Skip to main content
Cartilage logoLink to Cartilage
. 2022 Oct 15;13(4):119–132. doi: 10.1177/19476035221126349

A Fresh Glimpse into Cartilage Immune Privilege

Carissa Garrity 1, Boaz Arzi 2, Brian Haus 3, Cassandra A Lee 3, Natalia Vapniarsky 1,
PMCID: PMC9924976  PMID: 36250484

Abstract

The increasing prevalence of degenerative cartilage disorders in young patients is a growing public concern worldwide. Cartilage’s poor innate regenerative capacity has inspired the exploration and development of cartilage replacement treatments such as tissue-engineered cartilages and osteochondral implants as potential solutions to cartilage loss. The clinical application of tissue-engineered implants is hindered by the lack of long-term follow-up demonstrating efficacy, biocompatibility, and bio-integration. The historically reported immunological privilege of cartilage tissue was based on histomorphological observations pointing out the lack of vascularity and the presence of a tight extracellular matrix. However, clinical studies in humans and animals do not unequivocally support the immune-privilege theory. More in-depth studies on cartilage immunology are needed to make clinical advances such as tissue engineering more applicable. This review analyzes the literature that supports and opposes the concept that cartilage is an immune-privileged tissue and provides insight into mechanisms conferring various degrees of immune privilege to other, more in-depth studied tissues such as testis, eyes, brain, and cancer.

Keywords: cartilage immunology, tissue engineering, MHC, allogenic, immunological rejection, T cell

Introduction

The high prevalence of degenerative articular cartilage disorders among young and adult patients is a public health concern worldwide. Cartilage loss due to trauma or wear leads to osteoarthritis (OA). Clinical symptoms of OA include pain, joint swelling, and restricted range of motion, causing functional disability and reduced quality of life.1 In the United States alone, 1 in 25 working-age adults from 18 to 64 years are limited to arthritis.2 Out of this population, almost half experience work limitations, equating to $164 billion in lost wages.2 It is predicted that by 2040, doctor-diagnosed arthritic conditions will equate to 78.4 million people.2 Cartilage loss in pediatric patients is also increasing in frequency due to a significant rise in sports injuries and is of particular concern given the lifelong disability that subsequently occurs.

Cartilage’s innately poor regenerative capacity3,4 and progressively increasing demand for its regeneration have resulted in astonishing developments and achievements in the field of tissue engineering (TE) and regenerative therapies. However, despite advancements in cartilage TE and current clinical transplantation practices, the ability to replace deficient or damaged cartilage remains limited. Traditional methods such as fresh articular cartilage with bone (osteochondral) autografts and allografts are currently used as cartilage replacements in affected patients.5

Cartilage autografts are derived from the same individual and are harvested from a non-weightbearing area of the joint. However, the quantity of available cartilage is limited, and autograft collection incurs additional morbidity. Allografts are tissues obtained from a donor of the same species. A common example of allogenic cartilage treatments is allogenic osteochondral implantation (OCI), also called osteochondral allograft transplantation (OAT). The benefits of allografts are in the relative availability of donor tissue and the possibility of an off-the-shelf implant that can be introduced in a single surgery without inducing second-site morbidity in the patient.6,7 However, allografts present the risk of disease transmission and eliciting immune rejection.8-10 For instance, despite the average 12-year success rate of osteochondral allografts, up to 25% of allografts are failing, and 36% of patients require additional or revision surgeries.11 The lack of long-term durability and integration could be due to immunological destruction of the graft. Immunological rejection of a tissue graft is typically prevented by immunologic matching of donor and recipient before transplantation. Surprisingly, unlike solid organ allografts, immunologic matching, also known as major histocompatibility complex (MHC) matching, is not routinely performed with osteochondral allografts.12 This is due to the historical concept that cartilage is an “immune-privileged” tissue.13-15

Immune privilege is best summarized as a tissue’s specific ability to repress an immune response to implanted tissues or antigens,16 while immunogenicity is the ability to provoke the immune response.17 Mechanisms governing immune privilege in tissues such as brain,18 eye,19 placenta,20 ovary,21 and testis22 are continuously explored. However, studies addressing the immune status of cartilage are few, and there is a discrepancy between the evidence presented in the literature and how it is translated to clinical practice. For instance, contradicting the immune-privileged claim is the fact that only autologous but not allogenic chondrocytes are used in current cartilage replacement therapies.23,24 In addition, reports comparing the outcomes of allogenic OCI in immunologically matched and mismatched human patients8 and in dogs10 provide evidence that questions the concept of cartilage’s immune privilege. Specifically, in humans, the long-term outcomes of massive osteochondral implants were better when recipients and donors were immunologically matched.8 In dogs, the inflammatory response was much more severe in joints that received an immunologically mismatched allograft.10 Similarly, a histological study in rabbits comparing the host response to cartilage autografts, allografts, and xenografts (tissue from another species) reported strong innate and adaptive immune responses to xenografts, mild responses to allografts, and no response to autografts, in osteochondral implants.13

In contrast, there are numerous studies that provide strong evidence of immunosuppressive and potentially immune-privileged properties of chondrocytes and engineered cartilage neotissue. For instance, several studies have demonstrated that chondrocytes are not only unable to stimulate immune responses, but they also suppress the proliferation of activated immune cells.14,25,26 It is possible that the application of engineered tissue replacements is hindered by our lack of understanding of cartilage immunology and immune privilege. Here, we critically review the literature supporting and confronting the notion that cartilage is an immune-privileged tissue and provide an insight into novel methods that can advance the field of cartilage regeneration.

MHC as a Major Player in Transplantation

The primary role of the MHC molecules is to assist in the maintenance of an organism’s health by presenting foreign, self-, or altered self-peptides to immune cells (lymphocytes and, more specifically, T cells). The function of MHC molecules was first discovered in the context of transplantation in mice, hence, the name major histocompatibility complex.27 However, the primary role of MHC molecules is immunological surveillance and marking of altered or infected cells for the adaptive immune system’s elimination and mounting of the humoral and cellular immune response. The terminology for MHC equivalent in humans identifies the same molecules as (human leukocyte antigen) HLA; in swine as SLA; in a dog, DLA, in cat FLA, and so on for any other species. For this review, the MHC acronym will be used regardless of species. The immune response to non-self-MHC molecules and non-self-peptides presented by the MHC complexes are the primary causes of transplant rejection. For the benefit of understanding the immune status of cartilage tissue, it is imperative first to review the classes and the function of MHC in more detail.

There are 2 classes of MHC molecules. MHC class I molecules fit into 2 categories: classical (MHC Ia) and non-classical (MHC Ib).28 The MHC I molecules are expressed on all nucleated mammalian cells. The class II MHC is expressed by professional antigen-presenting cells (APCs), such as B cell, dendritic cells, and macrophages. As MHC I and MHC II play a critical role in the recognition and activation of immune cells by presenting self-, altered-self-, or non-self-peptides (alloantigens), these molecules are the main drivers of transplant rejection. Analogous to personal identification documents, they are located on the surface of the cells and present intracellular or extracellular components. Specifically, MHC II molecules present extracellular peptides taken up by phagocytosis, while MHC I molecules present intracellular peptides.28 If the MHC presents foreign or inappropriate peptides, activation and clonal expansion of effector T cells will follow, leading to the destruction of the targeted cells/organs. The peptides MHC molecules present are recognized by T lymphocytes via direct, indirect, and semi-direct pathways (Fig. 1A and B). For T cells to be properly activated by MHC, 3 signals should be perceived in conjunction: (1) MHC-peptide complex binding the respective T-cell receptor and co-receptor (CD4 or CD8), (2) binding to co-stimulatory receptors (CD28 and CD80/B7 on lymphocytes and APCs, respectively), and (3) cytokine secretion28 (Fig. 1C). Notably, the lack of MHC expression will be interpreted by natural killer (NK) cells as missing self, and cells with no MHC expression will be killed.

Figure 1.

Figure 1.

MHC antigen presentation pathways in the context of osteochondral implants. (A) In a scenario of osteochondral allograft implantation, APCs can be either donor or recipient derived. The donor APC will reside in the implant’s bone marrow cavity, while recipient APC will be present in the synovium and in the recipient’s bone marrow. (B) In the direct pathway of antigen presentation, donor APC will present donor peptides/antigens on MHC class Ia and MHC class II complexes. In the semi-direct pathway, recipient APC can fuse with donor cell or donor cell exosome membrane to present intact donor MHC Ia complexes with associated donor peptides. In the indirect pathway, a recipient APC presents digested donor peptides (green rectangles) on its MHC class II complex. As a result, the recipient APC can present digested donor peptides or whole donor MHC molecules, which will be subsequently recognized by lymphocytes. (C) Peptide presented by MHC class Ia will be recognized by a recipient cytotoxic T cell (CD8, T cell) and result in strong activation of the lymphocyte. Peptides presented by MHC class II complex will be recognized by T-helper lymphocytes (CD4, T cell). Besides MHC class Ia or MHC class II recognition, additional signals are necessary to activate CD4 T-helper cells or CD8 cytotoxic T cells. In addition to MHC class Ia with presented endogenous peptide binding to the T-cell receptor (TCR) (Signal 1), there should be an additional binding of co-receptor CD8 with MHC complex and CD28 (T cell) binding with CD80/B7 (APC) (Signal 2). Stimulation with appropriate cytokines secreted by the APC will further aid in the activation of the CD8 T cell (Signal 3). To keep activation in balance, the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expression by CD8 T cell will inhibit activation of this cell. A similar mode of 3-signal activation will be needed upon engagement of TCR on the CD4 T-helper cells with exogenous peptides presented by the MHC class II complexes. Upon sufficient activation of CD4 T cells, B cells are activated by recognizing peptides bound to its B-cell receptor and presented by MHC II from CD4 T cells. Once B cells are activated, they will generate antibodies against the peptide recognized by B-cell receptors. MHC = major histocompatibility complex; APC = antigen-presenting cell.

The non-classical MHC I molecules (MHC Ib) are far less studied and are thought to play a critical role in regulating immune activation.29 Specifically, the non-classical MHC I mediate inhibitory or activating stimuli in NK cells (cells of the innate immune system that are capable of killing other cells without the need for specific antibodies) and cytotoxic T cells.30 MHC Ib molecules function as a shield to ensure that the cells are not incorrectly lysed by immune cells.28 Non-classical MHC molecules present the leader peptide (or signal peptide, that initiates the process of assembling all the components of MHC I) derived from classical MHC I molecules to lymphocytes.28 These non-classical MHC-peptide complexes bind to the inhibitory receptors, NKG2A, KIR2D, and LIR-1 on NK cells or CD8 T cells to prevent the cell lysis28 (Fig. 2A). The expression of non-classical MHC molecules is thought to be associated with evasive immune mechanisms seen in tumors,31 placental trophoblasts,32,33 the eye,34 and testis.35

Figure 2.

Figure 2.

Non-classical MHC and altered-self recognition via classical MHC class I. (A) Tumor cells and trophoblasts are known to express the non-classical, MHC class 1b molecules that lead to deactivation and decreased activity of CD8 T cells and NK cells. The binding of minor MHC class 1b complexes is via KIR2D, NKG2A, and LIR-1 receptors. (B) In case of neoplastic transformation or other alteration of cells, the signal protein peptide for MHC I synthesis will be altered or mutated which will be readily recognized by CD8 T cells. The binding and subsequent activation of cytotoxic CD8 T cell will result in the release of granzyme and perforin enzymesmediated destruction of the altered cell. The binding of additional co-receptors such as Fas ligand (FasL) with Fas receptor (FasR) and LFA-1 with ICAM-1 on the CD8 T cell and the altered cell, respectively, will further activate the cytotoxic cell. MHC = major histocompatibility complex; NK = natural killer.

When MHC I complexes present a non-self- or altered peptide, cytotoxic T cells recognize the mismatch and destroy the immuno-incompatible cell (Fig. 2B). Although cartilage is considered an immune-privileged tissue, it is unclear if cartilage MHC expression levels and functionality is different compared with other tissues. For example, it would be essential to understand if the immune privilege of cartilage is due to non-classical MHC I components. Based on our knowledge, it is currently unknown if chondrocytes express non-classical MHC molecules. Many aspects of cartilage immunology still need to be explored before the status of cartilage’s immune privilege can be confirmed and fully understood. In the following sections of the review, we will discuss additional aspects of current knowledge of cartilage immunology.

MHC I matching prevents the recognition of donor MHC as an alloantigen (peptide derived from a different individual) and prevents rejection. MHC molecules are highly polymorphic and diverse across individuals. This diversity allows the host’s immune system to recognize a broad range of pathogens (viruses and bacteria). Still, unfortunately, as a side effect, recognition of non-self-MHC results in the mounting of the immune response following transplantation. Determining the degree of similarity of MHC molecules (MHC matching) between donor and recipient before transplantation helps to select individuals with similar MHC phenotypes.28 It is done by comparing the blood type and evaluating anti-MHC antibodies in the recipient’s serum.36 However, even in MHC identical individuals, rejection can still be facilitated by MHC presentation of other alloantigens. Therefore, recipients may require lifelong immunosuppressive therapy to prevent immunological rejection of the transplant.28 Consequently, it is not surprising to find down-regulation of MHC on tumors37 or the trophoblast,38 tissues that typically avoid immune recognition and subsequent destruction.

Immune Characteristics of Cartilage

Evidence Supporting Immune Privilege

Numerous studies have suggested that chondrocytes are immune evasive.14,25,26,39 Adkisson et al.14 showed that isolated chondrocytes inhibit T-cell proliferation in vitro through 3 main mechanisms: expression of CD80/B7 inhibitors, chondromodulin I, and secretion of indoleamine 2,3-dioxygenase (IDO). B7 inhibitors, such as PD-L1, directly bind to CD28/PD-1, hence preventing the co-stimulation needed for effector and naïve T-cell activation.28 Chondromodulin-I is a glycoprotein that promotes chondrogenesis40 and inhibits CD4 T-cell activation by downregulating the production of IL-2.41 IDO is an enzyme involved in tryptophan catabolism.42 By depleting the extracellular environment from tryptophan, IDO prevents T effector cell proliferation and induces cellular arrest in these cells.43 In addition, the metabolites of IDO are toxic to CD8 T cells and Th1 cells, while IDO itself is reported to promote T regulatory cell differentiation.43 IDO has been found in immune-privileged tissues such as the testis44 and has been shown to play a role in preventing allogenic rejection of fetuses during pregnancy.45 However, IDO has also been detected in non-privileged tissues such as the spleen and skin in mice.46 Due to IDO expression in both privileged and non-privileged tissue, it would be inaccurate to argue that the presence of IDO alone is sufficient to induce immune privilege (Fig. 3A).

Figure 3.

Figure 3.

Concepts supporting and refuting cartilage’s immune privilege. (A) The left side of the scheme presents factors supporting the privilege. ECM shielding is probably the most common argument. The presence of immunomodulatory molecules such as TGF-β in the ECM is another possibility. Some groups demonstrated expression of Fas ligand (FasL), CD80/B7 inhibitors, such as PD-L1/L2, and secretion of IDO by chondrocytes. (B) The right side presents factors disputing the immune privilege of cartilage. Chondrocytes, just like any other somatic cells, express MHC class 1a molecules and may express MHC class II upon inflammatory cytokines stimulation. APC and lymphocytes are present in the synovium of all synovial joints and will be able to detect non-self-antigens. Histological examination of not-matched osteochondral allografts determined the presence of pannus tissue. Pannus formation is usually induced by antigenic stimulation in cases of rheumatoid arthritis or bacterial infections. ECM = extracellular matrix; MHC = major histocompatibility complex; APC = antigen-presenting cell; IDO = indoleamine 2,3-dioxygenase.

An additional argument is that immune-evasive properties of chondrocytes are linked to their mesenchymal lineage.47-49 Mesenchymal stromal cells (MSCs) are the progenitor cells to chondrocytes with well-documented immunomodulatory properties.50 MSCs inhibit T-cell proliferation50-52 through the expression of non-classical MHC molecules and the secretion of soluble mediators such as TGF-β, IL-10, and IDO.53-56 Chondrocytes also secrete TGF-β and IDO. Despite the shared lineage between chondrocytes and MSCs, terminal chondrogenic differentiation can alter chondrocytes’ immune characteristics, making them more immunogenic.57 For example, rat MSC-derived chondrocytes were shown to acquire the surface expression of CD80/B7 and CD86/CD28, which rendered them more immunogenic than the parent MSCs lacking expression of these molecules.57 With that said, human articular chondrocytes have no expression of CD80 or CD86 at either protein or RNA levels.26 Species differences could explain the latter. While direct contact between lymphocytes and chondrocytes is possible in vitro, in reality, chondrocytes in situ are surrounded by a dense network of extracellular matrix (ECM). Thus, the third postulate for chondrocyte immune privilege claims that ECM sequesters antigens and shields against immune detection.25,58,59

The composition and density of ECM are essential for the function of the tissue but may also shield chondrocytes from direct contact with lymphocytes. A lymphocyte has an average diameter of 6 to 7 µm.60 Wolf et al.61 have shown that T cells can migrate through multicomponent synthetic lattices by amoeboid shape changes as long as the gap size remains below 1 to 2 µm. During transplant rejection of solid organs, lymphocytes must traverse the basement membrane (a type of ECM).62 The pore size of basement membrane is 0.75 to 3.85 µm,63 about half the diameter of a lymphocyte. The average pore size of cartilage ECM is 6 nm in diameter,64 a thousand times smaller than lymphocyte diameter. The latter suggests that the density of cartilage’s ECM may protect allogenic chondrocytes from lymphocyte detection. It remains to be determined what happens in case of ECM degradation and if lymphocytes can “find” allogenic chondrocytes upon even the slightest ECM breakage.

It is possible that, in addition to the structural barrier, the ECM contributes to the immune privilege of cartilage by harboring non-cellular immune modulators. For example, active and latent forms of TGF-β1 were reported to be present in the ECM.65 TGF-β is important in chondrogenesis,66 ECM synthesis,67 and inducing tolerance through the induction of T regulatory cell differentiation.28 High molecular weight hyaluronic acid, a major glycosaminoglycan of cartilage ECM, was reported to induce the expression of regulatory genes such as IL-10 in macrophages.68 Therefore, it is reasonable to assume that ECM density, entrapped bioactive proteins, and structural components directly may contribute to immune privilege of cartilage tissue.

Recently, a significant contribution of CD8+ T regulatory cells to the immune-evasive mechanisms of immune-privileged tissues69-72 was reported. CD8+ T regulatory cells have been found in immune-privileged tissues such as the eye73 and testis.74 The CD8+CD122+ T regulatory cells are naturally occurring regulatory cells that inhibit the proinflammatory cytokine (IFN-gamma) by activated CD8 and CD4 T.75 If CD8+ T regulatory cells reside in the joint or in the cartilage tissue, it could explain the lack of immune responses seen following cartilage transplantation.76 Further investigation is needed to determine if CD8+ T regulatory cells are present in cartilage grafts or within other components of the synovial joint.

Evidence Confronting Immune Privilege

While the arguments presented in the previous section support cartilages’ immune privilege, they are primarily based on in vitro or ex vivo experiments. It is unclear if these mechanisms translate to in vivo situations where the tissue integrity and joint environment are not controlled experimentally. A 12-year follow-up study of osteochondral transplants in 291 patients reported a 25% failure rate with a 36% rate of revision surgery.11 This review did not discuss the reasons for the implant failure.11 The authors do not specify if immunological matching was performed between the donors and recipients, but based on the general trend in the field, it is likely that it was not performed. As osteochondral plugs include cartilage, bone, and bone marrow, elements other than cartilage may compromise the successful integration of osteochondral transplants. Indeed, it was established a long ago that fresh and frozen bone allografts are highly immunogenic.77 The bone marrow elements were also proven immunogenic.78 In support of the latter radiologic and histologic studies investigating the failed osteochondral allografts from the talus reported edema around the implants, progressive cyst formation, irregularities at the subchondral plate, fragmentation, subchondral collapse, and importantly abundance of rejection-associated lymphocytic subsets (Fig. 3B).79

It is currently recommended practice to pulse lavage the osteochondral allografts to physically remove the residual marrow cells to reduce the potential contribution of immunogenicity by the bone marrow.80,81 However, a study by Ambra et al.82 investigating pulse lavage in removing bone marrow elements showed little efficiency in this procedure. A report by Friedlaender et al.8 compared the long-term outcomes of large OAT in MHC matched versus mismatched in 29 human patients. This study concluded that there is a benefit in MHC matching, particularly for class II MHC.8 Specifically, patients who had failed or fair outcomes (some or significant pain, functional limitation, and need for braces) had more anti-MHC antibodies either before or following the transplantation.8 A study by Hunt et al.12 determined that the presence of anti-MHC class I antibodies in immunologically mismatched recipients of fresh osteochondral allografts was associated with poor graft survival compared with recipients with no such antibodies. Also, allograft size was associated with the outcome.12 The larger osteochondral allografts were associated with a higher implant failure rate.12 Cumulatively, it can be deducted from the studies above that the high degree of osteochondral implant failure can be due to immune incompatibility of bone and bone marrow components; correspondingly, immunologic matching may reduce the chance of osteochondral implant failure.

However, it remains unclear if cartilage tissue itself is immunologically inert or if it is too contributing to the immunologic process in failing implants. In addition to a tight ECM (discussed earlier), cartilage’s lack of vascularity was proposed to contribute to its privileged immune status. Kandel et al.83 study evaluating failed osteochondral implants from the knee reported pannus formation, fibrillation, degeneration, and erosion of the chondral portions of the grafts. According to the medical dictionary definition, a pannus is a sheet of inflammatory granulation tissue that spreads from the synovial membrane and invades the joint. The synovial membrane is a highly vascularized structure. It is an immunological guardian of the joint containing phagocytic cells capable of antigen presentation and chemical signaling to recruit additional leukocytes from the circulation.84 A study in rats where chondrocytes suspended in hyaluronan were cross-transplanted among rats of 2 separate strains found that lymphocyte infiltration was seen not at the articular surface but the deep margin of the implant. The authors suggested that allogenic chondrocytes may have specific surface antigens that may attract leukocytes and NK cells from the bone marrow.85 Pannus formation was not reported in this study. Still, the lack of vascularity in the cartilage tissue did not protect it from the egress of leukocytes via other vascularized elements of the joint such as synovium or bone marrow. In concert, these observations suggest that cartilage’s lack of vascularity is not a sufficient mechanism to protect it from immune cells of the other vascularized compartments of the joint and that cartilage is potentially also immunogenic.

Reports from additional animal studies further subvert cartilage’s immune-privilege status. For example, we found no reports on cartilaginous or osteochondral allotransplantation in veterinary surgical clinical practice, despite abundant reports on successful allografting of cartilage under experimental preclinical settings.86,87 A study in dogs found that the inflammatory response was the most severe in joints implanted with immunologically mismatched osteochondral allografts.10 Also, pannus formation was typical in cases where fresh mismatched implants were introduced.10 The same study also reported that cryopreservation of the osteochondral implants enhanced the deleterious effects on the allografts.10 Assuming cartilage is immunogenic, it would be essential to analyze what components may potentially contribute to the immunogenicity—cells or ECM.

Studies on the immune response toward decellularized natural biomaterial matrixes that are essentially comprised of ECM molecules indicate that structural molecules can be immunogenic.88 Similarly, renal transplantation studies report antibodies to ECM components such as perlecan, fibronectin, and collagens in patients with chronic renal rejection.89,90 More specific to cartilage tissue, it was shown by Klatt et al.91 that nonfibrillar collagen type II enhanced gene expression of proinflammatory mediators and proteolytic enzymes in chondrocytes. Also, hyaluronic acid, a second major ECM component of cartilage, was shown to have differential signaling based on its molecular weight. Specifically, Rayahin et al.68 demonstrated that contrary to high molecular weight hyaluronic acid that has immunomodulatory properties, low molecular weight hyaluronic acid induced proinflammatory genes in macrophages. The latter 2 studies suggest that cartilage is shielded from immune system surveillance as long as its matrix is intact. However, the immune system reacts when ECM components and chondrocytes are exposed to immune cell-bearing joint elements such as synovium or bone marrow.

Chondrocyte surface antigens, such as MHC molecules, could further contribute to the aggravation of the immune response. It is well known that chondrocytes constitutively express MHC I and, in some species, also MHC II molecules.25 Studies by Hunt and Friedlaender (mentioned above) documented the presence of anti-MHC antibodies in patients with poor osteochondral transplantation outcomes.8,12 However, it remains to be determined if these antibodies were developed in response to MHC molecular presentation by chondrocytes or other elements of the osteochondral implant. Furthermore, it remains to be determined if and how antibodies contribute to implant failure or negative outcomes. Indeed, Williams et al.92 describe a mechanism known as accommodation, where the transplant can remain uninjured despite the presence of anti-MHC antibodies directed against it. Several studies confirm this phenomenon may be true in renal93 and heart94 transplantation. Dehoux and Gianello95 suggest that there may be a “delicate balance” between the production of neutralizing and non-neutralizing antibodies to prevent rejection while inducing accommodation. Identifying factors that would tip the balance in favor of accommodation would be an essential contribution to our understanding of cartilage immunology and the ability to engineer and direct outcomes of allotransplantation.96-98

Lessons from Other Immune-Privileged Tissues

Two tissues commonly studied for their immune-privilege mechanisms are the testis99 and the eye.100 A recent review defined the immune privilege of the testis as a tissue capable of inducing a systemic tolerance of alloantigens, xenoantigens, and immunogenic autoantigens.99 The Sertoli cell barrier and a specific macrophage phenotype in the interstitium contribute to the testis’s immune privilege.99 A specific phenotype of immunomodulatory interstitial testicular macrophages was identified in rats.101 These macrophages differed from the circulating cells by their ability to constitutively produce and secrete anti-inflammatory cytokines.101,102 It remains to be determined if similar macrophages are present in the cartilaginous joint.

The expression of transmembrane molecule Fas ligand (FasL) by ocular100 and testicular99 macrophages are additional factors that contribute to the immune privilege of these organs. The Fas receptor (FasR)-FasL interaction is a signal transduction pathway that regulates cell death.103 The cell bearing the receptor is targeted for death, while the cell bearing the ligand induces death.103 If a cell expresses the FasR, the cell is capable of being recognized and killed by immune cells. However, if the cell expresses FasL, it can induce apoptosis of immune cells and escape the destruction. While FasR presence on chondrocytes is unequivocal, FasL expression is controversial and was undetectable in healthy and osteoarthritic human cartilage by the Hashimoto group,104 but was reported by Fujihara et al.76 in human and murine auricular chondrocytes. Interestingly, a significant amount of soluble FasL was found in the synovial fluid of OA and rheumatoid arthritis patients.105 It remains to be elucidated if the soluble FasL in the synovium contributes to immune-modulatory properties seen in cartilage tissue. Although, the self-perpetuating nature of OA106 does not support this notion.

Extensive reports exist on cancer immuno-evasion.107,108 Selected examples of strategies employed by cancer cells to avoid recognition of altered-self include down-regulation of MHC Ia,109 expression of FasL,110 and the presence of tolerogenic tumor-associated APC111 in the tumor interstitium (Fig. 4). To the best of our knowledge, tolerance-inducing phenotypes of APCs have not been detected in cartilage tissue or the synovium. Further research is needed to characterize leukocyte populations within synovium and their influence on articular cartilage immunology. In addition, there have been no experiments inducing the systemic tolerance to xenoantigens or alloantigens transplanted directly into cartilage or the joint. Therefore, more studies are necessary to determine if the cartilage is indeed an immune-privileged tissue and what factors contribute to this privilege. Perhaps evaluating cartilage immune privilege in the context of the entire joint considering it an organ, would be insightful.

Figure 4.

Figure 4.

Selected factors known to confer an immune privilege to tissues are shown: (1) Secretion of an increased concentration of adenosine and lactate in the ECM of the tumors; (2) overexpression of FasL by tumor cells was shown to induce apoptosis in cytotoxic T cells; (3) down-regulation of MHC class 1a was reported in regular and transmissible tumors as well as trophoblasts; (4) presence of stromal regulatory T cells CD8+CD122+ was reported in testis and the eye; (5) overexpression of minor, MHC 1b complexes was reported in trophoblasts and tumor cells; (6) secretion of IDO, TGF-β, and IL-10 is a well-documented strategy to induce the T regulatory lymphocytes’ formation to suppress the immune recognition by eye, testis, and neoplastic cells; (7) presence of tolerogenic dendritic cells in the stroma of the eye and testis was reported. ECM = extracellular matrix; MHC = major histocompatibility complex; IDO = indoleamine 2,3-dioxygenase.

Methods to Manipulate Immunity

In principle, even with MHC matching for organ transplantation, immunosuppression is still required to avoid rejection.112,113 Regrettably, immunosuppression may lead to significant complications such as increased susceptibility to opportunistic infections.114 Thus, current research efforts are directed at the investigation of methods of masking recognizable antigens or manipulating genes responsible for immune recognition of non-self-antigens to offer a potential solution for manufacturing immunouniversal organs.115

In this context, betta 2 microglobulin (B2M) gene was knocked out in mice resulting in a complete absence of MHC Ia surface molecule expression, making these animals a universal organ donor.116 MHC-null pigs117 and individual specific cell lines such as hematopoietic blood progenitors and endothelial cells118 were developed using a similar strategy. Along these lines, immuno-engineered MHC-null embryonic stem cells119 and induced pluripotent stem cells120 were explored as promising cell sources for regenerative medicine, considering their unlimited proliferation capacity and the ability to differentiate into any functional tissue.

Although elimination of MHC expression renders cells invisible to lymphocyte recognition, NK cells can detect and destroy cells with “missing self.”28 In addition, slight differences in minor histocompatibility molecules or tissue-specific antigens may result in immune rejection.28 This problem can be solved by the induction of expression of single-chain dimers of non-classical MHC I complex.121 These single-chain fragments of MHC I proteins bind to inhibitory receptors such as CD94/NGK2A on NK cells and T cytotoxic T cells and prevent the cell from lysis. Indeed, this strategy was applied and proved effective in protecting induced pluripotent human stem cells genetically edited to express no MHC Ia but single-chain non-classical MHC Ib from lysis. This study utilized recombinant adeno-associated virus gene editing.121

Genetic deletion of MHC I is not the only way to manipulate immunity. Mechanisms that disrupt cytokine gradients and cytokine receptors responsible for effector T cell trafficking can be considered. Expectedly, nature mastered this approach in the maternal-fetal interface where Ccl5 and Cxcl9/10 genes are epigenetically silenced.122 As a result of this epigenetic silencing, effector cells cannot reach the fetus, thus preventing its recognition as non-self and subsequent distraction. In the context of cartilage, CCR2 (cytokine receptor) agonist administration to osteoarthritic mice attenuated macrophage accumulation in the synovium, reduced synovitis, and cartilage damage.123 This same study showed no benefit of blocking CCL5/CCLR5 axis in attenuating OA. These findings imply that T-cell trafficking signals may be specific to tissue types, and cytokine trafficking manipulation for immunomodulation purposes may need to be tailored to specific tissue types.

Inhibition of cytotoxic T-cell activity, induction of T regulatory cell phenotypes, and shift of macrophage and dendritic cells toward tolerogenic phenotypes are additional tactics that were shown success in immunomodulation. As shown in Figure 1 of this review, CTLA-4 is an inhibitory receptor, also known as CD152, expressed by T regulatory cells or effector T cells after activation that binds to CD80 or CD86 receptors on APC to induce an inhibitory signal. Administration of CTLA-4 immunoglobulins or tetra dimers thereof was shown to be effective in modulating the immune response in patients and animal models of rheumatoid arthritis.124 To overcome the challenges of delivery of such potent molecules to the implantation site, nanotechnology and nanoengineering have very clever methods to offer.125,126 For instance, to prevent the rejection of xenogeneic pancreatic islets, encapsulating these islets into a nanobot capable of a sustained release of CTLA-4 peptide was shown to prolong pancreatic islet survival in the xenogeneic transplantation model.127 CTLA4 biopatterned implant not only survived the transplantation and sustained normoglycemia in the experimental mice but also induced T regulatory differentiation at the implantation site.127

Induction of T regulatory phenotypes at the implantation site can be a more precise and accurate way to induce tolerance if APCs are given the correct type of antigen to present to T cells and “educate” these cells to be tolerant toward this particular antigen. The antigen is known in instances like pancreatic islet transplantation or autoimmune destruction. Hence, using dual nanoformulation delivery, impressive work has been done on inducing antigen-specific tolerance and preventing autoimmune type I diabetes in mice.128 Specifically, a delivery system comprised of 4 different nanoparticles was designed. Each particle was loaded with either tolerance-inducing factors (Vitamin D3 or TGF-β1) or dendritic cell colony-stimulating factor (GM-CSF), or insulin (antigen). These particles prevented the development of autoimmune destraction of pancreatic islets in experiemntal non-obese diebetic mice model, when administered as a cocktail.128 These particles prevented the development of autoimmune destruction of pancreatic islets in experiemntal non-obese diebetic mice model, when administered as a cocktail.128 In the context of cartilage, it is unknown if and which peptides may be immunogenic. Thus, more work needs to be done to identify these potentially antigenic molecules before immunoneutral cartilage tissue can be engineered.

Conclusions

The increasing prevalence of degenerative cartilage diseases is a growing public health concern worldwide. The transplantation of cartilage allografts is regarded as a safe solution due to the historical concept of cartilage being an immune-privileged tissue. However, several studies confront the concept of the immune-privilege status of cartilage by providing evidence of an immune response following allogenic cartilage transplantation. These contradictory findings highlight the need for studies that explore the immunology of cartilage.

The study of immune-tissues is highly instructive in developing universal tissues suitable for transplantation. The immune evasion mechanisms discovered thus far have contributed toward engineering immunologically invisible cell lines. The efficacy of MHC engineering and CRISPR multiplexing in various cell lines suggests that this technology could be translated to cartilage tissue. The application of this technology to cartilage tissue could create universal cartilage allografts that could potentially promote the clinical application of tissue-engineered cartilage and help millions of patients.

Footnotes

Acknowledgments and Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Ethical Statement: Ethical approval was not sought for the present study because it is a systematic review and does not directly involve patients.

ORCID iD: Natalia Vapniarsky Inline graphic https://orcid.org/0000-0003-3239-6233

References

  • 1. Hunter DJ, McDougall JJ, Keefe FJ. The symptoms of osteoarthritis and the genesis of pain. Rheum Dis Clin North Am. 2008;34(3):623-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Center for Disease Control and Prevention. Arthritis-related statistics. 2018. Jul 18. Available from: https://www.cdc.gov/arthritis/data_statistics/arthritis-related-stats.htm.
  • 3. Nam Y, Rim YA, Jung SM, Ju JH. Cord blood cell-derived iPSCs as a new candidate for chondrogenic differentiation and cartilage regeneration. Stem Cell Res Ther. 2017;8(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Akkiraju H, Nohe A. Role of chondrocytes in cartilage formation, progression of osteoarthritis and cartilage regeneration. J Dev Biol. 2015;3(4):177-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Martín AR, Patel JM, Zlotnick HM, Carey JL, Mauck RL. Emerging therapies for cartilage regeneration in currently excluded “red knee” populations. NPJ Regen Med. 2019;4:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Cinque ME, Kennedy NI, Moatshe G, Vinagre G, Chahla J, Hussain ZB, et al. Osteochondral allograft transplants for large trochlear defects. Arthrosc Tech. 2017;6(5):e1703-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Arzi B, Clark KC, Sundaram A, Spriet M, Verstraete FJM, Walker NJ, et al. Therapeutic efficacy of fresh, allogeneic mesenchymal stem cells for severe refractory feline chronic gingivostomatitis. Stem Cells Transl Med. 2017;6(8):1710-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Friedlaender GE, Strong DM, Tomford WW, Mankin HJ. Long-term follow-up of patients with osteochondral allografts. A correlation between immunologic responses and clinical outcome. Orthop Clin North Am. 1999;30(4):583-8. [DOI] [PubMed] [Google Scholar]
  • 9. Oakeshott RD, Farine I, Pritzker KP, Langer F, Gross AE. A clinical and histologic analysis of failed fresh osteochondral allografts. Clin Orthop Relat Res. 1988;233:283-94. [PubMed] [Google Scholar]
  • 10. Stevenson S, Dannucci GA, Sharkey NA, Pool RR. The fate of articular cartilage after transplantation of fresh and cryopreserved tissue-antigen-matched and mismatched osteochondral allografts in dogs. J Bone Joint Surg Am. 1989;71(9):1297-307. [PubMed] [Google Scholar]
  • 11. Assenmacher AT, Pareek A, Reardon PJ, Macalena JA, Stuart MJ, Krych AJ. Long-term outcomes after osteochondral allograft: a systematic review at long-term follow-up of 12.3 years. Arthroscopy. 2016;32(10):2160-8. [DOI] [PubMed] [Google Scholar]
  • 12. Hunt HE, Sadr K, Deyoung AJ, Gortz S, Bugbee WD. The role of immunologic response in fresh osteochondral allografting of the knee. Am J Sports Med. 2014;42(4):886-91. [DOI] [PubMed] [Google Scholar]
  • 13. Arzi B, DuRaine GD, Lee CA, Huey DJ, Borjesson DL, Murphy BG, et al. Cartilage immunoprivilege depends on donor source and lesion location. Acta Biomater. 2015;23:72-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Adkisson HD, Milliman C, Zhang X, Mauch K, Maziarz RT, Streeter PR. Immune evasion by neocartilage-derived chondrocytes: implications for biologic repair of joint articular cartilage. Stem Cell Res. 2010;4(1):57-68. [DOI] [PubMed] [Google Scholar]
  • 15. Elattar M, Dhollander A, Verdonk R, Almqvist KF, Verdonk P. Twenty-six years of meniscal allograft transplantation: is it still experimental? A meta-analysis of 44 trials. Knee Surg Sports Traumatol Arthrosc. 2011;19(2):147-57. [DOI] [PubMed] [Google Scholar]
  • 16. Forrester JV, Xu H, Lambe T, Cornall R. Immune privilege or privileged immunity? Mucosal Immunol. 2008;1(5):372-81. [DOI] [PubMed] [Google Scholar]
  • 17. Mahanty S, Prigent A, Garraud O. Immunogenicity of infectious pathogens and vaccine antigens. BMC Immunol. 2015;16:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Wenkel H, Streilein JW, Young MJ. Systemic immune deviation in the brain that does not depend on the integrity of the blood-brain barrier. J Immunol. 2000;164(10):5125-31. [DOI] [PubMed] [Google Scholar]
  • 19. Taylor AW. Ocular immune privilege and transplantation. Front Immunol. 2016;7:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Kanellopoulos-Langevin C, Caucheteux SM, Verbeke P, Ojcius DM. Tolerance of the fetus by the maternal immune system: role of inflammatory mediators at the feto-maternal interface. Reprod Biol Endocrinol. 2003;1:121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Minas V, Rolaki A, Kalantaridou SN, Sidiropoulos J, Mitrou S, Petsas G, et al. Intratumoral CRH modulates immuno-escape of ovarian cancer cells through FasL regulation. Br J Cancer. 2007;97(5):637-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Fijak M, Meinhardt A. The testis in immune privilege. Immunol Rev. 2006;213:66-81. [DOI] [PubMed] [Google Scholar]
  • 23. Jacobi M, Villa V, Magnussen RA, Neyret P. MACI—a new era? Sports Med Arthrosc Rehabil Ther Technol. 2011;3(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Ossendorf C, Steinwachs MR, Kreuz PC, Osterhoff G, Lahm A, Ducommun PP, et al. Autologous chondrocyte implantation (ACI) for the treatment of large and complex cartilage lesions of the knee. Sports Med Arthrosc Rehabil Ther Technol. 2011;3:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Osiecka-Iwan A, Hyc A, Radomska-Lesniewska DM, Rymarczyk A, Skopinski P. Antigenic and immunogenic properties of chondrocytes. Implications for chondrocyte therapeutic transplantation and pathogenesis of inflammatory and degenerative joint diseases. Cent Eur J Immunol. 2018;43(2):209-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Pereira RC, Martinelli D, Cancedda R, Gentili C, Poggi A. Human articular chondrocytes regulate immune response by affecting directly T cell proliferation and indirectly inhibiting monocyte differentiation to professional antigen-presenting cells. Front Immunol. 2016;7:415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Snell GD, Higgins GF. Alleles at the histocompatibility-2 locus in the mouse as determined by tumor transplantation. Genetics. 1951;36(3):306-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Murphy K, Weaver C. Janeway’s immunobiology. New York: Garland Science, Taylor and Francis Group LLC; 2017. p. 213-371 [Google Scholar]
  • 29. Allen RL. Non-classical immunology. Genome Biol. 2001;2(2):REPORTS4004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Halenius A, Gerke C, Hengel H. Classical and non-classical MHC I molecule manipulation by human cytomegalovirus: so many targets—but how many arrows in the quiver? Cell Mol Immunol. 2015;12(2):139-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Kochan G, Escors D, Breckpot K, Guerrero-Setas D. Role of non-classical MHC class I molecules in cancer immunosuppression. Oncoimmunology. 2013;2(11):e26491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Tilburgs T, Henry Evans J, Crespo ÂC, Strominger JL. The HLA-G cycle provides for both NK tolerance and immunity at the maternal-fetal interface. Proc Natl Acad Sci U S A. 2015;112(43):13312-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Rizzo R, Vercammen M, van de Velde H, Horn PA, Rebmann V. The importance of HLA-G expression in embryos, trophoblast cells, and embryonic stem cells. Cell Mol Life Sci. 2011;68(3):341-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Niederkorn JY, Chiang EY, Ungchusri T, Stroynowski I. Expression of a nonclassical MHC class Ib molecule in the eye. Transplantation. 1999;68(11):1790-9. [DOI] [PubMed] [Google Scholar]
  • 35. Fiszer D, Kurpisz M. Major histocompatibility complex expression on human, male germ cells: a review. Am J Reprod Immunol. 1998;40(3):172-6. [DOI] [PubMed] [Google Scholar]
  • 36. Berger A. HLA typing. BMJ. 2001;322(7280):218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Bijen CB, Bantema-Joppe EJ, de Jong RA, Leffers N, Mourits MJE, Eggink HF, et al. The prognostic role of classical and nonclassical MHC class I expression in endometrial cancer. Int J Cancer. 2010;126(6):1417-27. [DOI] [PubMed] [Google Scholar]
  • 38. Rapacz-Leonard A, Dąbrowska M, Janowski T. Major histocompatibility complex I mediates immunological tolerance of the trophoblast during pregnancy and may mediate rejection during parturition. Mediators Inflamm. 2014;2014:579279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Abe S, Nochi H, Ito H. Alloreactivity and immunosuppressive properties of articular chondrocytes from osteoarthritic cartilage. J Orthop Surg (Hong Kong). 2016;24(2):232-9. [DOI] [PubMed] [Google Scholar]
  • 40. Klinger P, Surmann-Schmitt C, Brem M, Swoboda B, Distler JH, Carl HD, et al. Chondromodulin 1 stabilizes the chondrocyte phenotype and inhibits endochondral ossification of porcine cartilage repair tissue. Arthritis Rheum. 2011;63(9):2721-31. [DOI] [PubMed] [Google Scholar]
  • 41. Setoguchi K, Misaki Y, Kawahata K, Shimada K, Juji T, Tanaka S, et al. Suppression of T cell responses by chondromodulin I, a cartilage-derived angiogenesis inhibitory factor: therapeutic potential in rheumatoid arthritis. Arthritis Rheum. 2004;50(3):828-39. [DOI] [PubMed] [Google Scholar]
  • 42. Soliman H, Mediavilla-Varela M, Antonia S. Indoleamine 2,3-dioxygenase: is it an immune suppressor? Cancer J. 2010;16(4):354-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Wu H, Gong J, Liu Y. Indoleamine 2, 3-dioxygenase regulation of immune response (Review). Mol Med Rep. 2018;17(4):4867-73. [DOI] [PubMed] [Google Scholar]
  • 44. Gualdoni GS, Jacobo PV, Sobarzo CM, Pérez CV, Matzkin ME, Höcht C, et al. Role of indoleamine 2,3-dioxygenase in testicular immune-privilege. Sci Rep. 2019;9(1):15919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, Marshall B, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281(5380):1191-3. [DOI] [PubMed] [Google Scholar]
  • 46. Dai X, Zhu BT. Indoleamine 2,3-dioxygenase tissue distribution and cellular localization in mice: implications for its biological functions. J Histochem Cytochem. 2010;58(1):17-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Du WJ, Reppel L, Leger L, Schenowitz C, Huselstein C, Bensoussan D, et al. Mesenchymal stem cells derived from human bone marrow and adipose tissue maintain their immunosuppressive properties after chondrogenic differentiation: role of HLA-G. Stem Cells Dev. 2016;25(19):1454-69. [DOI] [PubMed] [Google Scholar]
  • 48. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringdén O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol. 2003;31(10):890-6. [DOI] [PubMed] [Google Scholar]
  • 49. Zheng ZH, Li XY, Ding J, Jia JF, Zhu P. Allogeneic mesenchymal stem cell and mesenchymal stem cell-differentiated chondrocyte suppress the responses of type II collagen-reactive T cells in rheumatoid arthritis. Rheumatology (Oxford). 2008;47(1):22-30. [DOI] [PubMed] [Google Scholar]
  • 50. Peroni JF, Borjesson DL. Anti-inflammatory and immunomodulatory activities of stem cells. Vet Clin North Am Equine Pract. 2011;27(2):351-62. [DOI] [PubMed] [Google Scholar]
  • 51. Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, et al. Human mesenchymal stem cells modulate B-cell functions. Blood. 2006;107(1):367-72. [DOI] [PubMed] [Google Scholar]
  • 52. Ben-Ami E, Berrih-Aknin S, Miller A. Mesenchymal stem cells as an immunomodulatory therapeutic strategy for autoimmune diseases. Autoimmun Rev. 2011;10(7):410-5. [DOI] [PubMed] [Google Scholar]
  • 53. DelaRosa O, Lombardo E, Beraza A, Mancheño-Corvo P, Ramirez C, Menta R, et al. Requirement of IFN-gamma-mediated indoleamine 2,3-dioxygenase expression in the modulation of lymphocyte proliferation by human adipose-derived stem cells. Tissue Eng Part A. 2009;15(10):2795-806. [DOI] [PubMed] [Google Scholar]
  • 54. Ge W, Jiang J, Arp J, Liu W, Garcia B, Wang H. Regulatory T-cell generation and kidney allograft tolerance induced by mesenchymal stem cells associated with indoleamine 2,3-dioxygenase expression. Transplantation. 2010;90(12):1312-20. [DOI] [PubMed] [Google Scholar]
  • 55. Nasef A, Chapel A, Mazurier C, Bouchet S, Lopez M, Mathieu N, et al. Identification of IL-10 and TGF-beta transcripts involved in the inhibition of T-lymphocyte proliferation during cell contact with human mesenchymal stem cells. Gene Expr. 2007;13(4-5):217-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Selmani Z, Naji A, Zidi I, Favier B, Gaiffe E, Obert L, et al. Human leukocyte antigen-G5 secretion by human mesenchymal stem cells is required to suppress T lymphocyte and natural killer function and to induce CD4+CD25highFOXP3+ regulatory T cells. Stem Cells. 2008;26(1):212-22. [DOI] [PubMed] [Google Scholar]
  • 57. Ryan AE, Lohan P, O’Flynn L, Treacy O, Chen X, Coleman C, et al. Chondrogenic differentiation increases antidonor immune response to allogeneic mesenchymal stem cell transplantation. Mol Ther. 2014;22(3):655-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Chen CT. Cartilage extracellular matrix and integrity and OA. In: Rothschild Bruce M., editor. Principles of osteoarthritis: its definition, character derivation and modality-related recognition. IntechOpen; 2012. [Google Scholar]
  • 59. Mow VC, Wang CC, Hung CT. The extracellular matrix, interstitial fluid and ions as a mechanical signal transducer in articular cartilage. Osteoarthritis Cartilage. 1999;7(1):41-58. [DOI] [PubMed] [Google Scholar]
  • 60. Kuse R, Schuster S, Schübbe H, Dix S, Hausmann K. Blood lymphocyte volumes and diameters in patients with chronic lymphocytic leukemia and normal controls. Blut. 1985;50(4):243-8. [DOI] [PubMed] [Google Scholar]
  • 61. Wolf K, Müller R, Borgmann S, Bröcker E-B, Friedl P. Amoeboid shape change and contact guidance: T-lymphocyte crawling through fibrillar collagen is independent of matrix remodeling by MMPs and other proteases. Blood. 2003;102(9):3262-9. [DOI] [PubMed] [Google Scholar]
  • 62. Wiradjaja F, DiTommaso T, Smyth I. Basement membranes in development and disease. Birth Defects Res C Embryo Today. 2010;90(1):8-31. [DOI] [PubMed] [Google Scholar]
  • 63. Howat WJ, Holmes JA, Holgate ST, Lackie PM. Basement membrane pores in human bronchial epithelium: a conduit for infiltrating cells? Am J Pathol. 2001;158(2):673-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Linn FC, Sokoloff L. Movement and composition of interstitial fluid of cartilage. Arthritis Rheum. 1965;8:481-94. [DOI] [PubMed] [Google Scholar]
  • 65. Pedrozo HA, Schwartz Z, Gomez R, Ornoy A, Xin-Sheng W, Dallas SL, et al. Growth plate chondrocytes store latent transforming growth factor (TGF)-beta 1 in their matrix through latent TGF-beta 1 binding protein-1. J Cell Physiol. 1998;177(2):343-54. [DOI] [PubMed] [Google Scholar]
  • 66. Fortier LA, Barker JU, Strauss EJ, McCarrel TM, Cole BJ. The role of growth factors in cartilage repair. Clin Orthop Relat Res. 2011;469(10):2706-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Gharravi AM, Orazizadeh M, Hashemitabar M. Direct expansion of chondrocytes in a dynamic three-dimensional culture system: overcoming dedifferentiation effects in monolayer culture. Artif Organs. 2014;38(12):1053-8. [DOI] [PubMed] [Google Scholar]
  • 68. Rayahin JE, Buhrman JS, Zhang Y, Koh TJ, Gemeinhart RA. High and low molecular weight hyaluronic acid differentially influence macrophage activation. ACS Biomater Sci Eng. 2015;1(7):481-93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Niederkorn JY. Emerging concepts in CD8(+) T regulatory cells. Curr Opin Immunol. 2008;20(3):327-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Cone RE, Li X, Sharafieh R, O'Rourke J, Vella AT. The suppression of delayed-type hypersensitivity by CD8+ regulatory T cells requires interferon-gamma. Immunology. 2007;120(1):112-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Papatriantafyllou M, Moldenhauer G, Ludwig J, Tafuri A, Garbi N, Hollmann G, et al. Dickkopf-3, an immune modulator in peripheral CD8 T-cell tolerance. Proc Natl Acad Sci U S A. 2012;109(5):1631-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Kezuka T, Streilein JW. In vitro generation of regulatory CD8+ T cells similar to those found in mice with anterior chamber-associated immune deviation. Invest Ophthalmol Vis Sci. 2000;41(7):1803-11. [PubMed] [Google Scholar]
  • 73. Wilbanks GA, Streilein JW. Characterization of suppressor cells in anterior chamber-associated immune deviation (ACAID) induced by soluble antigen. Evidence of two functionally and phenotypically distinct T-suppressor cell populations. Immunology. 1990;71(3):383-9. [PMC free article] [PubMed] [Google Scholar]
  • 74. Cong L, Wang SF, Zhao ZL, Yang RY. Donor-antigen inoculation in the testis promotes skin allograft acceptance induced by conventional costimulatory blockade via induction of CD8 + CD122+ and CD4 + CD25+ regulatory T cells. Transplantation. 2016;100(4):763-71. [DOI] [PubMed] [Google Scholar]
  • 75. Rifa’i M, Shi Z, Zhang SY, Lee YH, Shiku H, Isobe K, et al. CD8+CD122+ regulatory T cells recognize activated T cells via conventional MHC class I-alphabetaTCR interaction and become IL-10-producing active regulatory cells. Int Immunol. 2008;20(7):937-47. [DOI] [PubMed] [Google Scholar]
  • 76. Fujihara Y, Takato T, Hoshi K. Macrophage-inducing FasL on chondrocytes forms immune privilege in cartilage tissue engineering, enhancing in vivo regeneration. Stem Cells. 2014;32(5):1208-19. [DOI] [PubMed] [Google Scholar]
  • 77. Langer F, Czitrom A, Pritzker KP, Gross AE. The immunogenicity of fresh and frozen allogeneic bone. J Bone Joint Surg Am. 1975;57(2):216-20. [PubMed] [Google Scholar]
  • 78. Stevenson S, Shaffer JW, Goldberg VM. The humoral response to vascular and nonvascular allografts of bone. Clin Orthop Relat Res. 1996;326:86-95. [DOI] [PubMed] [Google Scholar]
  • 79. Pomajzl RJ, Baker EA, Baker KC, Fleischer MM, Salisbury MR, Phillips DM, et al. Case series with histopathologic and radiographic analyses following failure of fresh osteochondral allografts of the talus. Foot Ankle Int. 2016;37(9):958-67. [DOI] [PubMed] [Google Scholar]
  • 80. Gortz S, Bugbee WD. Fresh osteochondral allografts: graft processing and clinical applications. J Knee Surg. 2006;19(3):231-40. [DOI] [PubMed] [Google Scholar]
  • 81. Lewandrowski KU, Rebmann V, Pässler M, Schollmeier G, Ekkernkamp A, Grosse-Wilde H, et al. Immune response to perforated and partially demineralized bone allografts. J Orthop Sci. 2001;6(6):545-55. [DOI] [PubMed] [Google Scholar]
  • 82. Ambra LF, de Girolamo L, Gomoll AH. Pulse lavage fails to significantly reduce bone marrow content in osteochondral allografts: a histological and DNA quantification study. Am J Sports Med. 2019;47(11):2723-8. [DOI] [PubMed] [Google Scholar]
  • 83. Kandel RA, Gross AE, Ganel A, McDermott AG, Langer F, Pritzker KP. Histopathology of failed osteoarticular shell allografts. Clin Orthop Relat Res. 1985;197:103-10. [PubMed] [Google Scholar]
  • 84. Kemble S, Croft AP. Critical role of synovial tissue-resident macrophage and fibroblast subsets in the persistence of joint inflammation. Front Immunol. 2021;12:715894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Moskalewski S, Hyc A, Osiecka-Iwan A. Immune response by host after allogeneic chondrocyte transplant to the cartilage. Microsc Res Tech. 2002;58(1):3-13. [DOI] [PubMed] [Google Scholar]
  • 86. McCarty EC, Fader RR, Mitchell JJ, Glenn RE, Jr, Potter HG, Spindler KP. Fresh osteochondral allograft versus autograft: twelve-month results in isolated canine knee defects. Am J Sports Med. 2016;44(9):2354-65. [DOI] [PubMed] [Google Scholar]
  • 87. Crist BD, Stoker AM, Pfeiffer FM, Kuroki K, Cook CR, Franklin SP, et al. Optimising femoral-head osteochondral allograft transplantation in a preclinical model. J Orthop Translat. 2016;5:48-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Morris AH, Stamer DK, Kyriakides TR. The host response to naturally-derived extracellular matrix biomaterials. Semin Immunol. 2017;29:72-91. [DOI] [PubMed] [Google Scholar]
  • 89. Cravedi P, Farouk S, Angeletti A, Edgar L, Tamburrini R, Duisit J, et al. Regenerative immunology: the immunological reaction to biomaterials. Transpl Int. 2017;30(12):1199-208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Filippone EJ, McCue PA, Farber JL. Transplant glomerulopathy. Mod Pathol. 2018;31(2):235-52. [DOI] [PubMed] [Google Scholar]
  • 91. Klatt AR, Paul-Klausch B, Klinger G, Kühn G, Renno JH, Banerjee M, et al. A critical role for collagen II in cartilage matrix degradation: collagen II induces pro-inflammatory cytokines and MMPs in primary human chondrocytes. J Orthop Res. 2009;27(1):65-70. [DOI] [PubMed] [Google Scholar]
  • 92. Williams JM, Holzknecht ZE, Plummer TB, Lin SS, Brunn GJ, Platt JL. Acute vascular rejection and accommodation: divergent outcomes of the humoral response to organ transplantation. Transplantation. 2004;78(10):1471-8. [DOI] [PubMed] [Google Scholar]
  • 93. Salama AD, Delikouras A, Pusey CD, Cook HT, Bhangal G, Lechler RI, et al. Transplant accommodation in highly sensitized patients: a potential role for Bcl-xL and alloantibody. Am J Transplant. 2001;1(3):260-9. [DOI] [PubMed] [Google Scholar]
  • 94. Jindra PT, Hsueh A, Hong L, Gjertson D, Shen X-D, Gao F, et al. Anti-MHC class I antibody activation of proliferation and survival signaling in murine cardiac allografts. J Immunol. 2008;180(4):2214-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Dehoux J-P, Gianello P. Accommodation and antibodies. Transplant Immunology. 2009;21(2):106-10. [DOI] [PubMed] [Google Scholar]
  • 96. Lewandowska K, Choi HU, Rosenberg LC, Zardi L, Culp LA. Fibronectin-mediated adhesion of fibroblasts: inhibition by dermatan sulfate proteoglycan and evidence for a cryptic glycosaminoglycan-binding domain. J Cell Biol. 1987;105(3):1443-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Schmidt G, Robenek H, Harrach B, Glössl J, Nolte V, Hörmann H, et al. Interaction of small dermatan sulfate proteoglycan from fibroblasts with fibronectin. J Cell Biol. 1987;104(6):1683-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Hunziker EB, Rosenberg LC. Repair of partial-thickness defects in articular cartilage: cell recruitment from the synovial membrane. J Bone Joint Surg Am. 1996;78(5):721-33. [DOI] [PubMed] [Google Scholar]
  • 99. Zhao S, Zhu W, Xue S, Han D. Testicular defense systems: immune privilege and innate immunity. Cell Mol Immunol. 2014;11(5):428-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Taylor AW. Ocular immune privilege. Eye. 2009;23(10):1885-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Winnall WR, Muir JA, Hedger MP. Rat resident testicular macrophages have an alternatively activated phenotype and constitutively produce interleukin-10 in vitro. J Leukoc Biol. 2011;90(1):133-43. [DOI] [PubMed] [Google Scholar]
  • 102. Bhushan S, Hossain H, Lu Y, Geisler A, Tchatalbachev S, Mikulski Z, et al. Uropathogenic E. coli induce different immune response in testicular and peritoneal macrophages: implications for testicular immune privilege. Plos One. 2011;6(12):e28452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Yamada A, Arakaki R, Saito M, Kudo Y, Ishimaru N. Dual role of Fas/FasL-mediated signal in peripheral immune tolerance. Front Immunol. 2017;8:403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Hashimoto S, Setareh M, Ochs RL, Lotz M. Fas/Fas ligand expression and induction of apoptosis in chondrocytes. Arthritis Rheum. 1997;40(10):1749-55. [DOI] [PubMed] [Google Scholar]
  • 105. Hashimoto H, Tanaka M, Suda T, Tomita T, Hayashida K, Takeuchi E, et al. Soluble Fas ligand in the joints of patients with rheumatoid arthritis and osteoarthritis. Arthritis Rheum. 1998;41(4):657-62. [DOI] [PubMed] [Google Scholar]
  • 106. Maldonado M, Nam J. The role of changes in extracellular matrix of cartilage in the presence of inflammation on the pathology of osteoarthritis. Biomed Res Int. 2013;2013:284873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Medema JP, de Jong J, Peltenburg LT, Verdegaal EM, Gorter A, Bres SA, et al. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc Natl Acad Sci U S A. 2001;98(20):11515-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Seliger B. Strategies of tumor immune evasion. Biodrugs. 2005;19(6):347-54. [DOI] [PubMed] [Google Scholar]
  • 109. BubenÃ-k J. Tumour MHC class I downregulation and immunotherapy (Review). Oncol Rep. 2003;10(6):2005-8. [PubMed] [Google Scholar]
  • 110. Wada A, Tada Y, Kawamura K, Takiguchi Y, Tatsumi K, Kuriyama T, et al. The effects of FasL on inflammation and tumor survival are dependent on its expression levels. Cancer Gene Ther. 2007;14(3):262-7. [DOI] [PubMed] [Google Scholar]
  • 111. Gur-Wahnon D, Borovsky Z, Liebergall M, Rachmilewitz J. The induction of APC with a distinct tolerogenic phenotype via contact-dependent STAT3 activation. Plos One. 2009;4(8):e6846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Gabardi S, Martin ST, Roberts KL, Grafals M. Induction immunosuppressive therapies in renal transplantation. Am J Health Syst Pharm. 2011;68(3):211-8. [DOI] [PubMed] [Google Scholar]
  • 113. Sasazuki T, Juji T, Morishima Y, Kinukawa N, Kashiwabara H, Inoko H, et al. Effect of matching of class I HLA alleles on clinical outcome after transplantation of hematopoietic stem cells from an unrelated donor. Japan Marrow Donor Program. N Engl J Med. 1998;339(17):1177-85. [DOI] [PubMed] [Google Scholar]
  • 114. Min DI, Monaco AP. Complications associated with immunosuppressive therapy and their management. Pharmacotherapy. 1991;11(5):119S-125S. [PubMed] [Google Scholar]
  • 115. Lanza R, Russell DW, Nagy A. Engineering universal cells that evade immune detection. Nat Rev Immunol. 2019;19:723-33. [DOI] [PubMed] [Google Scholar]
  • 116. Prange S, Zucker P, Jevnikar AM, Singh B. Transplanted MHC class I-deficient nonobese diabetic mouse islets are protected from autoimmune injury in diabetic nonobese recipients. Transplantation. 2001;71(7):982-5. [DOI] [PubMed] [Google Scholar]
  • 117. Sake HJ, Frenzel A, Lucas-Hahn A, Nowak-Imialek M, Hassel P, Hadeler KG, et al. Possible detrimental effects of beta-2-microglobulin knockout in pigs. Xenotransplantation. 2019;26(6):e12525. [DOI] [PubMed] [Google Scholar]
  • 118. Figueiredo C, Blasczyk R. A future with less HLA: potential clinical applications of HLA-universal cells. Tissue Antigens. 2015;85(6):443-9. [DOI] [PubMed] [Google Scholar]
  • 119. Deuse T, Seifert M, Phillips N, Fire A, Tyan D, Kay M, et al. Human leukocyte antigen I knockdown human embryonic stem cells induce host ignorance and achieve prolonged xenogeneic survival. Circulation. 2011;124(11 Suppl):S3-9. [DOI] [PubMed] [Google Scholar]
  • 120. Mattapally S, Pawlik KM, Fast VG, Zumaquero E, Lund FE, Randall TD, et al. Human leukocyte antigen class I and II knockout human induced pluripotent stem cell-derived cells: universal donor for cell therapy. J Am Heart Assoc. 2018;7(23):e010239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Gornalusse GG, Hirata RK, Funk SE, Riolobos L, Lopes VS, Manske G, et al. HLA-E-expressing pluripotent stem cells escape allogeneic responses and lysis by NK cells. Nat Biotechnol. 2017;35(8):765-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Nancy P, Tagliani E, Tay C-S, Asp P, Levy DE, Erlebacher A. Chemokine gene silencing in decidual stromal cells limits T cell access to the maternal-fetal interface. Science. 2012;336(6086):1317-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Raghu H, Lepus CM, Wang Q, Wong HH, Lingampalli N, Oliviero F, et al. CCL2/CCR2, but not CCL5/CCR5, mediates monocyte recruitment, inflammation and cartilage destruction in osteoarthritis. Ann Rheum Dis. 2017;76(5):914-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Cutolo M, Sulli A, Paolino S, Pizzorni C. CTLA-4 blockade in the treatment of rheumatoid arthritis: an update. Expert Rev Clin Immunol. 2016;12(4):417-25. [DOI] [PubMed] [Google Scholar]
  • 125. Conigliaro P, Scrivo R, Valesini G, Perricone R. Emerging role for NK cells in the pathogenesis of inflammatory arthropathies. Autoimmun Rev. 2011;10(10):577-81. [DOI] [PubMed] [Google Scholar]
  • 126. Cifuentes-Rius A, Desai A, Yuen D, Johnston APR, Voelcker NH. Inducing immune tolerance with dendritic cell-targeting nanomedicines. Nat Nanotechnol. 2021;16(1):37-46. [DOI] [PubMed] [Google Scholar]
  • 127. Zhang W, Gorantla VS, Campbell PG, Li Y, Yang Y, Komatsu C, et al. Biopatterned CTLA4/Fc matrices facilitate local immunomodulation, engraftment, and glucose homeostasis after pancreatic islet transplantation. Diabetes. 2016;65(12):3660-6. [DOI] [PubMed] [Google Scholar]
  • 128. Lewis JS, Stewart JM, Marshall GP, Carstens MR, Zhang Y, Dolgova NV, et al. Dual-sized microparticle system for generating suppressive dendritic cells prevents and reverses Type 1 diabetes in the nonobese diabetic mouse model. ACS Biomater Sci Eng. 2019;5(5):2631-46. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cartilage are provided here courtesy of SAGE Publications

RESOURCES