Network


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

Hotspot


Dive into the research topics where Glyn D. Palmer is active.

Publication


Featured researches published by Glyn D. Palmer.


Arthritis & Rheumatism | 2009

Interleukin-1β and tumor necrosis factor α inhibit chondrogenesis by human mesenchymal stem cells through NF-κB–dependent pathways†

N. Wehling; Glyn D. Palmer; Carmencita Pilapil; Fangjun Liu; James W. Wells; P. E. Müller; Christopher H. Evans; Ryan M. Porter

OBJECTIVE The differentiation of mesenchymal stem cells (MSCs) into chondrocytes provides an attractive basis for the repair and regeneration of articular cartilage. Under clinical conditions, chondrogenesis will often need to occur in the presence of mediators of inflammation produced in response to injury or disease. The purpose of this study was to examine the effects of 2 important inflammatory cytokines, interleukin-1beta (IL-1beta) and tumor necrosis factor alpha (TNFalpha), on the chondrogenic behavior of human MSCs. METHODS Aggregate cultures of MSCs recovered from the femoral intermedullary canal were used. Chondrogenesis was assessed by the expression of relevant transcripts by quantitative reverse transcription-polymerase chain reaction analysis and examination of aggregates by histologic and immunohistochemical analyses. The possible involvement of NF-kappaB in mediating the effects of IL-1beta was examined by delivering a luciferase reporter construct and a dominant-negative inhibitor of NF-kappaB (suppressor-repressor form of IkappaB [srIkappaB]) with adenovirus vectors. RESULTS Both IL-1beta and TNFalpha inhibited chondrogenesis in a dose-dependent manner. This was associated with a marked activation of NF-kappaB. Delivery of srIkappaB abrogated the activation of NF-kappaB and rescued the chondrogenic response. Although expression of type X collagen followed this pattern, other markers of hypertrophic differentiation responded differently. Matrix metalloproteinase 13 was induced by IL-1beta in a NF-kappaB-dependent manner. Alkaline phosphatase activity, in contrast, was inhibited by IL-1beta regardless of srIkappaB delivery. CONCLUSION Cell-based repair of lesions in articular cartilage will be compromised in inflamed joints. Strategies for enabling repair under these conditions include the use of specific antagonists of individual pyrogens, such as IL-1beta and TNFalpha, or the targeting of important intracellular mediators, such as NF-kappaB.


Journal of Immunology | 2008

Prostaglandin E2 Exerts Catabolic Effects in Osteoarthritis Cartilage: Evidence for Signaling via the EP4 Receptor

Mukundan Attur; H. Al-Mussawir; Jyoti C. Patel; Alison Kitay; M. Dave; Glyn D. Palmer; Michael H. Pillinger; Steven B. Abramson

Elevated levels of PGE2 have been reported in synovial fluid and cartilage from patients with osteoarthritis (OA). However, the functions of PGE2 in cartilage metabolism have not previously been studied in detail. To do so, we cultured cartilage explants, obtained from patients undergoing knee replacement surgery for advanced OA, with PGE2 (0.1–10 μM). PGE2 inhibited proteoglycan synthesis in a dose-dependent manner (maximum 25% inhibition (p < 0.01)). PGE2 also induced collagen degradation, in a manner inhibitable by the matrix metalloproteinase (MMP) inhibitor ilomastat. PGE2 inhibited spontaneous MMP-1, but augmented MMP-13 secretion by OA cartilage explant cultures. PCR analysis of OA chondrocytes treated with PGE2 with or without IL-1 revealed that IL-1-induced MMP-13 expression was augmented by PGE2 and significantly inhibited by the cycolooygenase 2 selective inhibitor celecoxib. Conversely, MMP-1 expression was inhibited by PGE2, while celecoxib enhanced both spontaneous and IL-1-induced expression. IL-1 induction of aggrecanase 5 (ADAMTS-5), but not ADAMTS-4, was also enhanced by PGE2 (10 μM) and reversed by celecoxib (2 μM). Quantitative PCR screening of nondiseased and end-stage human knee OA articular cartilage specimens revealed that the PGE2 receptor EP4 was up-regulated in OA cartilage. Moreover, blocking the EP4 receptor (EP4 antagonist, AH23848) mimicked celecoxib by inhibiting MMP-13, ADAMST-5 expression, and proteoglycan degradation. These results suggest that PGE2 inhibits proteoglycan synthesis and stimulates matrix degradation in OA chondrocytes via the EP4 receptor. Targeting EP4, rather than cyclooxygenase 2, could represent a future strategy for OA disease modification.


Osteoarthritis and Cartilage | 2008

Current concepts in the pathogenesis of osteoarthritis

Svetlana Krasnokutsky; Mukundan Attur; Glyn D. Palmer; Jonathan Samuels; Steven B. Abramson

Osteoarthritis (OA) is a degenerative joint disease that progressively causes loss of joint function and is the leading source of physical disability and impaired quality of life in industrialized nations. The burden of disease dramatically impacts health care usage and leads to total joint replacement in approximately a half-million Americans alone each year e and such consequences on society worldwide are expected to rise in coming decades with the continued expanding and aging population. There are no current interventions proven to restore cartilage or curtail the disease processes. Thus, OA often ultimately results in joint destruction, chronic pain, disability, depression and social isolation. Multiple etiologic risk factors and pathophysiologic processes all contribute to the progressive nature of the disease e and serve as targets of behavioral and pharmacologic interventions. Risk factors, such as age, gender, trauma, overuse, genetics and obesity each make contributions to initiate the process of injury in different components of the joint; then the effector biochemical processes involving the cartilage, bone, and synovium eventually intertwine and collectively damage all three components as well (Fig. 1). These effects on the tissues of all three joint compartments manifest as articular cartilage breakdown, osteophyte formation, subchondral sclerosis, bone marrow lesions and alterations of the synovium on both morphologic and biochemical levels often causing episodic synovitis. Thus, the molecular and cytokine-based events that drive joint damage in inflammatory arthritides have gradually emerged as pathogenic paradigms in OA, and will be highly relevant to the development of future OA therapeutics. With increasing appreciation of the contribution of all three joint compartments to disease progression, current research in OA pathogenesis, biomarkers and treatment has broadened immensely in recent years. In this review, we will focus on emerging pathogenic concepts that will hopefully help advance the search for effective disease-modifying osteoarthritis drugs (DMOADs).


Gene Therapy | 2004

Gene delivery to cartilage defects using coagulated bone marrow aspirate

Arnulf Pascher; Glyn D. Palmer; Andre F. Steinert; Thomas Oligino; Elvire Gouze; Jean Noel Gouze; Oliver B. Betz; Myron Spector; Paul D. Robbins; Christopher H. Evans; S. C. Ghivizzani

The long-term goal of the present study is to develop a clinically applicable approach to enhance natural repair mechanisms within cartilage lesions by targeting bone marrow-derived cells for genetic modification. To determine if bone marrow-derived cells infiltrating osteochondral defects could be transduced in situ, we implanted collagen–glycosaminoglycan (CG) matrices preloaded with adenoviral vectors containing various marker genes into lesions surgically generated in rabbit femoral condyles. Analysis of the recovered implants showed transgenic expression up to 21 days; however, a considerable portion was found in the synovial lining, indicating leakage of the vector and/or transduced cells from the matrix. As an alternative medium for gene delivery, we investigated the feasibility of using coagulated bone marrow aspirates. Mixture of an adenoviral suspension with the fluid phase of freshly aspirated bone marrow resulted in uniform dispersion of the vector throughout, and levels of transgenic expression in direct proportion to the density of nucleated cells in the ensuing clot. Furthermore, cultures of mesenchymal progenitor cells, previously transduced ex vivo with recombinant adenovirus, were readily incorporated into the coagulate when mixed with fresh aspirate. These vector-seeded and cell-seeded bone marrow clots were found to maintain their structural integrity following extensive culture and maintained transgenic expression in this manner for several weeks. When used in place of the CG matrix as a gene delivery vehicle in vivo, genetically modified bone marrow clots were able to generate similarly high levels of transgenic expression in osteochondral defects with better containment of the vector within the defect. Our results suggest that coagulates formed from aspirated bone marrow may be useful as a means of gene delivery to cartilage and perhaps other musculoskeletal tissues. Cells within the fluid can be readily modified with an adenoviral vector, and the matrix formed from the clot is completely natural, native to the host and is the fundamental platform on which healing and repair of mesenchymal tissues is based.


Molecular Therapy | 2003

Lentiviral-mediated gene delivery to synovium: potent intra-articular expression with amplification by inflammation

Elvire Gouze; Robert Pawliuk; Jean Noel Gouze; Carmencita Pilapil; Christina Fleet; Glyn D. Palmer; Christopher H. Evans; Philippe Leboulch; Steven C. Ghivizzani

Clinical translation of gene-based therapies for arthritis could be accelerated by vectors capable of efficient intra-articular gene delivery and long-term transgene expression. Previously, we have shown that lentiviral vectors transduce rat synovium efficiently in vivo. Here, we evaluated the functional capacity of transgene expression provided by lentiviral-mediated gene delivery to the joint. To do this, we measured the ability of a lentiviral vector containing the cDNA for human interleukin-1 receptor antagonist (LV-hIL-1Ra) to suppress intra-articular responses to IL-1beta. Groups of rats were injected in one knee with 5 x 10(7) infectious units of LV-IL-1Ra. After 24 h, a range of doses of fibroblasts (3 x 10(3), 10(4), 3 x 10(4), or 10(5) cells) genetically modified to overexpress IL-1beta was injected into both knees. Intra-articular delivery of LV-hIL-1Ra strongly prevented swelling in all treated knees, even in those receiving the greatest dose of IL-1beta(+) cells. Cellular infiltration, cartilage erosion, and invasiveness of inflamed synovium were effectively prevented in LV-hIL-1Ra-treated knees and were significantly inhibited in contralateral joints. Beneficial effects were also observed systemically in the lentivirus-treated animals. Interestingly, intra-articular expression of the IL-1Ra transgene was found to increase in relation to the number of IL-1beta(+) cells injected. Further experiments using GFP suggest this is due to the proliferation of cells, stably modified by the integrative lentivirus, in response to inflammatory stimulation.


Arthritis & Rheumatism | 2008

The antioxidant resveratrol protects against chondrocyte apoptosis via effects on mitochondrial polarization and ATP production

M. Dave; Mukundan Attur; Glyn D. Palmer; H. Al-Mussawir; Lauren Kennish; Jyoti C. Patel; Steven B. Abramson

OBJECTIVE To determine the effects of the antioxidant resveratrol on the functions of human chondrocytes in osteoarthritis (OA). METHODS Chondrocytes and cartilage explants were isolated from OA patients undergoing knee replacement surgery. Effects of resveratrol in the presence or absence of interleukin-1beta (IL-1beta) stimulation were assessed by measurement of prostaglandin E(2) (PGE(2)) and leukotriene B(4) (LTB(4)) synthesis, cyclooxygenase (COX) activity, matrix metalloproteinase (MMP) expression, and proteoglycan production. To explore the mechanisms of action of resveratrol, its effects on mitochondrial function and apoptosis were examined by assessing mitochondrial membrane potential, ATP levels, cytochrome c release, and annexin V staining. RESULTS Resveratrol inhibited both spontaneous and IL-1beta-induced PGE(2) production by >20% (P < 0.05) and by 80% (P < 0.001), respectively; similarly, LTB(4) production was reduced by >50% (P < 0.05). The production of PGE(2) was inhibited via a 70-90% suppression of COX-2 expression and enzyme activity (P < 0.05). Resveratrol also promoted anabolic effects in OA explant cultures, by elevating proteoglycan synthesis and decreasing production of MMPs 1, 3, and 13. Pretreatment of OA chondrocytes with resveratrol blocked mitochondrial membrane depolarization, loss of mitochondrial biomass, and IL-1beta-induced ATP depletion. Similarly, IL-1beta-mediated induction of the apoptotic markers cytochrome c and annexin V was also inhibited by resveratrol. Exogenous addition of PGE(2) abolished the protective effects of resveratrol on mitochondrial membrane integrity, ATP levels, expression of apoptotic markers, and DNA fragmentation. CONCLUSION Resveratrol protects against IL-1beta-induced catabolic effects and prevents chondrocyte apoptosis via its inhibition of mitochondrial membrane depolarization and ATP depletion. These beneficial effects of resveratrol are due, in part, to its capacity to inhibit COX-2-derived PGE(2) synthesis. Resveratrol may therefore protect against oxidant injury and apoptosis, which are main features of progressive OA.


Journal of Biological Chemistry | 2010

Foxo1, a Novel Regulator of Osteoblast Differentiation and Skeletogenesis

Cristina C. Teixeira; Yuexun Liu; Lwin Mon Thant; Jason T. Pang; Glyn D. Palmer; Mani Alikhani

Skeletogenesis depends on the activity of bone-forming cells derived from mesenchymal cells. The pathways that control mesenchymal cell differentiation are not well understood. We propose that Foxo1 is an early molecular regulator during mesenchymal cell differentiation into osteoblasts. In mouse embryos, Foxo1 expression is higher in skeletal tissues, while Foxo1 silencing has a drastic impact on skeletogenesis and craniofacial development, specially affecting pre-maxilla, nasal bone, mandible, tibia, and clavicle. Similarly, Foxo1 activity and expression increase in mouse mesenchymal cells under the influence of osteogenic stimulants. In addition, silencing Foxo1 blocks the expression of osteogenic markers such as Runx2, alkaline phosphatase, and osteocalcin and results in decreased culture calcification even in the presence of strong osteogenic stimulants. Conversely, the expression of these markers increases significantly in response to Foxo1 overexpression. One mechanism through which Foxo1 affects mesenchymal cell differentiation into osteoblasts is through regulation of a key osteogenic transcription factor, Runx2. Indeed, our results show that Foxo1 directly interacts with the promoter of Runx2 and regulates its expression. Using a tibia organ culture model, we confirmed that silencing Foxo1 decreases the expression of Runx2 and impairs bone formation. Furthermore, our data reveals that Runx2 and Foxo1 interact with each other and cooperate in the transcriptional regulation of osteoblast markers. In conclusion, our in vitro, ex vivo, and in vivo results strongly support the notion that Foxo1 is an early molecular regulator in the differentiation of mesenchymal cells into osteoblast.


The FASEB Journal | 2009

F-spondin, a neuroregulatory protein, is up-regulated in osteoarthritis and regulates cartilage metabolism via TGF-β activation

Mukundan Attur; Glyn D. Palmer; H. Al-Mussawir; M. Dave; Cristina C. Teixeira; Daniel B. Rifkin; C. Thomas G. Appleton; Frank Beier; Steven B. Abramson

In osteoarthritis (OA) articular chondrocytes undergo phenotypic changes culminating in the progressive loss of cartilage from the joint surface. The molecular mechanisms underlying these changes are poorly understood. Here we report enhanced (‐7‐fold) expression of F‐spondin, a neuronal extracellular ma‐trix glycoprotein, in human OA cartilage (P<0.005). OA‐specific up‐regulation of F‐spondin was also dem‐onstrated in rat knee cartilage following surgical meni‐sectomy. F‐spondin treatment of OA cartilage explants caused a 2‐fold increase in levels of the active form of TGF‐β1(P<0.01) and a 10‐fold induction of PGE2 (P< 0.005) in culture supernatants. PGE2 induction was found to be dependent on TGF‐β and the throm‐bospondin domain of the F‐spondin molecule. F‐spondin addition to cartilage explant cultures also caused a 4‐fold increase in collagen degradation (P< 0.05) and a modest reduction in proteoglycan synthesis (~20%;P<0.05), which were both TGF‐β and PGE2 dependent. F‐spondin treatment also led to increased secretion and activation of MMP‐13 (P<0.05). Together these studies identify F‐spondin as a novel protein in OAcartilage, where it may act in situ at lesional areas to activate latent TGF‐β and induce cartilage degradation via pathways that involve production of PGE2.—Attur, M. G., Palmer, G. D., Al‐Mussawir, H. E., Dave, M., Teixeira, C. C., Rifkin, D. B., Appleton, C. T. G., Beier, F., Abramson, S. B. F‐spondin, a neuroregulatory protein, is up‐regulated in osteoarthritis and regulates cartilage metabolism via TGF‐β activation. FASEB J. 23, 79‐89 (2009)


Methods in molecular medicine | 2004

In vitro gene transfer to chondrocytes and synovial fibroblasts by adenoviral vectors.

Jean Noel Gouze; Martin J. Stoddart; Elvire Gouze; Glyn D. Palmer; Steven C. Ghivizzani; Alan J. Grodzinsky; Christopher H. Evans

The major requirement of a successful gene transfer is the efficient delivery of an exogenous therapeutic gene to the appropriate cell type with subsequent high or regulated levels of expression. In this context, viral systems are more efficient than nonviral systems, giving higher levels of gene expression for longer periods. For the application of osteoarthritis (OA), gene products triggering anti-inflammatory or chondroprotective effects are of obvious therapeutic utility. Thus, their cognate genes are candidates for use in the gene therapy of OA. In this chapter, we describe the preparation, the use, and the effect of the transduction of chondrocytes or synovial fibroblasts with an adenoviral vector encoding the cDNA for glutamine: fructose-6-phosphate amidotransferase (GFAT). This is intended to serve as an example of a technology that can be used to evaluate the biological effects of overexpression of other cDNAs.


Expert Opinion on Biological Therapy | 2001

Gene therapy for rheumatoid arthritis

Elvire Gouze; Steven C. Ghivizzani; Glyn D. Palmer; Jean Noel Gouze; Paul D. Robbins; Christopher H. Evans

Rheumatoid arthritis (RA) is a disabling, painful disorder affecting 1% of the world’s population. Although the aetiology of RA remains unknown, recent advances in understanding its pathophysiology have led to the characterisation of several proteins whose activities may be anti-arthritic. Clinical application of such proteins has greatly improved the treatment of RA, but the disease remains incurable and difficult to manage in a substantial number of patients. Thus, there are continued efforts to develop new therapeutic strategies. Because RA is a chronic condition, effective treatment will probably require the presence of therapeutic agents for extended periods of time. In the case of proteins, this is problematic. Gene therapy may offer a solution to this problem. Experimental studies have confirmed the feasibility, efficacy and safety of gene therapy for the treatment of animal models of arthritis. Several different approaches have shown promise in this regard, including gene transfer to the synovial lining cells of individual joints and the systemic delivery of genes to extra-articular locations. One unexpected finding has been the ‘contralateral effect’ in which gene delivery to one joint of an animal with polyarticular disease leads to improvement of multiple joints. Investigation of this phenomenon has led to interest in cell trafficking and the genetic modification of antigen-presenting cells (APC). The first Phase I clinical trial tested the feasibility and safety of ex vivo gene transfer to the synovial lining of human joints. This clinical trial has been successfully completed and two other Phase I trials are in progress. A Phase II study is now being planned to investigate the efficacy of gene transfer to the joints of patients with early stage RA.

Collaboration


Dive into the Glyn D. Palmer's collaboration.

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

Oliver B. Betz

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge