Network


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

Hotspot


Dive into the research topics where Brian B. Graham is active.

Publication


Featured researches published by Brian B. Graham.


American Journal of Respiratory and Critical Care Medicine | 2012

Modern age pathology of pulmonary arterial hypertension.

Elvira Stacher; Brian B. Graham; James M. Hunt; Aneta Gandjeva; Steve D. Groshong; Vallerie V. McLaughlin; Marsha Jessup; William E. Grizzle; Michaela A. Aldred; Carlyne D. Cool; Rubin M. Tuder

RATIONALE The impact of modern treatments of pulmonary arterial hypertension (PAH) on pulmonary vascular pathology remains unknown. OBJECTIVES To assess the spectrum of pulmonary vascular remodeling in the modern era of PAH medication. METHODS Assessment of pulmonary vascular remodeling and inflammation in 62 PAH and 28 control explanted lungs systematically sampled. MEASUREMENTS AND MAIN RESULTS Intima and intima plus media fractional thicknesses of pulmonary arteries were increased in the PAH group versus the control lungs and correlated with pulmonary hemodynamic measurements. Despite a high variability of morphological measurements within a given PAH lung and among all PAH lungs, distinct pathological subphenotypes were detected in cohorts of PAH lungs. These included a subset of lungs lacking intima or, most prominently, media remodeling, which had similar numbers of profiles of plexiform lesions as those in lungs with more pronounced remodeling. Marked perivascular inflammation was present in a high number of PAH lungs and correlated with intima plus media remodeling. The number of profiles of plexiform lesions was significantly lower in lungs of male patients and those never treated with prostacyclin or its analogs. CONCLUSIONS Our results indicate that multiple features of pulmonary vascular remodeling are present in patients treated with modern PAH therapies. Perivascular inflammation may have an important role in the processes of vascular remodeling, all of which may ultimately lead to increased pulmonary artery pressure. Moreover, our study provides a framework to interpret and design translational studies in PAH.


Journal of Clinical Investigation | 2014

Systems-level regulation of microRNA networks by miR-130/301 promotes pulmonary hypertension

Thomas Bertero; Yu Lu; Sofia Annis; Andrew Hale; Balkrishen Bhat; Rajan Saggar; Rajeev Saggar; W. Dean Wallace; David J. Ross; Sara O. Vargas; Brian B. Graham; Rahul Kumar; Stephen M. Black; Sohrab Fratz; Jeffrey R. Fineman; James West; Kathleen J. Haley; Aaron B. Waxman; B. Nelson Chau; Katherine A. Cottrill; Stephen Y. Chan

Development of the vascular disease pulmonary hypertension (PH) involves disparate molecular pathways that span multiple cell types. MicroRNAs (miRNAs) may coordinately regulate PH progression, but the integrative functions of miRNAs in this process have been challenging to define with conventional approaches. Here, analysis of the molecular network architecture specific to PH predicted that the miR-130/301 family is a master regulator of cellular proliferation in PH via regulation of subordinate miRNA pathways with unexpected connections to one another. In validation of this model, diseased pulmonary vessels and plasma from mammalian models and human PH subjects exhibited upregulation of miR-130/301 expression. Evaluation of pulmonary arterial endothelial cells and smooth muscle cells revealed that miR-130/301 targeted PPARγ with distinct consequences. In endothelial cells, miR-130/301 modulated apelin-miR-424/503-FGF2 signaling, while in smooth muscle cells, miR-130/301 modulated STAT3-miR-204 signaling to promote PH-associated phenotypes. In murine models, induction of miR-130/301 promoted pathogenic PH-associated effects, while miR-130/301 inhibition prevented PH pathogenesis. Together, these results provide insight into the systems-level regulation of miRNA-disease gene networks in PH with broad implications for miRNA-based therapeutics in this disease. Furthermore, these findings provide critical validation for the evolving application of network theory to the discovery of the miRNA-based origins of PH and other diseases.


American Journal of Pathology | 2010

Tracheal Basal cells: a facultative progenitor cell pool.

Brook B. Cole; Russell W. Smith; Kimberly M. Jenkins; Brian B. Graham; Paul R. Reynolds; Susan D. Reynolds

Analysis of lineage relationships in the naphthalene-injured tracheal epithelium demonstrated that two multipotential keratin 14-expressing cells (K14ECs) function as progenitors for Clara and ciliated cells. These K14EC were distinguished by their self-renewal capacity and were hypothesized to reside at the stem and transit amplifying tiers of a tissue-specific stem cell hierarchy. In this study, we used gene expression and histomorphometric analysis of the steady-state and naphthalene-injured trachea to evaluate the predictions of this model. We found that the steady-state tracheal epithelium is maintained by two progenitor cell pools, secretory and basal cells, and the latter progenitor pool is further divided into two subsets, keratin 14-negative and -positive. After naphthalene-mediated depletion of the secretory and ciliated cell types, the two basal cell pools coordinate to restore the epithelium. Both basal cell types up-regulate keratin 14 and generate a broadly distributed, abundant, and highly mitotic cell pool. Furthermore, basal cell proliferation is associated with generation of differentiated Clara and ciliated cells. The uniform distribution of basal cell progenitors and of their differentiated progeny leads us to propose that the hierarchical organization of tracheal reparative cells be revised to include a facultative basal cell progenitor pool.


Cell Metabolism | 2013

Deletion of iron regulatory protein 1 causes polycythemia and pulmonary hypertension in mice through translational derepression of HIF2α

Manik C. Ghosh; De-Liang Zhang; Suh Young Jeong; Gennadiy Kovtunovych; Hayden Ollivierre-Wilson; Audrey Noguchi; Tiffany Tu; Thomas Senecal; Gabrielle Robinson; Daniel R. Crooks; Wing Hang Tong; Kavitha Ramaswamy; Anamika Singh; Brian B. Graham; Rubin M. Tuder; Zu Xi Yu; Michael Eckhaus; Jaekwon Lee; Danielle A. Springer; Tracey A. Rouault

Iron regulatory proteins (Irps) 1 and 2 posttranscriptionally control the expression of transcripts that contain iron-responsive element (IRE) sequences, including ferritin, ferroportin, transferrin receptor, and hypoxia-inducible factor 2α (HIF2α). We report here that mice with targeted deletion of Irp1 developed pulmonary hypertension and polycythemia that was exacerbated by a low-iron diet. Hematocrits increased to 65% in iron-starved mice, and many polycythemic mice died of abdominal hemorrhages. Irp1 deletion enhanced HIF2α protein expression in kidneys of Irp1(-/-) mice, which led to increased erythropoietin (EPO) expression, polycythemia, and concomitant tissue iron deficiency. Increased HIF2α expression in pulmonary endothelial cells induced high expression of endothelin-1, likely contributing to the pulmonary hypertension of Irp1(-/-) mice. Our results reveal why anemia is an early physiological consequence of iron deficiency, highlight the physiological significance of Irp1 in regulating erythropoiesis and iron distribution, and provide important insights into the molecular pathogenesis of pulmonary hypertension.


Critical Care Medicine | 2010

Diabetes mellitus does not adversely affect outcomes from a critical illness.

Brian B. Graham; Angela Keniston; Ognjen Gajic; Cesar Trillo Alvarez; Sofia Medvedev; Ivor S. Douglas

Objective:Chronic diabetes mellitus (DM) is a known cause of multisystem injury. The effect of DM in acute critical illness may also be detrimental, but is not specifically known. We hypothesized that the preexisting diagnosis of DM is an independent risk factor for mortality in critically ill patients. Design:Parallel retrospective and prospective cohort study. Setting:Two large patient datasets were used: the retrospective University HealthSystem Consortium database (UHC) and the prospective Mayo Clinic Acute Physiology And Chronic Health Evaluation III critical care database (Mayo). Patients:Inclusion criteria were admission to an intensive care unit and age ≥18 yrs. Patients with diabetic ketoacidosis or hyperosmolar nonketotic coma were excluded. A total of 1,509,890 patients (including 143,078 deaths) in the UHC cohort and 36,414 patients (including 3562 deaths) in the Mayo cohort were included in the study analysis. Measurements and Main Results:The primary outcome was in-hospital mortality compared between patients with a history of DM and all other patients. Other outcomes included in-hospital mortality in prespecified subgroups. In the UHC dataset, patients with DM had a lower unadjusted odds ratio (0.90, 95% confidence interval 0.89–0.91, p < .001) and a lower adjusted effect on mortality (odds ratio 0.75, 0.74–0.76, p < .001) compared with that seen in patients without DM. In the Mayo dataset, patients with DM had a comparable unadjusted odds ratio (1.07, 0.97–1.17, p = NS) and a lower adjusted effect on mortality (odds ratio 0.88, 0.79–0.98, p = .022) compared with that seen in patients without DM. A lower mortality in diabetic patients held across multiple demographic subgroups, including patients who underwent coronary-artery bypass grafting (UHC data: unadjusted odds ratio 0.66, 0.62–0.71, p < .001). Conclusions:Critically ill adults with DM do not have an increased mortality compared with that seen in patients without DM, and may have a decreased mortality. Further investigation needs to be done to determine the mechanism for this effect.


Journal of Immunology | 2014

Adventitial Fibroblasts Induce a Distinct Proinflammatory/Profibrotic Macrophage Phenotype in Pulmonary Hypertension

Karim C. El Kasmi; Steven C. Pugliese; Suzette R. Riddle; Jens M. Poth; Aimee L. Anderson; Maria G. Frid; Min Li; Soni Savai Pullamsetti; Rajkumar Savai; Maria A. Nagel; Mehdi A. Fini; Brian B. Graham; Rubin M. Tuder; Jacob E. Friedman; Holger K. Eltzschig; Ronald J. Sokol; Kurt R. Stenmark

Macrophage accumulation is not only a characteristic hallmark but is also a critical component of pulmonary artery remodeling associated with pulmonary hypertension (PH). However, the cellular and molecular mechanisms that drive vascular macrophage activation and their functional phenotype remain poorly defined. Using multiple levels of in vivo (bovine and rat models of hypoxia-induced PH, together with human tissue samples) and in vitro (primary mouse, rat, and bovine macrophages, human monocytes, and primary human and bovine fibroblasts) approaches, we observed that adventitial fibroblasts derived from hypertensive pulmonary arteries (bovine and human) regulate macrophage activation. These fibroblasts activate macrophages through paracrine IL-6 and STAT3, HIF1, and C/EBPβ signaling to drive expression of genes previously implicated in chronic inflammation, tissue remodeling, and PH. This distinct fibroblast-activated macrophage phenotype was independent of IL-4/IL-13–STAT6 and TLR–MyD88 signaling. We found that genetic STAT3 haplodeficiency in macrophages attenuated macrophage activation, complete STAT3 deficiency increased macrophage activation through compensatory upregulation of STAT1 signaling, and deficiency in C/EBPβ or HIF1 attenuated fibroblast-driven macrophage activation. These findings challenge the current paradigm of IL-4/IL-13–STAT6–mediated alternative macrophage activation as the sole driver of vascular remodeling in PH, and uncover a cross-talk between adventitial fibroblasts and macrophages in which paracrine IL-6–activated STAT3, HIF1α, and C/EBPβ signaling are critical for macrophage activation and polarization. Thus, targeting IL-6 signaling in macrophages by completely inhibiting C/EBPβ or HIF1α or by partially inhibiting STAT3 may hold therapeutic value for treatment of PH and other inflammatory conditions characterized by increased IL-6 and absent IL-4/IL-13 signaling.


Annals of the American Thoracic Society | 2013

Fasting 2-Deoxy-2-[18F]fluoro-d-glucose Positron Emission Tomography to Detect Metabolic Changes in Pulmonary Arterial Hypertension Hearts over 1 Year

Erika L. Lundgrin; Margaret Park; Jacqueline Sharp; W.H. Wilson Tang; James D. Thomas; Kewal Asosingh; Suzy Comhair; Frank P. DiFilippo; Donald R. Neumann; Laura Davis; Brian B. Graham; Rubin M. Tuder; Iva Dostanic; Serpil C. Erzurum

BACKGROUND The development of tools to monitor the right ventricle in pulmonary arterial hypertension (PAH) is of clinical importance. PAH is associated with pathologic expression of the transcription factor hypoxia-inducible factor (HIF)-1α, which induces glycolytic metabolism and mobilization of proangiogenic progenitor (CD34(+)CD133(+)) cells. We hypothesized that PAH cardiac myocytes have a HIF-related switch to glycolytic metabolism that can be detected with fasting 2-deoxy-2-[(18)F]fluoro-d-glucose positron emission tomography (FDG-PET) and that glucose uptake is informative for cardiac function. METHODS Six healthy control subjects and 14 patients with PAH underwent fasting FDG-PET and echocardiogram. Blood CD34(+)CD133(+) cells and erythropoietin were measured as indicators of HIF activation. Twelve subjects in the PAH cohort underwent repeat studies 1 year later to determine if changes in FDG uptake were related to changes in echocardiographic parameters or to measures of HIF activation. MEASUREMENTS AND RESULTS FDG uptake in the right ventricle was higher in patients with PAH than in healthy control subjects and correlated with echocardiographic measures of cardiac dysfunction and circulating CD34(+)CD133(+) cells but not erythropoietin. Among patients with PAH, FDG uptake was lower in those receiving β-adrenergic receptor blockers. Changes in FDG uptake over time were related to changes in echocardiographic parameters and CD34(+)CD133(+) cell numbers. Immunohistochemistry of explanted PAH hearts of patients undergoing transplantation revealed that HIF-1α was present in myocyte nuclei but was weakly detectable in control hearts. CONCLUSIONS PAH hearts have pathologic glycolytic metabolism that is quantitatively related to cardiac dysfunction over time, suggesting that metabolic imaging may be useful in therapeutic monitoring of patients.


American Journal of Pathology | 2010

Schistosomiasis-Induced Experimental Pulmonary Hypertension: Role of Interleukin-13 Signaling

Brian B. Graham; Margaret M. Mentink-Kane; Hazim El-Haddad; Shawn Purnell; Li Zhang; Ari Zaiman; Elizabeth F. Redente; David W. H. Riches; Paul M. Hassoun; Angela Pontes Bandeira; Hunter C. Champion; Ghazwan Butrous; Thomas A. Wynn; Rubin M. Tuder

The mechanisms underlying schistosomiasis-induced pulmonary hypertension (PH), one of the most common causes of PH worldwide, remain unclear. We sought to determine whether Schistosoma mansoni causes experimental PH associated with pulmonary vascular remodeling in an interleukin (IL)-13-dependent manner. IL-13Ralpha1 is the canonical IL-13 signaling receptor, whereas IL-13Ralpha2 is a competitive nonsignaling decoy receptor. Wild-type, IL-13Ralpha1(-/-), and IL-13Ralpha2(-/-) C57BL/6J mice were percutaneously infected with S. mansoni cercariae, followed by i.v. injection of eggs. We assessed PH with right ventricular catheterization, histological evaluation of pulmonary vascular remodeling, and detection of IL-13 and transforming growth factor-beta signaling. Infected mice developed pulmonary peri-egg granulomas and arterial remodeling involving predominantly the vascular media. In addition, gain-of-function IL-13Ralpha2(-/-) mice had exacerbated vascular remodeling and PH. Mice with loss of IL-13Ralpha1 function did not develop PH and had reduced pulmonary vascular remodeling. Moreover, the expression of resistin-like molecule-alpha, a target of IL-13 signaling, was increased in infected wild-type and IL-13Ralpha2(-/-) but not IL-13Ralpha1(-/-) mice. Phosphorylated Smad2/3, a target of transforming growth factor-beta signaling, was increased in both infected mice and humans with the disease. Our data indicate that experimental schistosomiasis causes PH and potentially relies on up-regulated IL-13 signaling.


Chest | 2010

Schistosomiasis-Associated Pulmonary Hypertension : Pulmonary Vascular Disease: The Global Perspective

Brian B. Graham; Angela Bandeira; Nicholas W. Morrell; Ghazwan Butrous; Rubin M. Tuder

Inflammation is likely a critical underlying etiology in many forms of severe pulmonary hypertension (PH), and schistosomiasis-associated PH, one of the most common causes of PH worldwide, is likely driven by the host response to parasite antigens. More than 200 million people are infected with schistosomiasis, the third most common parasitic disease, and approximately 1% of those chronically infected develop PH. Acute cutaneous infection causes inflammation at the site of parasite penetration followed by a subacute immune complex-mediated hypersensitivity response as the parasite migrates through the lungs. Chronic schistosomiasis infection induces a granulomatous inflammation around ova deposited in the tissue. In particular, Schistosoma mansoni migrates to the portal venous system and causes preportal fibrosis in a subset of individuals and appears to be a prerequisite for PH. Portal hypertension facilitates shunting of ova from the portal system to the pulmonary arterial tree, resulting in localized periovular pulmonary granulomas. The pulmonary vascular remodeling is likely a direct consequence of the host inflammatory response, and portopulmonary hypertension may be a significant contributor. New specific therapies available for PH have not been widely tested in patients with schistosomiasis and often are unavailable for those infected in resource-poor areas of the world where schistosomiasis is endemic. Furthermore, the current PH therapies in general target vasodilation rather than vascular remodeling and inflammation. Further research is needed into the pathogenic mechanism by which this parasitic infection results in pulmonary vascular remodeling and PH, which also may be informative regarding the etiology of other types of PH.


The New England Journal of Medicine | 2009

Kiss of Death

Brian B. Graham; Daniel R. Kaul; Sanjay Saint; William J. Janssen

n engl j med 360;24 nejm.org june 11, 2009 2564 From the Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver (B.B.G., W.J.J.); the Division of Infectious Disease (D.R.K.), Department of Internal Medicine (S.S.), and Department of Veterans Affairs Health Services Research and Development Center of Excellence and Department of Medicine (S.S.) — all at the University of Michigan, Ann Arbor; and the Department of Medicine, National Jewish Medical and Research Center, Denver (W.J.J.). Address reprint requests to Dr. Graham at the University of Colorado Health Sciences Center, 4200 E. 9th Ave., Box C-272, Denver, CO, 80262, or at brian. [email protected].

Collaboration


Dive into the Brian B. Graham's collaboration.

Top Co-Authors

Avatar

Rubin M. Tuder

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Rahul Kumar

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Kathleen J. Haley

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Sara O. Vargas

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Hale

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

David J. Ross

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rajan Saggar

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge