Jump to content

Triamcinolone-40 injection hospitalize patient


Recommended Posts

100 yo male with CKD, High cholesterol, HTN, Hypotension (SBP of 80 currently receiving colloids) with an INR of 1.13 presented to our service for bilateral knee effusions and severe bilateral knee pain secondary to severe osteoarthritis (he's 100...) Previous hx of effusions x 1yr which were aspirated, improving his pain. No documentation of Kenalog received. No history of gout. X-rays show large right knee effusion, small left knee effusion, complete joint space collapse bilaterally. Mobile prior to hospitalization with a 2WW. 

 

I aspirated 40cc of blood tinged fluid from the right knee and 20cc from the left knee. Appeared non-pyogenic sent for routine fluid analysis and did not administer any steroid. 

 

Steroid or no steroid? I did not. Looking to hear what you would do. Given the circumstances and his age I erred on the side of caution with the side effects. 

 

Other info:

BG trending down from 122 to 98 today

WBC hovering x 5 days at 13.22

Cr  2.18

Link to comment
Share on other sites

  • Moderator

That's tough one. I would discuss with him all the options and the high chance that steroids could put him in even worse spot. His kidney disease and age really puts him in a corner for recurrent/severe attack therapies. At this point it's pain control, if I were him. Hopefully get lucky and maybe the analysis will show something you can treat better.

Link to comment
Share on other sites

I've looked per Dr. Google for evidence of long-term benefit from a single steroid injection and there is some evidence of short term benefit.  My question is why?  The disease process itself has been shown to be non-inflammatory in cadavers (this is why Tylenol Arthritis is allowed to be sold/marketed as such).  If this is the case then what's the mechanism of action aside from placebo effect which is not to be overlooked?  Secondary inflammation to soft tissue structures is all I could come up with.

Link to comment
Share on other sites

  • Moderator

I've looked per Dr. Google for evidence of long-term benefit from a single steroid injection and there is some evidence of short term benefit. My question is why? The disease process itself has been shown to be non-inflammatory in cadavers (this is why Tylenol Arthritis is allowed to be sold/marketed as such). If this is the case then what's the mechanism of action aside from placebo effect which is not to be overlooked? Secondary inflammation to soft tissue structures is all I could come up with.

Plenty of info on uptodate if you have it. It's the standard of care to treat OA with intra-articular glucorticoids.

 

 

From uptodate

 

 

Efficacy of intraarticular glucocorticoids — The knee is the best-studied joint, with multiple trials demonstrating benefit of intraarticular glucocorticoids, usually compared with placebo [21,39]. However, more limited evidence has suggested efficacy in other joints, including the hip [43,44]. Clinically evident inflammation does not need to be present for intraarticular glucocorticoids to result in symptom relief [45]. The following examples illustrate the potential benefit of these agents:

 

●In a meta-analysis of 28 trials involving 1973 patients, intraarticular glucocorticoids were more effective at reducing pain than intraarticular placebo injections at one week following knee injection and were more likely to lead to improvement in patient global assessment [39]. Significantly more patients reported reduced pain after receiving glucocorticoids at two to three weeks (RR 1.81, 95% CI 1.09-3.00, and RR 3.11, 95% CI 1.61-6.01), but not at 4 to 24 weeks; there was no evidence of significant functional improvement following injection

●In larger 2015 network meta-analysis of 137 trials involving over 33,000 patients with knee OA, intraarticular glucocorticoids were also found to be more effective at reducing pain than intraarticular placebo after a follow-up period of three months [15].

The efficacy of glucocorticoid injections at sites other than the knee or hip is less well established. A prospective case series of injections of the carpometacarpal (CMC) joint in 25 patients showed significant improvement in pain at one month, but not at 3, 6, or 12 months following injection [46]. A randomized trial involving 40 patients with CMC joint arthritis assigned to receive a single injection of triamcinolone hexacetonide (0.25 mL) or saline failed to show greater benefit from the glucocorticoid injection, although both groups had decreased pain [47]. We use CMC joint glucocorticoid injections in patients who have not responded adequately to nonpharmacologic interventions based upon our clinical experience, which is supported by the opinion of other experts and the limited trial data available [5].

Link to comment
Share on other sites

  • Moderator

Yep, I found similar findings but it still doesn't explain the why.

 

It is an inflammatory process. Just different from other types.

 

 

Author:Richard F. Loeser, MDSection Editor:David Hunter, MD, PhDDeputy Editor:Monica Ramirez Curtis, MD, MPH

Contributor Disclosures

 

All topics are updated as new evidence becomes available and our peer review process is complete.

Literature review current through: Aug 2016. | This topic last updated: Jun 21, 2016.

INTRODUCTION — In the past, osteoarthritis (OA) was considered to be simply a degenerative “wear and tear” process and therefore often misnamed as degenerative joint disease. However, the pathogenesis of OA is much more complex than just wear and tear and the term “osteoarthritis,” where “-itis” is indicative of an inflammatory process, is indeed correct [1,2]. There are a variety of factors that play an important role in the pathogenesis of OA, including biomechanical factors, proinflammatory mediators, and proteases. By understanding the mechanisms driving joint tissue destruction in OA and identifying the key factors involved, new targets for therapy are emerging that will go beyond symptomatic relief to slowing or stopping the progression of OA [3].

 

This topic will review the pathogenesis of OA. The diagnosis, treatment, and other issues related to OA are discussed separately. (See "Initial pharmacologic therapy of osteoarthritis" and "Treatment of osteoarthritis resistant to initial pharmacologic therapy" and "Nonpharmacologic therapy of osteoarthritis" and "Overview of surgical therapy of knee and hip osteoarthritis" and "Investigational approaches to the management of osteoarthritis" and "Clinical manifestations and diagnosis of osteoarthritis".)

 

ROLE OF INFLAMMATION — Classically, inflammatory arthritis was defined in part based on cellular inflammation represented by increased numbers of leukocytes in the affected joint tissues and synovial fluid. Classic cellular inflammation is not prominent in osteoarthritis (OA), where the number of leukocytes in the joint fluid is normally low, and rarely exceeds 1000 to 2000 cells per milliliter. This is in contrast to forms of inflammatory arthritis, such as rheumatoid arthritis (RA), where the number of synovial fluid leukocytes will commonly exceed 2000 and will be accompanied by a more extensive synovial infiltrate of leukocytes with synovial fibroblast proliferation resulting in pannus formation. Although synovial inflammation is also present in OA and in some individuals can be indistinguishable from RA, the inflammatory component of OA is best appreciated at the molecular level and is characterized by the presence of a host of proinflammatory mediators, including cytokines and chemokines, that are part of an innate immune response to joint injury [2].

 

As will be further discussed below, proinflammatory factors appear to be driving the production of the proteolytic enzymes responsible for the degradation of the extracellular matrix that results in joint tissue destruction. Although destruction and loss of the articular cartilage is a central component of OA, all joint tissues are affected in some way, indicating that OA is a disease of the joint as an organ [4]. Mechanical factors certainly play a key role in OA and there is some debate in the field as to the extent to which OA is mediated by abnormal joint mechanics. However, the balance of evidence suggests that rather than simply causing joint tissue damage by wear and tear, excessive or abnormal joint loading also stimulates joint tissue cells to produce proinflammatory factors and proteases that mediate joint tissue destruction. (See 'Inflammatory mediators' below and 'Proteases' below.)

 

OVERVIEW OF PATHOLOGY — Osteoarthritis (OA) is one of the most common causes of chronic disability in adults due to pain and altered joint function that result from characteristic pathologic changes in the joint tissues and their processing in a biopsychosocial context (figure 1). The pathological findings described below are present to varying degrees in all people with OA, suggesting a common response of the joint to a variety of insults.

 

The order in which particular joint tissues are affected may depend on the initiating factors. With the exception of posttraumatic OA that starts with an acute injury to a key joint tissue component, such as a ligament tear, it is often difficult to know exactly which joint tissues are affected first. Plain radiographs underestimate the joint tissue involvement in OA since they only visualize a component of the condition including cartilage loss that results in joint space narrowing and bony changes that result in subchondral sclerosis, cysts, and osteophyte formation. Once these changes are apparent on radiographs, the condition has significantly advanced. Magnetic resonance imaging (MRI) studies can detect early disease and have provided evidence of matrix changes in cartilage, synovitis, bone marrow lesions, and degenerative changes in soft-tissue structures beyond the cartilage including ligaments and the knee menisci [4,5]. As OA progresses, it eventually affects the entire joint, resulting in failure of the component parts. However, OA does not progress to at a similar rate in all individuals and not everyone with early disease will develop more severe OA. Predicting which patients will advance to the end stages of the disease remains a challenge.

 

●Articular cartilage – Although commonly thought of as a “shock absorber,” articular cartilage primarily serves to provide a smooth, low-friction surface that allows for the normal gliding motion of the joint. Most of the load on the joint is absorbed by other tissues, including periarticular muscles and subchondral bone and, in the knee, the meniscus. Hyaluronic acid is the substance in the synovial fluid that provides viscosity, but it requires the presence of a large mucinous protein called lubricin (also known as proteoglycan-4 or superficial zone protein) to provide a low-friction state and protect the joint surface, including the superficial zone chondrocytes, from shear stresses [6]. The collagen fibers provide the tensile strength and form a network that restrains the very hydrophilic proteoglycans that provide resiliency.

The earliest pathologic changes in OA are commonly seen at the surface of the articular cartilage with fibrillation in focal regions that experience maximal loading. The cartilage initially swells as the collagen network loosens allowing the hydrophilic proteoglycans to attract water and expand. Chondrocytes, the only cell type present in cartilage, are normally quiescent. Chondrocytes are active cells maintaining cartilage through normal anabolic/catabolic activities. As OA develops, this activity dramatically accelerates: chondrocytes proliferate (to a modest degree) and form clusters, likely in response to loss of matrix. At least a portion of the cells undergo a phenotypic switch to a hypertrophic chondrocyte which is similar to the cells found in the hypertrophic zone of the growth plate that produce type X collagen and matrix metalloproteinase (MMP)-13. As OA progresses, extensive matrix degradation and loss occurs due to the continued production of proteases driven by proinflammatory cytokines and fragments of matrix proteins that feedback and stimulate chondrocytes in an autocrine and paracrine manner to produce more cytokines and proteases. As significant matrix damage occurs, chondrocyte death can be seen, resulting in areas of matrix devoid of cells.

●Bone – Thickening of the subchondral bone (bone sclerosis) occurs due to increased production of collagen that is improperly mineralized. Osteophytes (bony spurs) form at the joint margins, often at the insertion site of tendons or ligaments. In more advanced disease, bone cysts occur but bone erosions are not typically seen. An exception to the latter is erosive OA that most commonly noted in the distal joints of the hands (distal interphalangeals and proximal interphalangeals) and is associated with centrally located erosions that differ in location from the marginal erosions seen in rheumatoid arthritis (RA) and gout. Bone marrow lesions, evident on MRI, can be seen most commonly in areas with overlying cartilage loss and where mechanical loads are greatest. Pathologically, these focal lesions consist of microstructural damage to bone accompanied by localized necrosis and fibrosis [7].

●Synovium – Most people with symptomatic OA will exhibit some degree of synovial inflammation (synovitis) and/or synovial hypertrophy during the course of OA [8,9]. Unlike RA and other forms of so-called inflammatory arthritis, synovitis is not thought to be the initiating factor in primary OA. However, synovitis contributes to pain and disease progression, including cartilage destruction, mediated by the production of proinflammatory factors and proteins referred to as damage-associated molecular patterns (DAMPs), including the alarmins [2,4]. Secondary OA can be seen in joints previously affected by inflammatory arthritis, although the pathology is somewhat different from that of primary OA due to the prior effects of a more striking inflammatory component that causes more extensive joint destruction including bone erosions.

●Soft tissues – In addition to the cartilage, soft-tissue components of the joint, including ligaments, the joint capsule, and, in the knee, the menisci, are commonly affected by OA. These tissues exhibit disruption of their extracellular matrix and loss of cells. Thickening of the joint capsule along with osteophytes contribute to the enlargement observed in OA joints. In older adults with established OA, it is quite common to find tears in ligaments and the meniscus, which, without a history of prior joint injury, are most likely degenerative in nature [10]. In addition to the effects of meniscal tears on joint mechanics, studies have shown that torn menisci can be a source of inflammatory mediators in the joint [11]. Periarticular muscles and nerves are also affected by OA resulting in weakness and pain [12].

MULTIPLE PATHWAYS TO OSTEOARTHRITIS — The pathological changes in osteoarthritis (OA) joints described above are commonly present, particularly in advanced stages of the disease, no matter the inciting factor (see 'Overview of pathology' above). However, the pathway from initiating factors to disease can vary. This has led some in the field to think of OA as a spectrum of conditions or different OA phenotypes rather than a single disease. We discuss several factors below that are thought to play a role in the pathogenesis of OA.

 

Risk factors — Multiple risk factors have been linked to the pathogenesis of OA. Risk factors for OA include age, joint injury, obesity, genetics, anatomical factors including joint shape and alignment, and gender [13].

 

●Aging – Because OA is most clearly a condition associated with aging, a separate section below will review the aging changes that promote the development of OA. (See 'Aging' below.)

●Joint injury – OA that develops after injury to a joint is commonly called posttraumatic OA. The pathologic changes are often evident within 10 years after injury, with the time of onset influenced in part by the age of the individual at the time of injury [14]. OA can develop after injuries that result in ligament and/or meniscal tears, or after injuries such as fractures that involve the joint. Tears incite acute inflammation with joint swelling that is particularly severe when a major ligament, such as the anterior cruciate ligament (ACL), is torn. Studies have shown a host of inflammatory mediators, including tumor necrosis factor (TNF)-alpha (elevated sixfold) and interleukin (IL)-6 (elevated 1000-fold), are present shortly after injury [15]. The risk of developing OA after an ACL tear is the same whether the ligament is repaired or not [16]. This suggests that either the mechanics of the joint are not completely restored after ACL reconstruction or that the acute inflammation that occurs with the tear puts the OA process in motion and it is not stopped by reconstruction of the ligament. The latter is supported by studies demonstrating that markers of collagen and proteoglycan degradation are present acutely after injury and sustained over time [17].

●Obesity – Body weight is a risk factor for OA not only in weightbearing joints including the knee and hip, but also in the hand [18,19]. Excess weight will certainly produce increased load on the joint, but there is growing evidence for a metabolic contribution to OA. This would explain the association of obesity with hand OA and why not all overweight and obese individuals develop knee or hip OA [20]. Macrophages within adipose tissue are a source of proinflammatory cytokines, including IL-6 and TNF-alpha, and adipocytes produce adipokines such as leptin. The cytokines associated with obesity may promote a low-grade, systemic, proinflammatory state that could contribute to the development of OA, while leptin has been proposed to have direct effects on joint tissues that promote the development of OA [20]. Metabolic OA related to obesity may also involve altered lipid metabolism and increased activity of peroxisome proliferator-activated receptor (PPAR)-delta [21].

●Genetics – Heredity forms of OA due to certain rare mutations in collagen types II, IX, or XI, which are structural collagens found in articular cartilage, result in premature OA that can begin as early as adolescence, resulting in a severe destructive form of arthritis that affects multiple joints [22,23]. Because the vitreous of the eye also contains these collagen types, some patients also have eye disease. These mutations are causes of Stickler syndrome which affects 1 in 7500 to 9000 newborns (see "Syndromes with craniofacial abnormalities", section on 'Stickler and Marshall syndromes'). Less severe forms of OA also have a genetic component which, from twin studies, has been estimated to explain approximately 40 percent of the risk for OA [24]. Several risk loci have been discovered by genome wide-association studies with perhaps the most consistent association being found for polymorphisms in the gene that codes for growth and differentiation factor (GDF)-5, which is a bone morphogenetic family member that plays a role in joint development [24]. Mutations in GDF-5 are thought to predispose to OA due to altered joint shape. Genetic studies (other than the rare collagen mutations mentioned above) have found that individual genes provide a very slight increase in the risk for OA (odds ratios in the 1.2-1.4 range), suggesting that either multiple genes are needed for a more substantial OA risk or that environmental factors and/or epigenetics are important. (See 'Other pathways' below.)

●Anatomic factors – Joint shape, particularly of the hip, can influence the development of OA. Congenital acetabular dysplasia is associated with premature hip OA that often requires joint replacement. An important anatomic factor related to knee OA is lower-extremity alignment. Individuals who have a varus alignment (bow-legged) are at increased risk of medial tibial-femoral OA, while those with a valgus alignment (knocked-knee) are at risk for lateral tibial-femoral OA [25]. The relationship of anatomic factors to OA is best explained by altered joint mechanics as the initiating cause for OA. Altered mechanics that place excessive and abnormal loads on joint tissue cells activate mechanotransduction pathways that result in increased production of inflammatory mediators and proteolytic enzymes [26].

●Gender – OA of the hands and knees is more common in women than men, while hip OA is equally prevalent [19]. The strong association of OA with age could explain why OA is more common in the postmenopausal years, although there is some evidence that loss of estrogen could be a contributing factor [27,28].

Aging — OA is clearly related to aging, with both the incidence and prevalence of OA increasing with age [29]. However, it is also clear that aging of joint tissues and the development of OA are distinct processes. Rather than being one and the same, aging changes most likely make the joint more susceptible to the development of OA and promote progression. The aging changes within the joint that contribute to OA can be divided into aging of the extracellular matrix and cellular aging. Matrix changes include thinning of the articular cartilage with age, reduced hydration, and an accumulation of proteins containing advanced glycation end-products (AGEs) [30]. AGEs cause increased crosslinking of collagen, resulting in altered biomechanical properties characterized by increased “brittleness” [31]. AGEs are best known for their role in diabetes, where their production is facilitated by chronic elevations in glucose (think hemoglobin A1c, which is glycosylated hemoglobin). In cartilage, AGEs can form and accumulate independent of blood glucose levels. The most likely explanation is the very long half-life of matrix proteins in cartilage, particularly type II collagen, which has a half-life calculated at over 100 years [32]. The low turnover rate in cartilage allows for a very slow accumulation of AGEs that are not removed as they would be in tissues that have a higher turnover of their matrix such as bone.

 

Another age-related matrix change seen in cartilage, as well as the meniscus, is abnormal calcification referred to as “chondrocalcinosis.” Chondrocalcinosis is most commonly results from the accumulation of calcium pyrophosphate dihydrate (CPP) crystals, although basic calcium phosphate and hydroxyapatite crystals may also play a role [33]. Chondrocalcinosis is associated with episodes of acute crystal-induced mono- or oligoarticular arthritis or pseudogout that is most commonly due to CPPD crystals. It is thought, although still debated, that chondrocalcinosis could contribute to the development of OA [34]. Chondrocalcinosis is often seen in joints which also have radiographic evidence of OA; abnormal calcification could alter the mechanical properties of joint tissues, and crystal-induced inflammation that results from activation of the innate immune system in joint tissues could contribute to OA by stimulating production of proinflammatory mediators.

 

There are a host of cellular changes that can link aging and OA. These include mitochondrial dysfunction related to oxidative stress and mitochondrial DNA damage, reduced responsiveness to anabolic growth factor stimulation including insulin-like growth factor (IGF)-1 and transforming growth factor (TGF)-beta, cell senescence that results in the senescence-associated secretory phenotype, and a reduction in the process called autophagy which is a protective mechanism responsible for the degradation and removal of damaged cellular constituents [30]. These cellular changes contribute to an imbalance between anabolic activity mediated by growth factors that is necessary to produce and repair damaged matrix and catabolic activity mediated by proinflammatory mediators and proteases that promote joint tissue destruction. (See 'Inflammatory mediators' below and 'Proteases' below.)

 

Inflammatory mediators — Inflammatory mediators may play a role in the pathogenesis of OA as potential drivers of joint tissue destruction. The list of proinflammatory mediators found in the synovial fluid and tissues affected by OA continues to grow. Early studies focused on the role of the cytokine IL-1 which was initially named “catabolin” due to its ability to stimulate cartilage catabolic activity resulting in matrix degradation. The role of IL-1 in OA has been questioned because the levels found in OA joints (in the pg/mL range) are much lower than the levels required to cause cartilage degradation (ng/mL range). Other cytokines are present in OA synovial fluid at much greater levels than IL-1 or TNF-alpha, including IL-6, macrophage chemotactic protein (MCP)-1, interferon-induced protein (IP)-10, and monokine induced by interferon (MIG) [35]. Data from genetically modified mice have shown inconsistent protection from surgically induced OA in IL-1beta knockouts while IL-6 knockouts were protected from injury-induced OA but not age-associated OA, which was worse than the controls [36].

 

A number of other cytokines as well as chemokines have been found in synovial fluid or noted to be produced by articular chondrocytes and/or meniscal cells, including IL-7, IL-8, IL-15, IL-17, IL-18, oncostatin M (OSM), growth-related oncogene (GRO)-alpha, chemokine (C-C-motif) ligand 19 (CCL19), macrophage inflammatory protein (MIP)-1beta, and TGF-alpha [2,4]. Other inflammatory mediators include the alarmins (S100 proteins) and the damage-associated molecular patterns (DAMPs). These mediators promote synovitis by attracting macrophages into the joint and promote matrix degradation by stimulating the expression of various proteases (see 'Proteases' below). Surprisingly, some studies have also provided evidence for complement activation in OA joints. Inhibition of complement activation by gene deletion or pharmacologic modulation was found to protect mice from surgically induced OA [37].

 

Important questions arise from examining the extensive list of inflammatory mediators found in OA joints. Experts have questioned what causes joint tissues to produce proinflammatory factors, and which are the most important drivers of joint tissue destruction that could serve as targets for therapeutic intervention. There is growing evidence that OA is associated with activation of the innate immune response that can be initiated by tissue damage [2,38]. The mediators found in the OA joint are similar to those found in a chronic nonhealing wound. The articular cartilage does not exhibit an adequate repair response, perhaps because it is avascular, and so once sufficient matrix damage has occurred it is not reversible. Fragments of matrix proteins, including fibronectin, cartilage oligomeric protein (COMP), fibromodulin, proteoglycans, and collagen are released from the damage matrix. These matrix fragments stimulate the innate immune response and further promote the upregulation of degradative pathways through activation of toll-like receptors and integrins [2,4,39]. Many of the proinflammatory and catabolic pathways at work in the OA joint involve activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) family of transcriptional regulators [40].

 

Proteases — The destruction of joint tissues in OA is mediated by a variety of proteases including several matrix metalloproteinases (MMPs), cysteine proteinases including cathepsin K, and serine proteinases [41]. Our understanding of the proteases involved in OA is concentrated on those that mediate degradation of cartilage extracellular matrix proteins. Aggrecan is the large proteoglycan that provides much of the resiliency of cartilage. It is degraded, starting in the early stages of OA, by members of the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family referred to as aggrecanases (ADAMTS-4 and -5). Type II collagen, the most abundant collagen in cartilage, is responsible for the tensile strength of cartilage and is degraded by collagenases, which, like aggrecanases, are MMPs. MMP-13 is thought to be the major collagenase responsible for cartilage degradation in OA [41]. Once significant collagen degradation has occurred, it is felt that repair of the damaged matrix is not possible and progression of matrix loss is likely. Given the importance of aggrecanse-2 (ADAMTS-5) and MMP-13 in OA, development of specific inhibitors to these proteases has been of interest for use as potential disease-modifying therapy [42]. Tissue inhibitors of metalloproteinases (TIMPs) are endogenous inhibitors found in joint tissues and synovial fluid that may serve as an alternative to synthetic small molecule inhibitors.

 

MMPs are produced as pro-forms that require proteolytic cleavage to be activated. Serine proteases, including HtrA1 and activated protein C, can serve this role and therefore could also serve as therapeutic targets in OA. Cathepsin K is a cysteine proteinase that is expressed by osteoclasts that can degrade type I collagen in bone but also may degrade type II collagen in cartilage.

 

Other pathways — Several studies have provided evidence for a number of potential mediators of OA that are not considered proinflammatory mediators but appear to promote OA by either activating pathways that promote joint tissue destruction or inhibiting the ability of cells to repair damaged matrix.

 

●FGF signaling – The concept that OA might recapitulate developmental processes has facilitated an exploration of the role of bone morphogenetic proteins (BMPs), fibroblast growth factors (FGF), and the Wnts [43]. FGFs can promote chondrocyte proliferation and stimulate either anabolic or catabolic processes depending on which specific FGF receptor is activated. FGF-18 is a potent cartilage anabolic factor that signals through FGF receptor-3 and is being investigated as a potential intraarticular therapy for OA [3].

●BMP and Wnt signaling – Besides regulating joint development, the connection of BMPs and Wnts to OA is suggested by the concept that OA involves interactions between the subchondral bone (where BMPs and Wnts are also active) and the overlying articular cartilage [44]. BMP-2 and TGF-beta produced locally in the joint appear to be major mediators of osteophyte formation in OA [45,46]. Excessive activation of the Wnt-beta catenin-signaling pathway in cartilage promotes chondrocyte hypertrophy and expression of matrix-degrading enzymes, possibly through Wnt-induced signaling protein 1 [44,47]. The role of Wnt family members in mediating the cartilage-bone connection is less clear, with conflicting results from gene deletion and inhibitor studies depending on the animal model used and the family member targeted [43]. However, there are mouse model data to suggest that TGF-beta activity in subchondral bone mesenchymal stem cells may play a role in promoting degradation of the overlying cartilage [48]. More work is needed to determine how this may apply to human OA, but the findings demonstrate that in some cases OA may originate in the bone rather than the articular cartilage.

●Epigenetics – Another area of active investigation in the pathogenesis of OA is epigenetics. Epigenetic control of gene transcription includes an expanding lists of processes such as DNA methylation, histone modifications (acetylation and methylation), micro-RNAs, and long noncoding RNAs. Using samples of cartilage or bone from normal and OA joints, a number of studies have found significant differences in DNA methylation patterns and likewise a growing list of micro-RNAs and long noncoding RNAs that differ between normal cartilage and OA cartilage [43,49-51]. It is not yet clear which of the various epigenetic changes play a key role in the development of OA in humans.

●Sirtuins – The sirtuin (SIRT) family of nicotinamide adenine dinucleotide (NAD)-dependent deacetylases provides a mechanistic link for the regulation of gene expression in response to altered energy metabolism. SIRT1 was found to be a key factor in the promotion of longevity in response to dietary restriction [52]. SIRT1 serves as a histone deacetylase and mice lacking SIRT1 in cartilage develop spontaneous OA [49]. There are additional SIRTs, including SIRT6, found in cartilage that may also have an important function in maintaining cartilage homeostasis [53].

CLINICAL IMPLICATIONS — Available treatments for osteoarthritis (OA) target pain, and none have been proven to alter the structural progression of the disease. As the understanding of the mechanisms underlying OA improves, treatments are being developed that target specific mediators thought to promote joint tissue destruction. Because OA results from a change in the balance of catabolic and anabolic activity in the joint, both anti-catabolic agents and pro-anabolic agents are being developed and tested. (See "Initial pharmacologic therapy of osteoarthritis" and "Treatment of osteoarthritis resistant to initial pharmacologic therapy" and "Investigational approaches to the management of osteoarthritis".)

 

Until an agent that targets a specific mediator of OA is successfully shown to slow or stop structural progression, it will not be clear which mediators are key to the development and progression of OA. Given that joint tissues, including cartilage, are not capable of adequate intrinsic repair and that advanced disease involves the entire joint, resulting in significant alterations in joint mechanics, it is unlikely that treatment of advanced disease will be successful in reversing the OA process. Rather, targeting patients at earlier stages who are most likely to progress will be needed. Advances in imaging and biochemical biomarkers are bringing the field closer to identifying these patients [54].

 

Improvements in treating structural disease in OA will also require better phenotyping of patients based on the various pathways to OA. It is becoming evident that the pathways and mediators that promote the development of OA after joint injury (posttraumatic OA phenotype) are different from those associated with obesity (metabolic OA phenotype) and aging (age-related OA phenotype). In addition, in some individual factors in the bone may predominate (bone OA phenotype), or mechanical alterations (mechanical OA phenotype) or genetics (genetic OA phenotype) may be the driving forces.

 

SUMMARY

 

●Osteoarthritis (OA) was formerly considered to be simply a degenerative “wear and tear” process and therefore often misnamed as degenerative joint disease. However, the pathogenesis of OA is much more complex than just wear and tear and the term “osteoarthritis,” where “-itis” is indicative of an inflammatory process, is correct. (See 'Introduction' above.)

●OA is one of the most common causes of chronic disability in adults due to pain and altered joint function that is associated with characteristic pathologic changes in the joint tissues (figure 1). Pathological findings in articular cartilage, bone, synovium, and soft tissues are present to varying degrees in all people with OA, suggesting a common response of the joint to a variety of insults. (See 'Overview of pathology' above.)

●Multiple risk factors have been linked to the pathogenesis of OA, including age, joint injury, obesity, genetics, anatomical factors including joint shape and alignment, and gender. (See 'Risk factors' above and 'Aging' above.)

●Proinflammatory factors appear to be driving the production of the proteolytic enzymes responsible for the degradation of the extracellular matrix that results in joint tissue destruction. Although destruction and loss of the articular cartilage is a central component of OA, all joint tissues are affected in some way, indicating that OA is a disease of the whole joint as an organ. While mechanical factors play a key role in OA, excessive or abnormal joint loading also stimulates joint tissue cells to produce proinflammatory factors and proteases that mediate joint tissue destruction. (See 'Role of inflammation' above and 'Inflammatory mediators' above and 'Proteases' above.)

●There are a number of other potential mediators of OA that are not considered proinflammatory mediators but appear to promote OA by either activating pathways that promote joint tissue destruction or inhibiting the ability of cells to repair damaged matrix. (See 'Other pathways' above.)

●As the understanding of the mechanisms underlying OA improves, treatments are being developed that target specific mediators thought to affect bone turnover. Until an agent that targets a specific mediator of OA is successfully shown to slow or stop structural progression, it will not be clear which mediators are key to the development and progression of OA. (See 'Clinical implications' above.)

ACKNOWLEDGMENT — The editorial staff at UpToDate would like to acknowledge Kenneth Kalunian, MD; Susan Ritter, MD; and Peter Tugwell, MD, who contributed to an earlier version of this topic review.

 

REFERENCES

Lane NE, Brandt K, Hawker G, et al. OARSI-FDA initiative: defining the disease state of osteoarthritis. Osteoarthritis Cartilage 2011; 19:478.

Liu-Bryan R, Terkeltaub R. Emerging regulators of the inflammatory process in osteoarthritis. Nat Rev Rheumatol 2015; 11:35.

Yu SP, Hunter DJ. Emerging drugs for the treatment of knee osteoarthritis. Expert Opin Emerg Drugs 2015; 20:361.

Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum 2012; 64:1697.

Sharma L, Chmiel JS, Almagor O, et al. Significance of preradiographic magnetic resonance imaging lesions in persons at increased risk of knee osteoarthritis. Arthritis Rheumatol 2014; 66:1811.

Waller KA, Zhang LX, Elsaid KA, et al. Role of lubricin and boundary lubrication in the prevention of chondrocyte apoptosis. Proc Natl Acad Sci U S A 2013; 110:5852.

Taljanovic MS, Graham AR, Benjamin JB, et al. Bone marrow edema pattern in advanced hip osteoarthritis: quantitative assessment with magnetic resonance imaging and correlation with clinical examination, radiographic findings, and histopathology. Skeletal Radiol 2008; 37:423.

Loeuille D, Chary-Valckenaere I, Champigneulle J, et al. Macroscopic and microscopic features of synovial membrane inflammation in the osteoarthritic knee: correlating magnetic resonance imaging findings with disease severity. Arthritis Rheum 2005; 52:3492.

Baker K, Grainger A, Niu J, et al. Relation of synovitis to knee pain using contrast-enhanced MRIs. Ann Rheum Dis 2010; 69:1779.

Loeser RF. Aging processes and the development of osteoarthritis. Curr Opin Rheumatol 2013; 25:108.

Brophy RH, Rai MF, Zhang Z, et al. Molecular analysis of age and sex-related gene expression in meniscal tears with and without a concomitant anterior cruciate ligament tear. J Bone Joint Surg Am 2012; 94:385.

Roos EM, Herzog W, Block JA, Bennell KL. Muscle weakness, afferent sensory dysfunction and exercise in knee osteoarthritis. Nat Rev Rheumatol 2011; 7:57.

Blagojevic M, Jinks C, Jeffery A, Jordan KP. Risk factors for onset of osteoarthritis of the knee in older adults: a systematic review and meta-analysis. Osteoarthritis Cartilage 2010; 18:24.

Roos H, Adalberth T, Dahlberg L, Lohmander LS. Osteoarthritis of the knee after injury to the anterior cruciate ligament or meniscus: the influence of time and age. Osteoarthritis Cartilage 1995; 3:261.

Struglics A, Larsson S, Kumahashi N, et al. Changes in Cytokines and Aggrecan ARGS Neoepitope in Synovial Fluid and Serum and in C-Terminal Crosslinking Telopeptide of Type II Collagen and N-Terminal Crosslinking Telopeptide of Type I Collagen in Urine Over Five Years After Anterior Cruciate Ligament Rupture: An Exploratory Analysis in the Knee Anterior Cruciate Ligament, Nonsurgical Versus Surgical Treatment Trial. Arthritis Rheumatol 2015; 67:1816.

Riordan EA, Frobell RB, Roemer FW, Hunter DJ. The health and structural consequences of acute knee injuries involving rupture of the anterior cruciate ligament. Rheum Dis Clin North Am 2013; 39:107.

Kumahashi N, Swärd P, Larsson S, et al. Type II collagen C2C epitope in human synovial fluid and serum after knee injury--associations with molecular and structural markers of injury. Osteoarthritis Cartilage 2015; 23:1506.

Oliveria SA, Felson DT, Cirillo PA, et al. Body weight, body mass index, and incident symptomatic osteoarthritis of the hand, hip, and knee. Epidemiology 1999; 10:161.

Johnson VL, Hunter DJ. The epidemiology of osteoarthritis. Best Pract Res Clin Rheumatol 2014; 28:5.

Sellam J, Berenbaum F. Is osteoarthritis a metabolic disease? Joint Bone Spine 2013; 80:568.

Ratneswaran A, LeBlanc EA, Walser E, et al. Peroxisome proliferator-activated receptor δ promotes the progression of posttraumatic osteoarthritis in a mouse model. Arthritis Rheumatol 2015; 67:454.

Snead MP, Yates JR. Clinical and Molecular genetics of Stickler syndrome. J Med Genet 1999; 36:353.

Kannu P, Bateman JF, Randle S, et al. Premature arthritis is a distinct type II collagen phenotype. Arthritis Rheum 2010; 62:1421.

Valdes AM, Spector TD. Genetic epidemiology of hip and knee osteoarthritis. Nat Rev Rheumatol 2011; 7:23.

Moisio K, Chang A, Eckstein F, et al. Varus-valgus alignment: reduced risk of subsequent cartilage loss in the less loaded compartment. Arthritis Rheum 2011; 63:1002.

Andriacchi TP, Favre J. The nature of in vivo mechanical signals that influence cartilage health and progression to knee osteoarthritis. Curr Rheumatol Rep 2014; 16:463.

Cirillo DJ, Wallace RB, Wu L, Yood RA. Effect of hormone therapy on risk of hip and knee joint replacement in the Women's Health Initiative. Arthritis Rheum 2006; 54:3194.

Hussain SM, Cicuttini FM, Bell RJ, et al. Incidence of total knee and hip replacement for osteoarthritis in relation to circulating sex steroid hormone concentrations in women. Arthritis Rheumatol 2014; 66:2144.

Shane Anderson A, Loeser RF. Why is osteoarthritis an age-related disease? Best Pract Res Clin Rheumatol 2010; 24:15.

Lotz M, Loeser RF. Effects of aging on articular cartilage homeostasis. Bone 2012; 51:241.

Verzijl N, Bank RA, TeKoppele JM, DeGroot J. AGEing and osteoarthritis: a different perspective. Curr Opin Rheumatol 2003; 15:616.

Verzijl N, DeGroot J, Thorpe SR, et al. Effect of collagen turnover on the accumulation of advanced glycation end products. J Biol Chem 2000; 275:39027.

Lioté F, Ea HK. Clinical implications of pathogenic calcium crystals. Curr Opin Rheumatol 2014; 26:192.

Abhishek A, Doherty M. Epidemiology of calcium pyrophosphate crystal arthritis and basic calcium phosphate crystal arthropathy. Rheum Dis Clin North Am 2014; 40:177.

Sohn DH, Sokolove J, Sharpe O, et al. Plasma proteins present in osteoarthritic synovial fluid can stimulate cytokine production via Toll-like receptor 4. Arthritis Res Ther 2012; 14:R7.

Little CB, Fosang AJ. Is cartilage matrix breakdown an appropriate therapeutic target in osteoarthritis--insights from studies of aggrecan and collagen proteolysis? Curr Drug Targets 2010; 11:561.

Wang Q, Rozelle AL, Lepus CM, et al. Identification of a central role for complement in osteoarthritis. Nat Med 2011; 17:1674.

Liu-Bryan R, Terkeltaub R. The growing array of innate inflammatory ignition switches in osteoarthritis. Arthritis Rheum 2012; 64:2055.

Sofat N. Analysing the role of endogenous matrix molecules in the development of osteoarthritis. Int J Exp Pathol 2009; 90:463.

Olivotto E, Otero M, Marcu KB, Goldring MB. Pathophysiology of osteoarthritis: canonical NF-κB/IKKβ-dependent and kinase-independent effects of IKKα in cartilage degradation and chondrocyte differentiation. RMD Open 2015; 1:e000061.

Troeberg L, Nagase H. Proteases involved in cartilage matrix degradation in osteoarthritis. Biochim Biophys Acta 2012; 1824:133.

Tonge DP, Pearson MJ, Jones SW. The hallmarks of osteoarthritis and the potential to develop personalised disease-modifying pharmacological therapeutics. Osteoarthritis Cartilage 2014; 22:609.

Loeser RF. Osteoarthritis year in review 2013: biology. Osteoarthritis Cartilage 2013; 21:1436.

Luyten FP, Tylzanowski P, Lories RJ. Wnt signaling and osteoarthritis. Bone 2009; 44:522.

van der Kraan PM, van den Berg WB. Osteophytes: relevance and biology. Osteoarthritis Cartilage 2007; 15:237.

Blaney Davidson EN, Vitters EL, Bennink MB, et al. Inducible chondrocyte-specific overexpression of BMP2 in young mice results in severe aggravation of osteophyte formation in experimental OA without altering cartilage damage. Ann Rheum Dis 2015; 74:1257.

Blom AB, Brockbank SM, van Lent PL, et al. Involvement of the Wnt signaling pathway in experimental and human osteoarthritis: prominent role of Wnt-induced signaling protein 1. Arthritis Rheum 2009; 60:501.

Zhen G, Wen C, Jia X, et al. Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nat Med 2013; 19:704.

Gabay O, Sanchez C. Epigenetics, sirtuins and osteoarthritis. Joint Bone Spine 2012; 79:570.

Tsezou A. Osteoarthritis year in review 2014: genetics and genomics. Osteoarthritis Cartilage 2014; 22:2017.

Liu Q, Zhang X, Dai L, et al. Long noncoding RNA related to cartilage injury promotes chondrocyte extracellular matrix degradation in osteoarthritis. Arthritis Rheumatol 2014; 66:969.

Leibiger IB, Berggren PO. Sirt1: a metabolic master switch that modulates lifespan. Nat Med 2006; 12:34.

Nagai K, Matsushita T, Matsuzaki T, et al. Depletion of SIRT6 causes cellular senescence, DNA damage, and telomere dysfunction in human chondrocytes. Osteoarthritis Cartilage 2015; 23:1412.

Hunter DJ, Nevitt M, Losina E, Kraus V. Biomarkers for osteoarthritis: current position and steps towards further validation. Best Pract Res Clin Rheumatol 2014; 28:61.

Topic 5498 Version 20.0

[emoji767] 2016 UpToDate, Inc. All rights reserved.

Link to comment
Share on other sites

Far be it for me to question this but it's amazing that we have been so dead set on it not being inflammatory in nature for years based on "studies". Appreciate your source info.. Good for me that it's a moot point since I don't see chronic conditions.

 

Addendum: did a search this morning of a subscription service that I have and found studies showing that exercise alone of OA joints were helpful short term (12 weeks), even those with effusions (Cochrane meta-analysis). Always skeptical of M-A studies. Garbage in, garbage out.

Link to comment
Share on other sites

Archived

This topic is now archived and is closed to further replies.

×
×
  • Create New...

Important Information

Welcome to the Physician Assistant Forum! This website uses cookies to ensure you get the best experience on our website. Learn More