Searched for: in-biosketch:yes
person:atturm01
Effects of fatty acid supplementation in modulation of gut microbiome and T-regulatory cells in health and psoriatic disease [Meeting Abstract]
Malik, F; Manasson, J; Herrera, A; Attur, M; Reddy, S M; Yang, L; Koralov, S; Scher, J U
Background/Purpose: Psoriatic Arthritis (PsA) affects up to 30% patients with psoriasis and is characterized by wide spread synovio-entheseal inflammation. Physiologically, the human gut microbiota metabolizes dietary fiber into shortchain fatty acids (FA)- which exert anti-inflammatory effects by increasing activity of regulatory T cells (Tregs).Moreover, we have previously shown decreased abundance of Akkermansia and Ruminococcus and concomitant decrease in mediumchain FA (MCFA) levels in stool of PsA patients. We therefore hypothesized that FA supplementation may have favorable effects on gut microbiome and lead to increase in tolerance, potentially serving as therapeutic target in psoriatic disease.
Method(s): Wild type (WT)animals were fed SCFA-rich diet for 14 days followed by 16S rRNA sequencing and microbiota analysis of pellet specimens.We then evaluated effects ofMCFA-rich diet in healthy subjects. Peripheral blood and stool samples were collected at days 0, 7 and 14 for 16s rRNA sequencing and FACS. Finally, we conducted a small, prospective, proof-ofprinciple study in new-onset, drug-naive psoriatic disease patients (with or without PsA). Each participant received MCFA (1 gm 4 times a day for 6 weeks). Clinical history was obtained at baseline. Skin and joint exam were performed at baseline and follow up. Serum and stool samples were collected at baseline, weeks 3, and 6 for 16S rRNA sequencing and FACS, respectively. Wilcoxon signed-rank test was used to compare differences in Tregs before and after MCFA-rich administration.
Result(s): SCFA rich diet in WT mice led to statistically significant perturbations in gut bacterial composition 14 days into intervention, with a dramatic increase in commensals (Fig 1A; p<0.001), most notably in Akkermansia(Fig 1B). MCFA administration to healthy subjects (n=7) also led to significant changes in community structure (Fig 2A; p=0.03) and associated increases in circulating Treg cells (Fig 2B; p<0.001). These findings were also observed in psoriatic disease patients (n=4) showing a significant alteration in specific taxa, including Actinobacteria (Fig 2 C; p<0.05) and Mollicutes (p=0.09) and concomitant increase in circulatory Treg cells (Fig 2D)
Conclusion(s): In both health and psoriatic disease, MCFA supplementation is associated with distinct changes in human gut microbiota composition and peripheral Treg cells. These findings rationalize the need for a larger placebo controlled, prospective trial to study the effects of MCFA in patients with psoriasis and PsA as a potential therapy alone or in combination with DMARDs. (Figure Presented)
EMBASE:626435145
ISSN: 2326-5205
CID: 3704992
THE TRANSLATIONAL LANDSCAPE IN ARTICULAR CHONDROCYTES TREATEDWITH INTERLEUKIN-1 REVEALS NOVEL POTENTIAL PLAYERS IN OSTEOARTHRITIS [Meeting Abstract]
Kolupaeva, V.; Katsara, O.; Attur, M.
ISI:000432189700165
ISSN: 1063-4584
CID: 3132612
Safety, Tolerability, and Pharmacodynamics of an Anti-Interleukin-1 alpha/beta Dual Variable Domain Immunoglobulin in Patients With Osteoarthritis of the Knee: A Randomized Phase 1 Study
Wang, Susanne X; Abramson, Steven B; Attur, Mukundan; Karsdal, Morten A; Preston, Richard A; Lozada, Carlos J; Kosloski, Matthew P; Hong, Feng; Jiang, Ping; Saltarelli, Mary J; Hendrickson, Barbara A; Medema, Jeroen K
OBJECTIVE: To investigate the safety, tolerability, pharmacokinetics, and pharmacodynamics of ABT-981, a human dual variable domain immunoglobulin simultaneously targeting interleukin (IL)-1alpha and IL-1beta, in patients with knee osteoarthritis. METHOD: This was a randomized, double-blind, placebo-controlled, single-center study of multiple subcutaneous (SC) injections of ABT-981 in patients with mild-to-moderate osteoarthritis of the knee (NCT01668511). Three cohorts received ABT-981 (0.3, 1, or 3 mg/kg) or placebo every other week for a total of 4 SC injections, and one cohort received ABT-981 (3 mg/kg) or placebo every 4 weeks for a total of 3 SC injections. Assessment of safety and tolerability were the primary objectives. A panel of serum and urine biomarkers of inflammation and joint degradation were evaluated. RESULTS: A total of 36 patients were randomized (ABT-981, n=28; placebo, n=8); 31 (86%) completed the study. Adverse event (AE) rates were comparable between ABT-981 and placebo (54% vs 63%). The most common AE reported with ABT-981 versus placebo was injection site erythema (14% vs 0%). ABT-981 significantly reduced absolute neutrophil count and serum concentrations of IL-1alpha/IL-1beta, high-sensitivity C-reactive protein, and matrix metalloproteinase (MMP)-derived type 1 collagen. Serum concentrations of MMP-derived type 3 collagen and MMP-degraded C-reactive protein demonstrated decreasing trends with ABT-981. Antidrug antibodies were found in 37% of patients but were not associated with the incidence or severity of AEs. CONCLUSION: ABT-981 was generally well tolerated in patients with knee osteoarthritis and engaged relevant tissue targets, eliciting an anti-inflammatory response. Consequently, ABT-981 may provide clinical benefit to patients with inflammation-driven osteoarthritis.
PMID: 28964890
ISSN: 1522-9653
CID: 2720402
Combinatorial Peripheral Blood Inflammatory and MRI-Based Biomarkers Predict Radiographic Joint Space Narrowing in Knee OA [Meeting Abstract]
Samuels, Svetlana Krasnokutsky; Zhou, Hua; Attur, Mukundan; Samuels, Jonathan; Chang, Gregory; Bencardino, Jenny; Ma, Sisi; Rybak, Leon; Abramson, Steven B
ISI:000411824105040
ISSN: 2326-5205
CID: 2766802
Toward understanding the role of cartilage particulates in synovial inflammation
Silverstein, Amy M; Stefani, Robert M; Sobczak, Evie; Tong, Eric L; Attur, Mukundan G; Shah, Roshan P; Bulinski, J Chloe; Ateshian, Gerard A; Hung, Clark T
OBJECTIVE: Arthroscopy with lavage and synovectomy can remove tissue debris from the joint space and the synovial lining to provide pain relief to patients with osteoarthritis (OA). Here, we developed an in vitro model to study the interaction of cartilage wear particles with fibroblast-like synoviocytes (FLS) to better understand the interplay of cartilage particulates with cytokines on cells of the synovium. METHOD: In this study sub-10mum cartilage particles or 1mum latex particles were co-cultured with FLS +/- 10 ng/mL interleukin-1alpha (IL-1 alpha) or tumor necrosis factor- alpha (TNF-alpha). Samples were analyzed for DNA, glycosaminoglycan (GAG), and collagen and media samples were analyzed for media GAG, nitric oxide (NO) and prostaglandin-E2 (PGE2). The nature of the physical interaction between the particles and FLS was determined by microscopy. RESULTS: Both latex and cartilage particles could be phagocytosed by FLS. Cartilage particles were internalized and attached to the surface of both dense monolayers and individual cells. Co-culture of FLS with cartilage particulates resulted in a significant increase in cell sheet DNA and collagen content as well as NO and PGE2 synthesis compared to control and latex treated groups. CONCLUSION: The proliferative response of FLS to cartilage wear particles resulted in an overall increase in ECM content, analogous to the thickening of the synovial lining observed in OA patients. Understanding how cartilage particles interface with the synovium may provide insight into how this interaction contributes to OA progression and may guide the role of lavage and synovectomy for degenerative disease.
PMCID:5554538
PMID: 28365462
ISSN: 1522-9653
CID: 2521292
Serum Urate Levels Predict Joint Space Narrowing in Non-gout Patients with Medial Knee Osteoarthritis
Krasnokutsky, Svetlana; Oshinsky, Charles; Attur, Mukundan; Ma, Sisi; Zhou, Hua; Zheng, Fangfei; Chen, Meng; Patel, Jyoti; Samuels, Jonathan; Pike, Virginia C; Regatte, Ravinder; Bencardino, Jenny; Rybak, Leon; Abramson, Steven; Pillinger, Michael H
OBJECTIVE: OA pathogenesis includes both mechanical and inflammatory features. Studies have implicated synovial fluid urate (UA) as a potential OA biomarker, possibly reflecting chondrocyte damage. Whether serum urate (sUA) levels reflect/contribute to OA is unknown. We investigated whether sUA predicts OA progression in a non-gout knee OA population. METHODS: Eighty-eight subjects with medial knee OA (BMI <33) but without gout were included. Baseline sUA was measured in previously banked serum. At 0 and 24 months, subjects underwent standardized weight-bearing fixed-flexion posteroanterior knee radiographs to determine joint space width (JSW) and Kellgren-Lawrence (KL) grades. Joint space narrowing (JSN) was determined as JSW change from 0 to 24 months. Twenty-seven subjects underwent baseline contrast-enhanced 3T knee MRI for synovial volume (SV) assessment. RESULTS: sUA correlated with JSN in both univariate (r=0.40, p=0.01) and multivariate analyses (r=0.28, p=0.01). There was a significant difference in mean JSN after dichotomizing at sUA of 6.8 mg/dL, the solubility point for serum urate, even after adjustment (JSN of 0.90 mm for sUA>/=6.8; JSN of 0.31 mm for sUA<6.8, p<0.01). Baseline sUA distinguished progressors (JSN>0.2mm) and fast progressors (JSN>0.5mm) from nonprogressors (JSN=0.0mm) in multivariate analyses (area under the receiver operating characteristic curve 0.63, p=0.03; AUC 0.62, p=0.05, respectively). sUA correlated with SV (r=0.44, p<0.01), a possible marker of JSN, though this correlation did not persist after controlling for age, gender and BMI (r=0.13, p=0.56). CONCLUSIONS: In non-gout patients with knee OA, sUA predicted future JSN and may serve as a biomarker for OA progression
PMCID:5449226
PMID: 28217895
ISSN: 2326-5205
CID: 2460142
TARGETING A SELECTIVE POOL OF MRNAS TO INTERFERE WITH PROGRESSION OF OSTEOARTHRITIS (OA) [Meeting Abstract]
Katsara, O; Attur, M; Kolupaeva, V
ISI:000406888100257
ISSN: 1522-9653
CID: 2675522
KNEE OSTEOARTHRITIS IMPROVEMENT AND RELATED BIOMARKER PROFILES ARE SUSTAINED AT 24 MONTHS FOLLOWING BARIATRIC SURGERY [Meeting Abstract]
Chen, SX; Bomfim, F; Mukherjee, T; Wilder, E; Aharon, S; Toth, K; Browne, L; Vieira, RLa Rocca; Patel, J; Ren-Fielding, C; Parikh, M; Abramson, SB; Attur, M; Samuels, J
ISI:000406888100099
ISSN: 1522-9653
CID: 2675532
Cell contact-mediated response of fibroblast-like synoviocytes to cartilaginous debris [Meeting Abstract]
Silverstein, A M; Stefani, R M; Sobczak, E; Attur, M G; Shah, R P; Bulinski, J C; Ateshian, G A; Hung, C T
INTRODUCTION: The synovium of patients with osteoarthritis (OA), presenting with capsular synovial hyperplasia, has cartilage and bone debris bound to the surface or embedded within the tissue [1]. Arthroscopy with lavage and synovectomy can remove tissue debris from the joint space and the synovial lining to provide pain relief to patients with OA. Despite the results of six decades of preliminary animal model and in vitro studies, which have confirmed the deleterious interaction of cartilage wear particles with the synovium, the mechanisms that mediate this interaction remain poorly understood [2-4]. Cartilage wear particles are inherently more complex to study than their non-biologic counterparts, as they can interact with the cells through phagocytosis or integrin binding as well as undergo degradation by cells. In the current study, we hypothesize that the physical interaction between fibroblast-like synoviocytes (FLS) and cartilage wear particles is mediated by phagocytosis and surface binding. METHODS: Tissue Harvest: Synovial tissue and articular cartilage explants were harvested from bovine calf knees (n=3). Synovial tissue was digested with collagenase IV, and the isolated FLS were plated and expanded for two passages. Cartilage explants were maintained in serum free media until use [5]. Particulate Generation: Cartilage particles were generated by manual abrasion with 120 grit sandpaper. The cartilage particle solution was filtered with a 10mum nylon mesh filter to achieve desired particle size. Latex (~ 1mum) and cartilage wear particles (~0.9mum) were counted and sized as in [6]. Imaging: FLS were cultured with FITC labeled latex particles or cartilage particles labeled with dichlorotriazinyl aminofluorescein, fixed and counterstained with TRITC phalloidin (actin) and DAPI (nuclei) after 48 hours. Immunohistochemistry: High density FLS monolayers were cultured for 5 days, fixed and stained for type I collagen and lubricin and counterstained with DAPI. Monolayer Culture: FLS were plated at high density overnight and co-cultured with 250 latex or cartilage particles per cell for 5 days (n=6-8) [7]. Particles alone were also cultured. Conditioned Media: Dense monolayers were cultured +/- 250 cartilage particles per cell for 5 days. In parallel, acellular media only and particle only groups were cultured under the same conditions. Conditioned media was removed from the four original culture groups (media only, particles only, FLS only, FLS + particles). After filtering out the remaining particles, the conditioned media was supplemented with fresh media before application to recipient FLS monolayers for 5 days. Biochemistry: DNA, collagen (COL), nitric oxide (NO) and prostaglandin E2 (PGE2) were determined using the Picogreen, DMMB, OHP, Griess and PGE2 Parameter assays, respectively. Biochemical content of cartilage particles alone was subtracted from the measured sample values to determine the contribution of SF alone. Statistics: Comparisons were analyzed using ANOVA with Tukey's post-hoc test (p<0.05). RESULTS: Similar to the native synovium, FLS organized into a multi-layer cell sheets with positive staining for collagen type I and lubricin (Figure 1A,B,C). Both latex and cartilage particles were capable of being phagocytosed by FLS. Cartilage particles, not only appeared internalized, but attached to the surface of both dense monolayers and individual cells. Staining for the actin cytoskeleton revealed nuclear (internal) and cortical (external) remodeling of the actin cytoskeleton around cartilage wear particles (Figure 1D), compared to only internal remodeling of latex particulates (Figure 1E). Co-culture of FLS with cartilage particulates for 5 days resulted in a significant increase in cell sheet DNA and collagen content compared to control and latex treated groups (Figure 2A,B). Co-culture with cartilage wear particles induced an inflammatory response as measured by the up-regulation of both NO and PGE2 synthesis compared to both control and latex treated samples. Latex particles only had a deleterious effect on collagen synthesis (Figure 2B). No differences in proliferation or extracellular matrix (ECM) synthesis were observed between media only and particle + media groups and FLS + media only and particles + media + FLS. As such, the response of FLS to cartilage particles appears to require particulate surface binding and/or phagocytosis (Figure 3). DISCUSSION: In this study, we developed a 2D model of the synovium to study the interaction of cartilage wear particles with FLS. Based on our findings, we accept our hypothesis that the physical interaction between FLS and cartilage wear particles is mediated by phagocytosis and surface binding. Both latex and cartilage wear particles showed re-arrangement of the actin cytoskeleton around the wear particulate (Figure1 D,E) as has been observed previously with titanium particles [8]. Here, the actin cytoskeleton remodeled at the surface to form a U-shaped organization around the cartilage particle contact site (Figure 1D). Cartilage particles induced more proliferation and secreted more inflammatory factors than latex particles alone. The proliferative response of FLS to cartilage wear particles resulted in an overall increase in ECM content, representative of the thickening of the synovial lining observed in OA patients. Additional insight into the underlying mechanisms that mediate cartilage particulate-FLS interactions may be gleaned from literature studies that have looked at cellular activation through ECM binding to FLS integrins using ECM (e.g. collagen, fibronectin, etc.)-coated beads [9]. These studies have shown that binding to ECM-coated beads increases cellular proliferation and decreases collagenase synthesis, which supports the present results for FLS co-culture with cartilage wear particles (Figure 2). Parallel studies pursued by our laboratory are investigating how the interaction of cartilage wear particles and FLS are affected by physical loading, such as fluid shear that arises in situ from synovial fluid-synovium interactions during joint articulation. (Figure Presented)
EMBASE:616815192
ISSN: 1554-527x
CID: 2610282
Increased activity of chondrocyte translational apparatus accompanies osteoarthritis
Katsara, Olga; Attur, Mukundan; Ruoff, Rachel; Abramson, Steven B; Kolupaeva, Victoria
Objectives Degeneration of articular cartilage is central to OA pathology; however, the molecular mechanisms leading to these irreversible changes are still poorly understood. Here, we investigated how changes in the chondrocyte translational apparatus may contribute to the pathology of OA. Methods Normal and OA human knee cartilage was used to analyze the activity of different components of the translational machinery. Chondrocytes isolated from lesional and non-lesional areas of OA cartilage were used to estimate relative rate of protein synthesis by metabolic labeling. Experimental OA was induced by transection of the anterior cruciate ligament in rats to investigate changes in the translational apparatus associated with OA. The role of IL-1beta signaling was assessed in vitro using rat articular chondrocytes. Expression of mRNAs was analyzed by qPCR and protein levels by immunohistochemistry and Western blotting. Results We identified several novel traits of OA chondrocytes, including upregulation of the Serine/Threonine kinases AKT2 and AKT3 at the post-transcriptional level and increased rate of total protein synthesis, likely due to inactivation of 4E-BP1, a known repressor of cap-dependent translation. We found that 4E-BP1 inactivation is mTOR-dependent and crucial for upregulation of protein synthesis in general and in particular for MMP13 and ADAMTS5 expression. In addition, IL-1beta treatment led to 4E-BP1 inactivation and upregulation of protein synthesis in articular chondrocytes. Conclusions Precise control of protein synthesis is vital for cartilage homeostasis and its dysregulation contributes to the molecular pathology of OA. Our study therefore identifies a novel set of potential therapeutic targets
PMCID:5329137
PMID: 27696794
ISSN: 2326-5205
CID: 2273982