Network


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

Hotspot


Dive into the research topics where Parasuram Krishnamoorthy is active.

Publication


Featured researches published by Parasuram Krishnamoorthy.


The American Journal of Medicine | 2011

Attributable Risk Estimate of Severe Psoriasis on Major Cardiovascular Events

Nehal N. Mehta; YiDing Yu; Rebecca Pinnelas; Parasuram Krishnamoorthy; Daniel B. Shin; Andrea B. Troxel; Joel M. Gelfand

BACKGROUND Recent studies suggest that psoriasis, particularly if severe, may be a risk factor for major adverse cardiac events, such as myocardial infarction, stroke, and mortality from cardiovascular disease. We compared the risk of major adverse cardiac events between patients with psoriasis and the general population and estimated the attributable risk of severe psoriasis. METHODS We performed a cohort study in the General Practice Research Database. Severe psoriasis was defined as receiving a psoriasis diagnosis and systemic therapy (N=3603). Up to 4 patients without psoriasis were selected from the same practices and start dates for each patient with psoriasis (N=14,330). RESULTS Severe psoriasis was a risk factor for major adverse cardiac events (hazard ratio 1.53; 95% confidence interval, 1.26-1.85) after adjusting for age, gender, diabetes, hypertension, tobacco use, and hyperlipidemia. After fully adjusted analysis, severe psoriasis conferred an additional 6.2% absolute risk of 10-year major adverse cardiac events. CONCLUSION Severe psoriasis confers an additional 6.2% absolute risk of a 10-year rate of major adverse cardiac events compared with the general population. This potentially has important therapeutic implications for cardiovascular risk stratification and prevention in patients with severe psoriasis. Future prospective studies are needed to validate these findings.


Archives of Dermatology | 2011

Systemic and Vascular Inflammation in Patients With Moderate to Severe Psoriasis as Measured by [18F]-Fluorodeoxyglucose Positron Emission Tomography –Computed Tomography (FDG-PET/CT): A Pilot Study

Nehal N. Mehta; YiDing Yu; Babak Saboury; Negar Foroughi; Parasuram Krishnamoorthy; Anna Raper; Amanda Baer; Jules Antigua; Abby S. Van Voorhees; Drew A. Torigian; Abass Alavi; Joel M. Gelfand

OBJECTIVE To evaluate the feasibility of using [18F]-fluorodeoxyglucose positron emission tomography-computed tomography (FDG-PET/CT) to detect and quantify systemic inflammation in patients with psoriasis. DESIGN Case series with a nested case-control study. SETTING Referral dermatology and preventive cardiology practices. PARTICIPANTS Six patients with psoriasis affecting more than 10% of their body surface area and 4 controls age and sex matched to 4 of the patients with psoriasis for a nested case-control study. MAIN OUTCOME MEASURES The FDG uptake in the liver, musculoskeletal structures, and aorta measured by mean standardized uptake value, a measure of FDG tracer uptake by macrophages and other inflammatory cells. RESULTS FDG-PET/CT identified numerous foci of inflammation in 6 patients with psoriasis within the skin, liver, joints, tendons, and aorta. Inflammation in the joints was observed in a patient with psoriatic arthritis as well as in 1 patient with no history of joint disease or joint symptoms. In a nested case-control study, FDG-PET/CT imaging demonstrated increased vascular inflammation in multiple segments of the aorta compared with controls. These findings persisted after adjustment for traditional cardiovascular risk factors in multivariate analysis (mean β = 0.33; P < .001). Patients with psoriasis further demonstrated increased hepatic inflammation after adjusting for cardiovascular risk factors (β = 0.18; P < .001), but the association was no longer significant when adjusted for alcohol intake (β = -0.25; P = .07). CONCLUSION FDG-PET/CT is a sensitive tool for identifying inflammation and can be used to identify clinically observed inflammation in the skin and subclinical inflammation in the blood vessels, joints, and liver of patients with psoriasis.


Atherosclerosis | 2012

Abnormal lipoprotein particles and cholesterol efflux capacity in patients with psoriasis

Nehal N. Mehta; Ron C. Li; Parasuram Krishnamoorthy; YiDing Yu; William Farver; Amrith Rodrigues; Anna Raper; Mackenzie Wilcox; Amanda Baer; Stephanie DerOhannesian; Megan L. Wolfe; Muredach P. Reilly; Daniel J. Rader; Abby VanVoorhees; Joel M. Gelfand

OBJECTIVES Psoriasis is a Th-1/17 mediated inflammatory disease associated with increased risk of cardiovascular disease (CVD). Inflammation may modulate lipoprotein particle number and directly impair HDL functions, in particular reverse cholesterol transport (RCT). We sought to study how chronic in vivo inflammation modulates lipoprotein particle composition using nuclear magnetic resonance spectroscopy (NMR) and HDL efflux in psoriasis. METHODS AND RESULTS We prospectively enrolled a consecutive sample of patients with psoriasis (n = 122) and compared lipoprotein and metabolic risk factors to patients without psoriasis (n = 134). Fasting lipids, insulin, glucose were measured by standard assays, and lipoprotein concentration and size were measured by NMR. In a random subset (n = 100 each group), HDL efflux capacity was quantified using a validated ex vivo system involving the incubation of macrophages with apolipoprotein B-depleted serum from patients. Traditional lipid concentrations were similar in both groups except for HDL concentration which was lower in psoriasis (43 mg/dl (36-58) vs 50 (42-62), p < 0.01). However, NMR showed an atherogenic profile in psoriasis similar to that observed in diabetes, with significant increase in LDL particle concentration [1210.5 (1002-1498) vs 1115 (935-1291), p = 0.02] with decrease in LDL size [20.6 (20.3-21.1) vs 21.3 (20.6-21.1), p < 0.001] beyond CV risk factors and HOMA-IR (p = 0.001). Finally, HDL efflux capacity was lower in psoriasis compared to controls in fully adjusted models (beta -0.14, p = 0.001). CONCLUSIONS These data support a more atherogenic lipoprotein profile by NMR and decreased HDL efflux capacity in psoriasis patients compared to controls beyond CVD risk factors. The abnormal lipoprotein particle composition and HDL efflux capacity in psoriasis may provide a link between psoriasis and CVD.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Severity of Psoriasis Associates With Aortic Vascular Inflammation Detected by FDG PET/CT and Neutrophil Activation in a Prospective Observational Study

Haley B. Naik; Balaji Natarajan; Elena Stansky; Mark A. Ahlman; Heather L. Teague; Taufiq Salahuddin; Qimin Ng; Aditya A. Joshi; Parasuram Krishnamoorthy; Jenny Dave; Shawn Rose; Julia Doveikis; Martin P. Playford; Ronald B. Prussick; Alison Ehrlich; Mariana J. Kaplan; Benjamin N. Lockshin; Joel M. Gelfand; Nehal N. Mehta

Objective—To understand whether directly measured psoriasis severity is associated with vascular inflammation assessed by 18F-fluorodeoxyglucose positron emission tomography computed tomography. Approach—In-depth cardiovascular and metabolic phenotyping was performed in adult psoriasis patients (n=60) and controls (n=20). Psoriasis severity was measured using psoriasis area severity index. Vascular inflammation was measured using average aortic target-to-background ratio using 18F-fluorodeoxyglucose positron emission tomography computed tomography. Results—Both the psoriasis patients (28 men and 32 women, mean age 47 years) and controls (13 men and 7 women, mean age 41 years) were young with low cardiovascular risk. Psoriasis area severity index scores (median 5.4; interquartile range 2.8–8.3) were consistent with mild-to-moderate skin disease severity. Increasing psoriasis area severity index score was associated with an increase in aortic target-to-background ratio (&bgr;=0.41, P=0.001), an association that changed little after adjustment for age, sex, and Framingham risk score. We observed evidence of increased neutrophil frequency (mean psoriasis, 3.7±1.2 versus 2.9±1.2; P=0.02) and activation by lower neutrophil surface CD16 and CD62L in blood. Serum levels of S100A8/A9 (745.1±53.3 versus 195.4±157.8 ng/mL; P<0.01) and neutrophil elastase-1 (43.0±2.4 versus 30.8±6.7 ng/mL; P<0.001) were elevated in psoriasis. Finally, S100A8/A9 protein was related to both psoriasis skin disease severity (&bgr;=0.53; P=0.02) and vascular inflammation (&bgr;=0.48; P=0.02). Conclusions—Psoriasis severity is associated with vascular inflammation beyond cardiovascular risk factors. Psoriasis increased neutrophil activation and neutrophil markers, and S100A8/A9 was related to both skin disease severity and vascular inflammation.


American Journal of Cardiology | 2014

Meta-Analysis of Left Ventricular Hypertrophy and Sustained Arrhythmias

Saurav Chatterjee; Chirag Bavishi; Partha Sardar; Vikram Agarwal; Parasuram Krishnamoorthy; Tomasz Grodzicki; Franz H. Messerli

Presence of left ventricular hypertrophy (LVH) has been reported to be associated with supraventricular and ventricular arrhythmias, but the association has not been systematically quantified and evaluated. A systematic search of studies in MEDLINE, EMBASE, CINAHL, Scopus, Web of Science, and Cochrane Central Register of Controlled Trials databases was undertaken through April 2014. Studies reporting on LVH and sustained arrhythmias such as atrial fibrillation and supraventricular tachycardias (SVTs) and ventricular arrhythmias (tachycardia and fibrillation) were identified. Pooled effect estimates were calculated with random-effects models (DerSimonian and Laird). A total of 10 eligible studies with 27,141 patients were included in the analysis. The incidence of SVT in patients with LVH was 11.1% compared with 1.1% among patients without LVH (p<0.001). Patients with LVH had 3.4-fold greater odds of developing SVT (odds ratio 3.39, 95% confidence interval 1.57 to 7.31) than those without LVH, although significant heterogeneity was present (I2=98%). Meta-regression analyses revealed the heterogeneity to have originated from differences in the baseline covariates such as age, male gender, hypertension, and diabetes of the individual studies. The incidence of ventricular arrhythmias was 5.5% compared with 1.2% in patients without LVH (p<0.001). The occurrence of ventricular tachycardia or fibrillation was 2.8-fold greater, in the presence of LVH (odds ratio 2.83, 95% confidence interval 1.78 to 4.51), and there was no significant heterogeneity (I2=9%). Presence of LVH in hypertensive patients is associated with a greater risk of sustained supraventricular/atrial and ventricular arrhythmias, and there is an unmet need for identifying and refining risk stratification for this group.


Journal of the American Heart Association | 2014

Endothelial cell-, platelet-, and monocyte/macrophage-derived microparticles are elevated in psoriasis beyond cardiometabolic risk factors.

Junko Takeshita; Emile R. Mohler; Parasuram Krishnamoorthy; Jonni S. Moore; Wade T. Rogers; Lifeng Zhang; Joel M. Gelfand; Nehal N. Mehta

Background Psoriasis, especially when severe, is a risk factor for cardiometabolic disease beyond traditional risk factors. The mechanism of atherogenesis in psoriasis remains unknown. Cell membrane vesicles (ie, microparticles), released upon cell activation or apoptosis, have recently been associated with cardiometabolic disease and may play a pathogenic role. Microparticle levels, particularly from endothelial cells and platelets, are elevated in patients with cardiovascular disorders, metabolic syndrome, other inflammatory diseases, autoimmune conditions, and have been shown to be predictive of cardiovascular outcomes. Methods and Results Concentrations of microparticles with positive expression for any of 7 cell surface markers (Annexin V, CD3, CD31, CD41a, CD64, CD105, and CD144) were measured in blood samples from psoriasis patients (n=53) and control subjects without psoriasis (n=41). Platelet‐free plasma was separated from whole blood by one‐step centrifugation for microparticle analysis. Microparticles were fluorescently labeled and characterized by flow cytometry. Higher concentrations of CD105 (5.5/μL versus 2.5/μL, P<0.001), CD31 (31/μL versus 18/μL, P=0.002), CD41a (50/μL versus 22/μL, P<0.001), and CD64 (5.0/μL versus 4.1/μL, P=0.02) singly positive microparticles corresponding to endothelial cell‐, platelet‐, and monocyte/macrophage‐derived microparticles, respectively, were found in psoriasis patients compared with controls. These differences persisted after adjustment for traditional cardiometabolic risk factors including body mass index. Conclusions Increased microparticle concentrations, independent of cardiometabolic risk factors, in patients with psoriasis suggest that the presence of increased endothelial cell, platelet, and monocyte/macrophage activation with cell turnover may contribute to the heightened atherogenesis associated with psoriasis.


Journal of The American Academy of Dermatology | 2012

The impact of psoriasis on 10-year Framingham risk

Nehal N. Mehta; Parasuram Krishnamoorthy; YiDing Yu; Omar Khan; Anna Raper; Abby S. Van Voorhees; Andrea B. Troxel; Joel M. Gelfand

Psoriasis is a chronic inflammatory skin disease, and when severe, may be a risk factor for major adverse cardiac events (MACE) including myocardial infarction1, stroke and cardiovascular (CV) death. Indeed, recent studies evaluating the risk of MACE with IL-12/23 inhibitors in randomized trials emphasize the need for efforts at decreasing the risk of cardiovascular disease in psoriasis patients. 2. Risk stratification and treatment goals for cholesterol and blood pressure in psoriasis remains understudied, and the Framingham Risk score (FRS) for CV risk may underestimate long term risk of MACE in psoriasis patients. The Adult Treatment Panel for lipid management recommends that providers consider emerging risk factors when deciding on lipid treatment but does not provide specific guidelines. We described an initial estimate of this excess attributable risk of severe psoriasis on MACE3 and herein describe how this AR may affect FRS in psoriasis. We consecutively enrolled patients (n=138), and calculated FRS before and after adding the AR of psoriasis (6.2%)3, and estimated the reclassification rate by examining patients moved from a lower risk category to a higher risk category. Patients with psoriasis were relatively young with higher than expected tobacco use, alcohol use, body mass index and lipids (Table 1). Nonetheless, their risk for MACE by FRS was low (<10%) (male mean FRS 7.4 ± 7.75, female 5.9 ± 5.86), attributable to their young age. After considering the estimated AR, (mean FRS 13.36 ± 7.10, male 13.92 ± 7.77, female 12.48 ± 5.86), the majority of patients were re-classified to a higher-risk category: 73% (95% CI 61.5%–82.3%) of low risk patients were reclassified as intermediate, and 53% (95% CI 36.4%–64.1%) of intermediate risk patients were reclassified as high risk (Table 2). These findings suggest that adding the estimated AR of psoriasis on MACE to the FRS results in clinically important changes in prevention strategies3. Table 1 Demographic characteristics of study sample Table 2 Category of risk before and after adding psoriasis attributable risk with treatment goals Due to the young age of our population, the majority of patients were in the low risk category, which would not warrant aggressive risk reduction strategies. However, when considering an AR estimate of MACE, the majority of our patients were reclassified into the intermediate risk category which would warrant change in treatment plans and goals for over 60% of our patients (Table 2), an important finding in this population potentially at higher risk for CV disease with increased subclinical vascular inflammation4. We recognize this proof of concept study is limited by the generalizability of UK data to a US population. However, guidelines for identifying CVD risk factors are similar. Secondly, our AR was derived in severe psoriasis defined by treatment with systemic therapy or phototherapy; similarly, over 80% of our patients had been exposed to these therapies permitting this estimate to be applicable in this proof of concept study. The major implication of these findings is that patients with psoriasis may warrant more aggressive control of established CV risk factors. Measures to decrease CV risk include lifestyle and dietary education through targeted counseling. The percentage of these patients requiring drug interventions such as statins and anti-hypertensive is unknown but likely to be small. Finally, this illustration provides a quantitative approach for CV risk estimation in psoriasis, an approach gaining acceptance in the care of patients with rheumatoid arthritis5.


Clinical and Experimental Dermatology | 2014

Psoriasis is associated with decreased plasma adiponectin levels independently of cardiometabolic risk factors

R. C. Li; Parasuram Krishnamoorthy; Julia Doveikis; M. Wilcox; P. Thomas; Daniel J. Rader; Muredach P. Reilly; A. Van Voorhees; Joel M. Gelfand; Nehal N. Mehta

Psoriasis is an inflammatory skin disease that may be associated with an adverse cardiometabolic profile including modulated plasma adiponectin and leptin levels. Whether these levels are independent of cardiometabolic risk factors, which are also prevalent in psoriasis, is not known.


Circulation Research | 2016

GlycA Is a Novel Biomarker of Inflammation and Subclinical Cardiovascular Disease in Psoriasis

Aditya A. Joshi; Joseph B. Lerman; Tsion M. Aberra; Mehdi Afshar; Heather L. Teague; Justin Rodante; Parasuram Krishnamoorthy; Qimin Ng; Tarek Z Aridi; Taufiq Salahuddin; Balaji Natarajan; Benjamin N. Lockshin; Mark A. Ahlman; Marcus Y. Chen; Daniel J. Rader; Muredach P. Reilly; Alan T. Remaley; David A. Bluemke; Martin P. Playford; Joel M. Gelfand; Nehal N. Mehta

RATIONALE GlycA, an emerging inflammatory biomarker, predicted cardiovascular events in population-based studies. Psoriasis, an inflammatory disease associated with increased cardiovascular risk, provides a model to study inflammatory biomarkers in cardiovascular disease (CVD). Whether GlycA associates with psoriasis and how it predicts subclinical CVD beyond high-sensitivity C-reactive protein in psoriasis is unknown. OBJECTIVE To investigate the relationships between GlycA and psoriasis and between GlycA and subclinical CVD. METHODS AND RESULTS Patients with psoriasis and controls (n=412) participated in a 2-stage study. We measured GlycA by nuclear magnetic resonance spectroscopy. National Institutes of Health (NIH) participants underwent 18-F Fluorodeoxyglucose Positron Emission Tomography Computed Tomography (18-FDG PET/CT) scans to assess vascular inflammation (VI) and coronary computed tomographic angiography to quantify coronary artery disease burden. Psoriasis cohorts were young (mean age=47.9), with low cardiovascular risk and moderate skin disease. high-sensitivity C-reactive protein and GlycA were increased in psoriasis compared with controls (GlycA: [PENN: 408.8±75.4 versus 289.4±60.2, P<0.0001; NIH: 415.8±63.2 versus 346.2±46, P<0.0001]) and demonstrated a dose-response with psoriasis severity. In stage 2, VI (β=0.36, P<0.001) and coronary artery disease (β=0.29, P=0.004) associated with GlycA beyond CV risk factors in psoriasis. In receiver operating characteristic analysis, GlycA added value in predicting VI (P=0.01) and coronary artery disease (P<0.01). Finally, initiating anti-tumor necrosis factor therapy (n=16) reduced psoriasis severity (P<0.001), GlycA (463.7±92.5 versus 370.1±78.5, P<0.001) and VI (1.93±0.36 versus 1.76±0.19, P<0.001), whereas GlycA remained associated with VI (β=0.56, P<0.001) post treatment. CONCLUSIONS GlycA associated with psoriasis severity and subclinical CVD beyond traditional CV risk and high-sensitivity C-reactive protein. Moreover, psoriasis treatment reduced GlycA and VI. These findings support the potential use of GlycA in subclinical CVD risk assessment in psoriasis and potentially other inflammatory diseases.


Journal of Translational Medicine | 2012

A human model of inflammatory cardio-metabolic dysfunction; a double blind placebo-controlled crossover trial.

Nehal N. Mehta; Sean P Heffron; Parth Patel; Jane F. Ferguson; Rachana Shah; Christine Hinkle; Parasuram Krishnamoorthy; Rhia Shah; Jennifer Tabita-Martinez; Karen Terembula; Stephen R. Master; Michael R. Rickels; Muredach P. Reilly

BackgroundChronic inflammation may contribute to insulin resistance (IR), metabolic syndrome and atherosclerosis although evidence of causality is lacking in humans. We hypothesized that very low-dose experimental endotoxemia would induce adipose tissue inflammation and systemic IR during a low-grade but asymptomatic inflammatory response and thus provide an experimental model for future tests of pharmacologic and genomic modulation of cardio-metabolic traits in humans.MethodsTen healthy, human volunteers (50% male, 90% Caucasian, mean age 22.7 ± 3.8) were randomized in a double-masked, placebo-controlled, crossover study to separate 36-hour inpatient visits (placebo versus intravenous-LPS 0.6 ng/kg). We measured clinical symptoms via the McGill pain questionnaire and serial vital signs. Plasma and serum were collected for measurement of cytokines, C-reactive protein, insulin and glucose, serial whole blood & subcutaneous adipose tissue mRNA expression were measured by real-time PCR. HOMA-IR, a well-validated measure of IR was calculated to estimate insulin resistance, and frequently sampled intravenous glucose tolerance testing (FSIGTT) was performed to confirm an insulin resistant state. We performed ANOVA and within subject ANOVA to understand the differences in cytokines, adipose tissue inflammation and IR before and after LPS or placebo.ResultsThere was no significant difference between placebo and LPS in clinical responses of symptom scores, body temperature or heart rate. However, low-dose endotoxemia induced a rapid and transient 25-fold induction of plasma TNF-alpha and 100-fold increase in plasma IL-6 (Figure 1B) (p < 0.001 for both) both peaking at two hours, followed by modest inflammation in adipose tissue with increases in mRNA levels of several inflammatory genes known to modulate adipose and systemic insulin resistance. Adipose tissue mRNA levels of IL-6 (peak 6-fold, ANOVA F = 27.5, p < 0.001) and TNF-alpha (peak 1.8-fold, F = 2.9, p = 0.01) increased with MCP-1 (peak 10-fold, F = 5.6, p < 0.01) and fractalkine (CX3CL1) (peak 15-fold, F = 13.3, p < 0.001). Finally, HOMA-IR was 32% higher following LPS compared to placebo (p < 0.01) and insulin sensitivity declined by 21% following LPS compared to placebo (p < 0.05).ConclusionsWe present a low dose human endotoxemia model of inflammation which induces adipose tissue inflammation and systemic insulin resistance in the absence of overt clinical response. Such a model has the potential for broad and safe application in the study of novel therapeutics and genomic influences in cardio-metabolic disease.

Collaboration


Dive into the Parasuram Krishnamoorthy's collaboration.

Top Co-Authors

Avatar

Jalaj Garg

Lehigh Valley Hospital

View shared research outputs
Top Co-Authors

Avatar

Nehal N. Mehta

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chandrasekar Palaniswamy

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Joel M. Gelfand

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Neeraj Shah

Lehigh Valley Hospital

View shared research outputs
Top Co-Authors

Avatar

Hasan Ahmad

New York Medical College

View shared research outputs
Top Co-Authors

Avatar

Janani Rangaswami

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

YiDing Yu

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge