granulomas and unchallenged (or naïve) mice to determine their activation state and their response to specific TLR agonists, including S. mansoni egg antigens. Unlike naïve macrophages, macrophages from Th2‐polarized mice constitutively expressed significantly higher “found in inflammatory zone‐1” (FIZZ1) and ST2 (M2 markers) and significantly lower NO synthase 2, CCL3, MIP‐2, TNF‐α, and IL‐12 (M1 markers). Also, compared with naïve macrophages, Th2‐polarized macrophages exhibited enhanced responses to the presence of specific TLR agonists, which consistently induced significantly higher levels of gene and protein levels for M2 and M1 markers in these cells. Together, these data show that signals received by bone marrow precursors during S. mansoni egg‐induced granuloma responses dynamically alter the development of macrophages and enhance the TLR responsiveness of these cells, which may ultimately have a significant effect on the pulmonary granulomatous response.


Journal of Immunology | 2007

The Chemokine CCL6 Promotes Innate Immunity via Immune Cell Activation and Recruitment

Ana Lucia Coelho; Matthew Schaller; Claudia F. Benjamim; Amos Orlofsky; Cory M. Hogaboam; Steven L. Kunkel

Septic syndrome is a consequence of innate immune failure. Recent studies showed that the CC chemokine CCL6 enhanced antimicrobial immunity during experimental sepsis through an unknown mechanism. The present study demonstrates that transgenic CCL6 expression abolishes mortality in a septic peritonitis model via the modulation of resident peritoneal cell activation and, more importantly, through the recruitment of IFN-producing NK cells and killer dendritic cells into the peritoneum. Thus, CCL6 attenuates the immune failure during sepsis, in part, through a protective type 1-cytokine mediated mechanism.


American Journal of Pathology | 2011

Targeting ST2L Potentiates CpG-Mediated Therapeutic Effects in a Chronic Fungal Asthma Model

Hemanth Ramaprakash; Takehiko Shibata; Karen E. Duffy; Ugur B. Ismailoglu; Rachel M. Bredernitz; Ana Paula Moreira; Ana Lucia Coelho; Anuk Das; Natalie Fursov; Geoffrey L. Chupp; Cory M. Hogaboam

IL-33 and its soluble receptor and cell-associated receptor (ST2L) are all increased in clinical and experimental asthma. The present study addressed the hypothesis that ST2L impairs the therapeutic effects of CpG in a fungal model of asthma. C57BL/6 mice were sensitized to Aspergillus fumigatus and challenged via i.t. instillation with live A. fumigatus conidia. Mice were treated with IgG alone, anti-ST2L monoclonal antibody (mAb) alone, CpG alone, IgG plus CpG, or anti-ST2L mAb plus CpG every other day from day 14 to day 28 and investigated on day 28 after conidia. Lung ST2L and toll-like receptor 9 protein expression levels concomitantly increased in a time-dependent manner during fungal asthma. Therapeutic blockade of ST2L with an mAb attenuated key pathological features of this model. At subtherapeutic doses, neither anti-ST2L mAb nor CpG alone affected fungal asthma severity. However, airway hyperresponsiveness, mucus cell metaplasia, peribronchial fibrosis, and fungus retention were markedly reduced in asthmatic mice treated with the combination of both. Whole lung CXCL9 levels were significantly elevated in the combination group but not in the controls. Furthermore, in asthmatic mice treated with the combination therapy, dendritic cells generated significantly greater IL-12p70 with CpG in vitro compared with control dendritic cells. The combination of anti-ST2L mAb with CpG significantly attenuated experimental asthma, suggesting that targeting ST2L might enhance the therapeutic efficacy of CpG during allergic inflammation.


Journal of Immunology | 2014

Axl Receptor Blockade Ameliorates Pulmonary Pathology Resulting from Primary Viral Infection and Viral Exacerbation of Asthma

Takehiko Shibata; David M. Habiel; Ana Lucia Coelho; Steven L. Kunkel; Nicholas W. Lukacs; Cory M. Hogaboam

Viruses use Tyro3, Axl, and Mertk (TAM) receptor tyrosine kinases to infect and modulate the immune properties of various cell types, which led us to investigate whether TAM receptor activation affected primary viral infection and viral exacerbation of asthma in experimental models. In these lung-specific models, we observed that Axl was the most abundantly induced TAM receptor protein. During primary respiratory syncytial virus (RSV) infection, anti-Axl mAb treatment significantly increased the number of IFN-γ–producing T cells and NK cells and significantly suppressed RSV replication and whole lung levels of IL-4 and IL-13. Intrapulmonary H1N1 infection induced lethal pulmonary inflammation, but anti-Axl mAb treatment of infected mice significantly increased the number of IFN-β–producing macrophages and dendritic cells and significantly suppressed neutrophil infiltration. Consequently, the lethal effect of H1N1 infection in this model was significantly reduced in the mAb-treated group compared with the IgG control-treated group. Targeting Axl also inhibited airway hyperresponsiveness, IL-4 and IL-13 production, and goblet cell metaplasia in an Aspergillus fumigatus–induced asthma model. Finally, infection of mice with RSV during fungal asthma significantly exacerbated airway inflammation, goblet cell metaplasia, and airway remodeling, but all of these features in this viral exacerbation model were ameliorated by anti-Axl mAb treatment. Taken together, these results demonstrate that Axl modulates the pulmonary immune response during viral and/or allergic pathology, and they also suggest that targeting this TAM receptor might provide a novel therapeutic approach in these infectious diseases.


PLOS ONE | 2013

Toll like receptor 3 plays a critical role in the progression and severity of acetaminophen-induced hepatotoxicity.

Karen A. Cavassani; Ana Paula Moreira; David M. Habiel; Toshihiro Ito; Ana Lucia Coelho; Ronald M. Allen; Bin Hu; Janna Raphelson; William F. Carson; Matthew Schaller; Nicholas W. Lukacs; M. Bishr Omary; Cory M. Hogaboam; Steven L. Kunkel

Toll-like receptor (TLR) activation has been implicated in acetaminophen (APAP)-induced hepatotoxicity. Herein, we hypothesize that TLR3 activation significantly contributed to APAP-induced liver injury. In fasted wildtype (WT) mice, APAP caused significant cellular necrosis, edema, and inflammation in the liver, and the de novo expression and activation of TLR3 was found to be necessary for APAP-induced liver failure. Specifically, liver tissues from similarly fasted TLR3-deficient (tlr3−/−) mice exhibited significantly less histological and biochemical evidence of injury after APAP challenge. Similar protective effects were observed in WT mice in which TLR3 was targeted through immunoneutralization at 3 h post-APAP challenge. Among three important death ligands (i.e. TNFα, TRAIL, and FASL) known to promote hepatocyte death after APAP challenge, TNFα was the only ligand that was significantly reduced in APAP-challenged tlr3−/− mice compared with APAP-challenged WT controls. In vivo studies demonstrated that TLR3 activation contributed to TNFα production in the liver presumably via F4/80+ and CD11c+ immune cells. In vitro studies indicated that there was cooperation between TNFα and TLR3 in the activation of JNK signaling in isolated and cultured liver epithelial cells (i.e. nMuLi). Moreover, TLR3 activation enhanced the expression of phosphorylated JNK in APAP injured livers. Thus, the current study demonstrates that TLR3 activation contributes to APAP-induced hepatotoxicity.

Collaboration


Dive into the Ana Lucia Coelho's collaboration.

Top Co-Authors

Avatar

Cory M. Hogaboam

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

David M. Habiel

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge