Network


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

Hotspot


Dive into the research topics where Angela J. Rogers is active.

Publication


Featured researches published by Angela J. Rogers.


American Journal of Respiratory and Critical Care Medicine | 2012

Inflammasome-regulated Cytokines Are Critical Mediators of Acute Lung Injury

Tamas Dolinay; Young Sam Kim; Judie A. Howrylak; Gary M. Hunninghake; Chang Hyeok An; Anthony F. Massaro; Angela J. Rogers; Lee Gazourian; Kiichi Nakahira; Jeffrey A. Haspel; Roberto Landazury; Sabitha Eppanapally; Jason D. Christie; Nuala J. Meyer; Lorraine B. Ware; David C. Christiani; Stefan W. Ryter; Rebecca M. Baron; Augustine M. K. Choi

RATIONALE Despite advances in clinical management, there are currently no reliable diagnostic and therapeutic targets for acute respiratory distress syndrome (ARDS). The inflammasome/caspase-1 pathway regulates the maturation and secretion of proinflammatory cytokines (e.g., IL-18). IL-18 is associated with injury in animal models of systemic inflammation. OBJECTIVES We sought to determine the contribution of the inflammasome pathway in experimental acute lung injury and human ARDS. METHODS We performed comprehensive gene expression profiling on peripheral blood from patients with critical illness. Gene expression changes were assessed using real-time polymerase chain reaction, and IL-18 levels were measured in the plasma of the critically ill patients. Wild-type mice or mice genetically deficient in IL-18 or caspase-1 were mechanically ventilated using moderate tidal volume (12 ml/kg). Lung injury parameters were assessed in lung tissue, serum, and bronchoalveolar lavage fluid. MEASUREMENTS AND MAIN RESULTS In mice, mechanical ventilation enhanced IL-18 levels in the lung, serum, and bronchoalveolar lavage fluid. IL-18-neutralizing antibody treatment, or genetic deletion of IL-18 or caspase-1, reduced lung injury in response to mechanical ventilation. In human patients with ARDS, inflammasome-related mRNA transcripts (CASP1, IL1B, and IL18) were increased in peripheral blood. In samples from four clinical centers, IL-18 was elevated in the plasma of patients with ARDS (sepsis or trauma-induced ARDS) and served as a novel biomarker of intensive care unit morbidity and mortality. CONCLUSIONS The inflammasome pathway and its downstream cytokines play critical roles in ARDS development.


Science Translational Medicine | 2013

Sepsis: An integrated clinico-metabolomic model improves prediction of death in sepsis

Raymond J. Langley; Ephraim L. Tsalik; Jennifer C. van Velkinburgh; Seth W. Glickman; Brandon J. Rice; Chunping Wang; Bo Chen; Lawrence Carin; Arturo Suarez; Robert P. Mohney; D. Freeman; Mu Wang; Jinsam You; Jacob Wulff; J. Will Thompson; M. Arthur Moseley; Stephanie Reisinger; Brian T. Edmonds; Brian W. Grinnell; David R. Nelson; Darrell L. Dinwiddie; Neil A. Miller; Carol J. Saunders; Sarah S. Soden; Angela J. Rogers; Lee Gazourian; Anthony F. Massaro; Rebecca M. Baron; Augustine M. K. Choi; G. Ralph Corey

A molecular signature, derived from integrated analysis of clinical data, the metabolome, and the proteome in prospective human studies, improved the prediction of death in patients with sepsis, potentially identifying a subset of patients who merit intensive treatment. Understanding Survival of the Fittest in Sepsis Differentiating mild infections from life-threatening ones is a complex decision that is made millions of times a year in U.S. emergency rooms. Should a patient be sent home with antibiotics and chicken soup? Or should he or she be hospitalized for intensive treatment? Sepsis—a serious infection that is associated with a generalized inflammatory response—is one of the leading causes of death. In two prospective clinical studies reported by Langley et al., patients arriving at four urban emergency departments with symptoms of sepsis were evaluated clinically and by analysis of their plasma proteome and metabolome. Survivors and nonsurvivors at 28 days were compared, and a molecular signature was detected that appeared to differentiate these outcomes—even as early as the time of hospital arrival. The signature was part of a large set of differences between these groups, showing that better energy-producing fatty acid catabolism was associated with survival of the fittest in sepsis. A test developed from the signature was able to predict sepsis survival and nonsurvival reproducibly and better than current methods. This test could help to make all important decisions in the emergency room more accurate. Sepsis is a common cause of death, but outcomes in individual patients are difficult to predict. Elucidating the molecular processes that differ between sepsis patients who survive and those who die may permit more appropriate treatments to be deployed. We examined the clinical features and the plasma metabolome and proteome of patients with and without community-acquired sepsis, upon their arrival at hospital emergency departments and 24 hours later. The metabolomes and proteomes of patients at hospital admittance who would ultimately die differed markedly from those of patients who would survive. The different profiles of proteins and metabolites clustered into the following groups: fatty acid transport and β-oxidation, gluconeogenesis, and the citric acid cycle. They differed consistently among several sets of patients, and diverged more as death approached. In contrast, the metabolomes and proteomes of surviving patients with mild sepsis did not differ from survivors with severe sepsis or septic shock. An algorithm derived from clinical features together with measurements of five metabolites predicted patient survival. This algorithm may help to guide the treatment of individual patients with sepsis.


The Lancet Respiratory Medicine | 2015

Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial

Jennifer G. Wilson; Kathleen D. Liu; Hanjing Zhuo; Lizette Caballero; Melanie McMillan; Xiaohui Fang; Katherine Cosgrove; Rosemary Vojnik; Carolyn S. Calfee; Jae-Woo Lee; Angela J. Rogers; Joseph E. Levitt; Jeanine P. Wiener-Kronish; Ednan K. Bajwa; Andrew D. Leavitt; David H. McKenna; B. Taylor Thompson; Michael A. Matthay

BACKGROUND No effective pharmacotherapy for acute respiratory distress syndrome (ARDS) exists, and mortality remains high. Preclinical studies support the efficacy of mesenchymal stem (stromal) cells (MSCs) in the treatment of lung injury. We aimed to test the safety of a single dose of allogeneic bone marrow-derived MSCs in patients with moderate-to-severe ARDS. METHODS The STem cells for ARDS Treatment (START) trial was a multicentre, open-label, dose-escalation, phase 1 clinical trial. Patients were enrolled in the intensive care units at University of California, San Francisco, CA, USA, Stanford University, Stanford, CA, USA, and Massachusetts General Hospital, Boston, MA, USA, between July 8, 2013, and Jan 13, 2014. Patients were included if they had moderate-to-severe ARDS as defined by the acute onset of the need for positive pressure ventilation by an endotracheal or tracheal tube, a PaO2:FiO2 less than 200 mm Hg with at least 8 cm H2O positive end-expiratory airway pressure (PEEP), and bilateral infiltrates consistent with pulmonary oedema on frontal chest radiograph. The first three patients were treated with low dose MSCs (1 million cells/kg predicted bodyweight [PBW]), the next three patients received intermediate dose MSCs (5 million cells/kg PBW), and the final three patients received high dose MSCs (10 million cells/kg PBW). Primary outcomes included the incidence of prespecified infusion-associated events and serious adverse events. The trial is registered with ClinicalTrials.gov, number NCT01775774. FINDINGS No prespecified infusion-associated events or treatment-related adverse events were reported in any of the nine patients. Serious adverse events were subsequently noted in three patients during the weeks after the infusion: one patient died on study day 9, one patient died on study day 31, and one patient was discovered to have multiple embolic infarcts of the spleen, kidneys, and brain that were age-indeterminate, but thought to have occurred before the MSC infusion based on MRI results. None of these severe adverse events were thought to be MSC-related. INTERPRETATION A single intravenous infusion of allogeneic, bone marrow-derived human MSCs was well tolerated in nine patients with moderate to severe ARDS. Based on this phase 1 experience, we have proceeded to phase 2 testing of MSCs for moderate to severe ARDS with a primary focus on safety and secondary outcomes including respiratory, systemic, and biological endpoints. FUNDING The National Heart, Lung, and Blood Institute.


Genomics | 2009

Genetic association analysis of copy-number variation (CNV) in human disease pathogenesis.

Iuliana Ionita-Laza; Angela J. Rogers; Christoph Lange; Benjamin A. Raby; Charles Lee

Structural genetic variation, including copy-number variation (CNV), constitutes a substantial fraction of total genetic variability and the importance of structural genetic variants in modulating human disease is increasingly being recognized. Early successes in identifying disease-associated CNVs via a candidate gene approach mandate that future disease association studies need to include structural genetic variation. Such analyses should not rely on previously developed methodologies that were designed to evaluate single nucleotide polymorphisms (SNPs). Instead, development of novel technical, statistical, and epidemiologic methods will be necessary to optimally capture this newly-appreciated form of genetic variation in a meaningful manner.


PLOS Medicine | 2013

Circulating Mitochondrial DNA in Patients in the ICU as a Marker of Mortality: Derivation and Validation

Kiichi Nakahira; Sun-Young Kyung; Angela J. Rogers; Lee Gazourian; Sojung Youn; Anthony F. Massaro; Carolina Quintana; Juan C. Osorio; Zhaoxi Wang; Yang Zhao; Laurie Lawler; Jason D. Christie; Nuala J. Meyer; Finnian R. Mc Causland; Sushrut S. Waikar; Aaron B. Waxman; Raymond T. Chung; Raphael Bueno; Ivan O. Rosas; Rebecca M. Baron; David C. Christiani; Gary M. Hunninghake; Augustine M. K. Choi

In this paper, Choi and colleagues analyzed levels of mitochondrial DNA in two prospective observational cohort studies and found that increased mtDNA levels are associated with ICU mortality, and improve risk prediction in medical ICU patients. The data suggests that mtDNA could serve as a viable plasma biomarker in MICU patients.


Current Opinion in Genetics & Development | 2009

Asthma genetics and genomics 2009

Scott T. Weiss; Benjamin A. Raby; Angela J. Rogers

Asthma Genetic Association studies have been plagued by methodologic problems that are common in all studies of complex traits: small sample size, lack of replication, and lack of control of population stratification. Despite this, the field has identified 43 replicated genes from association studies. The most frequently replicated are: TNF alpha, IL4, FCERB, Adam 33, and GSTP1. Several genes have been identified by linkage and fine mapping (ADAM33, DPP10, GPR154, and PHF11) and one gene has been identified by GWAS (ORMD3). The major issue is that these genes have been looked at one at a time rather than in some more holistic manner where epistasis is considered. For asthma genetics to begin to have an impact on clinical medicine we need to consider epistatic interaction.


American Journal of Respiratory and Critical Care Medicine | 2010

A Role for Wnt Signaling Genes in the Pathogenesis of Impaired Lung Function in Asthma

Sunita Sharma; Kelan G. Tantisira; Vincent J. Carey; Amy Murphy; Jessica Lasky-Su; Juan C. Celedón; Ross Lazarus; Barbara J. Klanderman; Angela J. Rogers; Manuel Soto-Quiros; Lydiana Avila; Thomas J. Mariani; Roger Gaedigk; Stephen Leeder; John S. Torday; David Warburton; Benjamin A. Raby; Scott T. Weiss

RATIONALE Animal models demonstrate that aberrant gene expression in utero can result in abnormal pulmonary phenotypes. OBJECTIVES We sought to identify genes that are differentially expressed during in utero airway development and test the hypothesis that variants in these genes influence lung function in patients with asthma. METHODS Stage 1 (Gene Expression): Differential gene expression analysis across the pseudoglandular (n = 27) and canalicular (n = 9) stages of human lung development was performed using regularized t tests with multiple comparison adjustments. Stage 2 (Genetic Association): Genetic association analyses of lung function (FEV(1), FVC, and FEV(1)/FVC) for variants in five differentially expressed genes were conducted in 403 parent-child trios from the Childhood Asthma Management Program (CAMP). Associations were replicated in 583 parent-child trios from the Genetics of Asthma in Costa Rica study. MEASUREMENTS AND MAIN RESULTS Of the 1,776 differentially expressed genes between the pseudoglandular (gestational age: 7-16 wk) and the canalicular (gestational age: 17-26 wk) stages, we selected 5 genes in the Wnt pathway for association testing. Thirteen single nucleotide polymorphisms in three genes demonstrated association with lung function in CAMP (P < 0.05), and associations for two of these genes were replicated in the Costa Ricans: Wnt1-inducible signaling pathway protein 1 with FEV(1) (combined P = 0.0005) and FVC (combined P = 0.0004), and Wnt inhibitory factor 1 with FVC (combined P = 0.003) and FEV(1)/FVC (combined P = 0.003). CONCLUSIONS Wnt signaling genes are associated with impaired lung function in two childhood asthma cohorts. Furthermore, gene expression profiling of human fetal lung development can be used to identify genes implicated in the pathogenesis of lung function impairment in individuals with asthma.


PLOS ONE | 2014

Metabolomic derangements are associated with mortality in critically ill adult patients.

Angela J. Rogers; Michael J. McGeachie; Rebecca M. Baron; Lee Gazourian; Jeffrey A. Haspel; Kiichi Nakahira; Gary M. Hunninghake; Benjamin A. Raby; Michael A. Matthay; Ronny M. Otero; Vance G. Fowler; Emanuel P. Rivers; Christopher W. Woods; Stephen F. Kingsmore; Raymond J. Langley; Augustine M. K. Choi

Objective To identify metabolomic biomarkers predictive of Intensive Care Unit (ICU) mortality in adults. Rationale Comprehensive metabolomic profiling of plasma at ICU admission to identify biomarkers associated with mortality has recently become feasible. Methods We performed metabolomic profiling of plasma from 90 ICU subjects enrolled in the BWH Registry of Critical Illness (RoCI). We tested individual metabolites and a Bayesian Network of metabolites for association with 28-day mortality, using logistic regression in R, and the CGBayesNets Package in MATLAB. Both individual metabolites and the network were tested for replication in an independent cohort of 149 adults enrolled in the Community Acquired Pneumonia and Sepsis Outcome Diagnostics (CAPSOD) study. Results We tested variable metabolites for association with 28-day mortality. In RoCI, nearly one third of metabolites differed among ICU survivors versus those who died by day 28 (N = 57 metabolites, p<.05). Associations with 28-day mortality replicated for 31 of these metabolites (with p<.05) in the CAPSOD population. Replicating metabolites included lipids (N = 14), amino acids or amino acid breakdown products (N = 12), carbohydrates (N = 1), nucleotides (N = 3), and 1 peptide. Among 31 replicated metabolites, 25 were higher in subjects who progressed to die; all 6 metabolites that are lower in those who die are lipids. We used Bayesian modeling to form a metabolomic network of 7 metabolites associated with death (gamma-glutamylphenylalanine, gamma-glutamyltyrosine, 1-arachidonoylGPC(20:4), taurochenodeoxycholate, 3-(4-hydroxyphenyl) lactate, sucrose, kynurenine). This network achieved a 91% AUC predicting 28-day mortality in RoCI, and 74% of the AUC in CAPSOD (p<.001 in both populations). Conclusion Both individual metabolites and a metabolomic network were associated with 28-day mortality in two independent cohorts. Metabolomic profiling represents a valuable new approach for identifying novel biomarkers in critically ill patients.


American Journal of Respiratory and Critical Care Medicine | 2014

Integrative "omic" analysis of experimental bacteremia identifies a metabolic signature that distinguishes human sepsis from systemic inflammatory response syndromes.

Raymond J. Langley; Jennifer L. Tipper; Shannon Bruse; Rebecca M. Baron; Ephraim L. Tsalik; James Huntley; Angela J. Rogers; Richard J. Jaramillo; Denise O'Donnell; William Mega; Mignon Keaton; Elizabeth Kensicki; Lee Gazourian; Anthony F. Massaro; Ronny M. Otero; Vance G. Fowler; Emanuel P. Rivers; Christopher W. Woods; Stephen F. Kingsmore; Mohan L. Sopori; Mark A. Perrella; Augustine M. K. Choi; Kevin S. Harrod

RATIONALE Sepsis is a leading cause of morbidity and mortality. Currently, early diagnosis and the progression of the disease are difficult to make. The integration of metabolomic and transcriptomic data in a primate model of sepsis may provide a novel molecular signature of clinical sepsis. OBJECTIVES To develop a biomarker panel to characterize sepsis in primates and ascertain its relevance to early diagnosis and progression of human sepsis. METHODS Intravenous inoculation of Macaca fascicularis with Escherichia coli produced mild to severe sepsis, lung injury, and death. Plasma samples were obtained before and after 1, 3, and 5 days of E. coli challenge and at the time of killing. At necropsy, blood, lung, kidney, and spleen samples were collected. An integrative analysis of the metabolomic and transcriptomic datasets was performed to identify a panel of sepsis biomarkers. MEASUREMENTS AND MAIN RESULTS The extent of E. coli invasion, respiratory distress, lethargy, and mortality was dependent on the bacterial dose. Metabolomic and transcriptomic changes characterized severe infections and death, and indicated impaired mitochondrial, peroxisomal, and liver functions. Analysis of the pulmonary transcriptome and plasma metabolome suggested impaired fatty acid catabolism regulated by peroxisome-proliferator activated receptor signaling. A representative four-metabolite model effectively diagnosed sepsis in primates (area under the curve, 0.966) and in two human sepsis cohorts (area under the curve, 0.78 and 0.82). CONCLUSIONS A model of sepsis based on reciprocal metabolomic and transcriptomic data was developed in primates and validated in two human patient cohorts. It is anticipated that the identified parameters will facilitate early diagnosis and management of sepsis.


Pharmacogenomics | 2009

Predictors of poor response during asthma therapy differ with definition of outcome.

Angela J. Rogers; Kelan G. Tantisira; Anne L. Fuhlbrigge; Augusto A. Litonjua; Jessica Lasky-Su; Stanley J. Szefler; Robert C. Strunk; Robert S. Zeiger; Scott T. Weiss

AIMS To evaluate phenotypic and genetic variables associated with a poor long-term response to inhaled corticosteroid therapy for asthma, based independently on lung function changes or asthma exacerbations. MATERIALS & METHODS We tested 17 phenotypic variables and polymorphisms in FCER2 and CRHR1 in 311 children (aged 5-12 years) randomized to a 4-year course of inhaled corticosteroid during the Childhood Asthma Management Program (CAMP). RESULTS Predictors of recurrent asthma exacerbations are distinct from predictors of poor lung function response. A history of prior asthma exacerbations, younger age and a higher IgE level (p < 0.05) are associated with recurrent exacerbations. By contrast, lower bronchodilator response to albuterol and the minor alleles of RS242941 in CRHR1 and T2206C in FCER2 (p < 0.05) are associated with poor lung function response. Poor lung function response does not increase the risk of exacerbations and vice versa (p = 0.72). CONCLUSION Genetic and phenotypic predictors of a poor long-term response to inhaled corticosteroids differ markedly depending on definition of outcome (based on exacerbations vs lung function). These findings are important in comparing outcomes of clinical trials and in designing future pharmacogenetic studies.

Collaboration


Dive into the Angela J. Rogers's collaboration.

Top Co-Authors

Avatar

Benjamin A. Raby

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Rebecca M. Baron

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lee Gazourian

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jessica Lasky-Su

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Scott T. Weiss

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy Murphy

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony F. Massaro

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge