SUMMARY
Our understanding of the role of B cells in organ transplantation remains incomplete and continues to grow. The majority of research has focused on the detrimental role of antibodies that drive the development of pathogenesis of the transplanted organ. However, it has been shown that not all donor-specific antibodies are harmful and in some circumstances can even promote tolerance through the mechanism of accommodation. Furthermore, B cells can have effects on transplanted organs through their interaction with T cells, namely antigen presentation, cytokine production, and costimulation. More recently, the role and importance of Bregs was introduced to the field of transplantation. Due to this functional and ontogenetic heterogeneity, targeting B cells in transplantation may bring undesired immunologic side effects including increased rejection. Therefore, the selective control of B cells that contribute to the humoral response against donor antigens will continue to be an important and challenging area of research and potentially lead to improved long-term transplant outcomes.
Keywords: alloantibody, antibody-mediated rejection, B cell, tolerance
Introduction
Historical perspectives—B cells in transplant rejection
In 1990, a Nobel Prize was awarded to Drs. Murray and Thomas “for their discoveries concerning organ and cell transplantation in the treatment of human disease,” celebrating the benefits of clinical transplantation. Over the last 30 years, the number of transplants has increased even further, with more than 19 000 transplants performed in the United States in 2018 [1]. Kidney allograft survival dramatically improved between 1956 and 1990, partially due to advancement of immunosuppressive agents that target T lymphocytes. One-year unadjusted graft survival now exceeds 97% and 93% for primary living and deceased donor kidneys, respectively [2,3]. However, the rate of improvement of long-term graft survival over the past five decades does not follow the remarkable positive trend of short-term graft survival in organ transplantation (Figs 1 and 2).
Figure 1.

A schematic and simplified view of the different pathways through which B cells contribute to transplant rejection. B cells contribute to allograft rejection after differentiating into antibody-secreting plasma cells (blue). Additionally, B cells shape the T-cell response through a combination of antigen presentation, cytokine production, and costimulation (green). Lastly, B cells have direct effects on the allograft that can be initiated by an ischemic injury (purple).
Figure 2.

Overview of commonly used pharmacological agents targeting B cells during different developmental stages.
The gradual loss of graft function has been described by various terms and is most often attributed to chronic rejection. As reviewed by our group and others, the etiology of chronic rejection is multifactorial [4–6] and includes progression of underlying kidney disease, drug toxicity, and immune injury. In his commentary on an earlier review by us, Paul Terasaki stated, “The mantra, ‘chronic rejection is multifactorial’ is the major reason for the lack of progress in reducing the rate of chronic rejection these past 30 years.” [7]. By this, he was claiming that antibody was the sole important cause of graft failure rather than other etiologies, and perhaps reacting to the emphasis on the T cell as the agent of rejection. Alloantibody-induced pathogenesis had been initially recognized in the 1960s by Patel and Terasaki [8], who showed that donor-specific antibodies (DSAs) were associated with immediate kidney transplantation failure. Later, Terasaki and Cai [9,10] showed that human leukocyte antigen (HLA) antibodies are associated with chronic rejection. As they claimed, the T-cell-centric concept is deeply ingrained in the transplant community, and alloantibody or B cells had not been fully considered as a major barrier to tolerance until recently.
Current perspectives - B cells in organ transplantation
B cells were initially considered to be associated with graft rejection but were not considered the major component of rejection or tolerance in organ transplantation but rather an adjunct to T-cell-mediated rejection [11,12]. These early conclusions were mainly due to the more obvious role of cellular immunity under suboptimal or no immunosuppression in early graft rejection [11].
In the current immunosuppressive era with low rates of acute cellular rejection, the presence of alloantibody remains associated with poorer outcomes [13]. Post-transplant donor-specific antibody (DSA) and de novo DSA (dnDSA) are major risk factors and barriers to long-term stable graft survival [14,15]. Once DSA develops, almost 40% of affected patients lose their graft in contrast to patients with no dnDSA [16]. Furthermore, patients with preformed DSA, who comprise 40% of transplant waitlists, showed higher risk of rejection, either acute or chronic antibody-mediated rejection (ABMR) regardless of type of organ transplantation [17–19]. Alloantibody is also a major barrier to transplant tolerance. Conceptually, B cells and their downstream effector plasma cells (PCs) play a major role in acute and chronic ABMR [20]. Memory B cells rapidly differentiate to PCs following a secondary anamnestic response [21]. The clinical impact of B cells and antibodies, especially PCs that secrete antibodies against donor antigens, including HLA and non-HLA–specific antibodies, has received increasing attention in the past decade [22–28]. This has included a defined B-cell signature associated with clinical kidney transplant tolerance [29]. Given the association of a B-cell signature with tolerance, the B cell and its associated alloimmune response seems to be a key determinant of stable long-term graft survival or operational tolerance.
B cells are functionally heterogeneous populations. Unique B-cell subsets have been defined based on location, ability to migrate, and contribution to T-dependent or T-independent response [30,31]. However, some B-cell subtypes are well known for their regulatory effect via IL-10, IL-35, or TGF-ß, including transitional B cells, B10 effector cells, and other regulatory B cells [32–35]. In this review, we will focus on potential detrimental and beneficial B-cell functions/mechanisms in transplantation.
Antibodies in organ transplantation
Alloantibody as a barrier to transplant
Early in clinical transplantation, it was recognized that pretransplant cytotoxic levels of donor-specific HLA antibody (DSA) were associated with an increased risk for hyperacute rejection and/or allograft dysfunction across all organ types [8,36]. As more sensitive solid-phase immunoassays for DSA detection were developed, the correlation between DSA positivity and ABMR or allograft loss diminished [37–41]. However, surveillance biopsies and more sensitive methods for detecting antibody-mediated injury reestablished a link between ABMR and diminished long-term survival in DSA-positive transplants [42–47]. The decision to transplant across a DSA barrier or wait for a more compatible organ depends on many factors, including the urgency for transplantation, center size, and risk-aversion policy, and center infrastructure that permits rapid initiation of antirejection therapy and close post-transplant monitoring of protocol biopsies and DSA. Single and multicenter studies have developed risk stratifications correlating preformed DSA strength at initiation of desensitization or time of transplantation with risk of ABMR and reduced allograft survival [48–51]. Detailed examinations of DSA characteristics have identified a greater risk for allograft loss and ABMR with complement-fixing DSA and IgG3 subclass DSA [24,52–55]. However, even in the absence of DSA at time of transplantation, the risk for ABMR in sensitized candidates may not be completely eliminated, given the possibility of unrecognized HLA-specific memory with the potential for recall responses immediately post-transplant [56–59]. Risk assessments for humoral alloimmunity require a full assessment of the patient’s current and past sensitization events as well as the overall quality of the transplanted organ to include HLA mismatch [60]. Therefore, while avoiding DSA is preferred, it may not be possible in very broadly sensitized transplant candidates even in the era of kidney paired donation and broader deceased donor organ sharing across larger geographical regions [61,62]. Whether preformed DSA represents a barrier to transplantation is multifactorial and must be determined in the context of the patient, the organ type, and the transplant center.
The development of post-transplant de novo DSA can occur in the absence of pretransplant HLA sensitization and has been shown to impact long-term allograft survival. DSA that arises post-transplant is primarily directed toward donor HLA class II mismatches and occurs in the setting of inadequate immunosuppression and/or increased HLA class II mismatching [63–72]. The incidence of de novo DSA depends upon allograft type ranging from 12% in primary kidney transplants with a median time to development of 4 years [16] and up to 30% in lung recipients within the first year post-transplant [45,73]. De novo DSA is more strongly associated with ABMR and allograft loss and thus appears to be more pathologic than preformed DSA. The reasons for this observation may reflect the generation of antibodies with higher specificity and affinity for mismatched donor HLA, increased immunogenicity of HLA class II molecules, or the upregulation and exposure to donor HLA class II in the context of inflammation and infection [45,73–76]. Greater attention to alloimmune risk assessments at time of transplant may better inform individualized immunosuppression and post-transplant monitoring strategies to detect incomplete suppression of humoral alloimmune response and avoid DSA formation [60,74].
Accommodation: enigmatic role of alloantibody
In contrast to the above-described harmful effects of alloantibodies to vascularized grafts, under some circumstances humoral immunity causes little or no damage to an organ graft. Recent studies have shown that 30% of nonsensitized patients develop de novo DSA post-transplantation without demonstrating clinical signs of rejection [63,77]. This condition is referred to as accommodation [78,79]. Accommodation describes a biologic state in which grafts function despite noxious stimuli, like alloantibodies, against them, which was first described in the 1980s in the context of clinical ABO incompatible renal transplantation [80,81].
Accommodation can be mediated by the graft or by the host. The proposed mechanism of host accommodation includes a qualitative change in the humoral immune response with altered affinity and/or specificity for the graft [82]. One example in humans is the shift of IgG subclass to IgG2, which inefficiently activates complement and therefore indirectly blocks the effect of more cytotoxic IgG subclasses, as described by Yu et al.[83]. However, data also suggest that a healthy balance between complement-fixing and noncomplement-fixing antibodies may be required to induce accommodation [84]. A xenotransplantation model has shown evidence that antibodies against the graft are required to induce accommodation, and accommodation was not evident when anti-donor antibodies were suppressed [85]. In a cardiac xenograft model, investigators hypothesized that control of the complement cascade can support accommodation [86]. In sensitized murine models, investigators observed durable accommodation in heart and kidney allotransplantation models with terminal complement inhibition (anti-C5 mAb) [87,88]. Other observations in sensitized human renal transplant recipients have shown that removal of anti-HLA antibodies by immunoadsorption prior to transplantation is leading to accommodation in selected patients [89]. Subsequently, it was postulated that graft exposure with a low concentration of DSAs induces accommodation instead of causing graft injury. This was demonstrated in an in vitro model by Salama et al. [90], who identified Bclxl in graft endothelial cells as a possible key pathway involved. Recent data confirmed that 72% of nonsensitized pediatric kidney recipients showed evidence of de novo DSA within the graft when DSA was present in the serum, but the presence of graft DSAs was not, per se, predictive of graft loss [91].
Therefore, accommodation should not be interpreted as resistance to injury but is better described as a process that repairs injury and regenerates tissue functions. Barbosa et al. [92] summarized this new model of accommodation as a period of vulnerability countered by transiently induced cytoprotection that is followed by ongoing loss of vulnerability, reflecting persistent cellular, and biochemical changes. Viewed in this way, it is understandable that excess accommodation can also have downsides that include a lack of viral control and control of malignant tumor cells as described for hepatitis C and multiple cancer entities [93–95].
From the above-described understanding, modern strategies focus on controlling but not inhibiting the interaction between circulating antibodies and the graft. However, treatment strategies involving the complement cascade (eculizumab) or targeted plasma cell inhibition (bortezomib) have only been partially successful to date [96]. Overall, our understanding of accommodation is limited and largely driven by data generated before the current era of highly sensitive assays to measure allospecific antibodies, protocol graft biopsies, and more precise histologic criteria for ABMR.
The role of B cells beyond antibody
For a more complete understanding of B cells and their role in transplantation, aspects of B-cell biology other than antibody production must be considered. Setting antibody aside, B cells function in other ways, some of which promote rejection, and some of which have the potential to promote tolerance.
B cells as antigen-presenting cells
Although chronic rejection is commonly considered an antibody-mediated process, Zeng et al. [97] have provided evidence that B cells are capable of promoting chronic rejection independently of antibody production. In a mouse model of heart allotransplantation using costimulation blockade-based immunosuppression (CTLA4Ig and anti-CD40L), they showed that animals genetically modified to lack circulating antibody (AID/μSKO mice) still went on to develop chronic allograft vasculopathy (CAV), a pathognomonic feature of chronic rejection found in heart, kidney, liver, and pancreas transplantation. On the other hand, animals genetically modified to lack both B cells and circulating antibody (μMT mice) were protected from CAV. CAV could be elicited in μMT mice by infusing B cells from AID/μS KO mice, even though these B cells were incapable of antibody production. One possible mechanism to account for this phenomenon could be the role of B cells as antigen-presenting cells. In another study of mouse cardiac allotransplantation, Noorchasm and colleagues reported that indirect alloantigen presentation by recipient B cells plays a critical role in the activation of alloreactive CD4+ T cells [98]. In mice, B cells have also been reported to play an important role in helping alloreactive CD4+ and CD8+ T cells differentiate into memory T cells [99]. In general, B cells may contribute to allograft rejection independently of antibody production by shaping the T-cell response through a combination of antigen presentation, cytokine production, and costimulation [100–103]. Additionally, one important role of B cells may include supporting the basic architecture of lymphoid tissue to allow optimal interaction between T cells, dendritic cells, and other components of the immune response [100–102].
B-cell response to tissue injury
The traditionally understood pathological role of B cells in transplantation is about B-cell differentiation into antibody-secreting cells. However, as described above, B-cell function as antigen-presenting cells was also recognized [9,97]. Additionally, it is well known that B cells acutely respond to ischemia/reperfusion injury [104–106]. Recently, Cippa et al. [107] suggested that kidney injury in ischemia/reperfusion injury and transplantation are both mediated by a B-cell response to dysfunctional tissue repair. Interestingly, patients who developed chronic rejection already showed elevated B-cell activities and other common gene signatures for acute kidney injury including genes related to fibrosis (e.g., COL1A1, DPT, and MMP7) and inflammation (e.g., CD52, CXCL10, and CCL21). Based on a nontransplant (ischemic injury) mouse model, they found that the B-cell response to tissue injury is able to contribute to chronic kidney injury or chronic rejection in the absence of an alloimmune response. Based on transcriptional analyses, it was hypothesized that memory B cells infiltrate, expand, and gradually switch to a plasma cell population after ischemic injury and later became CD138 negative polyclonal B cells. Such cells may play a crucial role in developing ectopic germinal centers in the kidney, causing chronic kidney injury. Overall, B cells could have an important role in late immune-mediated kidney injury and repair responses including chronic rejection.
Role of B cells in promoting tolerance
B cells are often thought to boost inflammatory responses. However, like their counterpart T cell, B cells can also suppress the immune response. Originally, B cells with immune regulatory function were identified by their function, such as their capacity to produce inhibitory cytokines (IL-10). Additionally, their ability to induce or recruit regulatory T cells (Tregs) has recently been implicated [108,109].
In transplantation, B cells were previously thought to have only a pathogenic role; however, growing evidence demonstrates that B cells may play a pivotal role in the induction and maintenance of transplant tolerance [110]. Regulatory B cells (Bregs) have proven their importance in controlling immunity in a number of mouse models of allergy and autoimmunity [103,111,112]. Although a number of subsets have been described as Bregs, the two B-cell subsets that are best characterized are CD5+CD19+CD1dhi B10 and CD19+CD21hiCD23hi CD24hi transitional-2 (T2) Breg cells. While no true consensus definition of Bregs has been agreed upon, characterizations generally center around the secretion of IL-10 [113]. Likewise, no signature set of phenotypic markers for Bregs is analogous to the markers CD25 and FoxP3 that characterize Tregs. Candidate phenotypic profiles for Bregs include CD5+CD1dhi B cells and T-cell Ig and mucin domain protein 1 (TIM-1+) B cells [114–116]. Mechanistically, Bregs function through IL-10 and through secretion of other cytokines (TGF-β, IL-35) to suppress CD4+ T-cell proliferation, suppress CD8+ effector T-cell function, induce T-cell apoptosis through binding the FAS and PD-1 receptors, induce Tregs, suppress antigen-presenting and cytokine secretion by dendritic cells and M1 macrophages, and suppress natural killer (NK) cells and neutrophils [113]. Evidence for Bregs’ salutary effect is sometimes inferred from experiments involving pan-B-cell depletion, which has been shown to accelerate rejection in models of heart and skin allotransplantation [117–119] and in human clinical heart and kidney transplant trials [120,121]. In accordance with this, agents that target specific Breg cell populations, such as daratumumab (anti-CD38mAb), showed skewing toward memory T-cell dominance in multiple myeloma [122] and accelerated T-cell-mediated rejection in a NHP model [123] possibly by reducing immune regulatory cells including Bregs, which are known to express CD38 [124,125]. More direct evidence for the role of Bregs in contributing to tolerance comes mainly from B-cell profiling in operationally tolerant transplant patients.
Recently, Lino et al. [126] characterized a subset of “natural regulatory plasma cells” identified by expression of the marker LAG-3. These cells appear to be a pre-existing “natural” subset and rapidly produce IL-10 in response to toll-like receptor stimulation. They also express the inhibitory receptors PD-L1, PD-L2, and CD200. Finally, B cells have been identified as the critical antigen-presenting cells involved in anergizing CD4+ memory T cells. In work by Dalai et al. [127], B2 follicular B cells—not DCs—were responsible for inducing anergy in the CD4+ memory T-cell compartment in a mouse model.
B-cell signature of transplant tolerance
A major goal of the transplant community has been to identify a “signature” of tolerance in transplant patients. This might allow patients demonstrating this “tolerance signature” to wean off immunosuppression and maintain normal graft function. While much of the focus in studying operational tolerance has emphasized the role of T cells, recent evidence has pointed to a B-cell signature of tolerance in transplant recipients. To study this phenomenon, a population of operationally tolerant kidney transplant patients who were no longer taking immunosuppression but maintained normal graft function were identified and compared to control patients that remained on immunosuppression with stable graft function. In 2010, two groups identified a transcriptional signature in peripheral blood showing that upregulation of B-cell-related genes and their molecular pathways were associated with tolerance [29,128]. Furthermore, Newell et al. [29] demonstrated that this B-cell signature had upregulated in cells found in urine and found increased numbers of naive and transitional B cells in peripheral blood by flow cytometry. Additional work by these groups and others has identified that operationally tolerant patients have distinct B-cell phenotypes in peripheral blood that exhibit a more regulatory phenotype than patients with stable graft function on immunosuppression [129–132].
Collectively, these studies have suggested a role for B cells, in particular Bregs, in promoting or maintaining tolerance in kidney transplant recipients. Several studies in animal models have also shown a key role for B cells in transplantation tolerance since B-cell depletion can prevent tolerance induction by various methods, perhaps due to the loss of immune regulation by B cells [115,118,133,134]. More recent evidence in the operationally tolerant human kidney transplant recipients has identified the same B-cell signature of tolerance by transcriptional analysis of peripheral blood in both spontaneously tolerant individuals as well as those who underwent specific tolerance induction protocols [135]. In addition, a significant proportion of patients with stable long-term graft function while on immunosuppression demonstrated this B-cell tolerance signature [135–137]. A recent meta-analysis of the gene signatures of operationally tolerant kidney transplant patients validated the concept that the majority of biomarkers associated with tolerance were in fact B-cell-related [138].
B-cell targeting, pros and cons
B-cell-targeted therapies have shown success in treating and reducing the incidence of ABMR, treating sensitized patients, and inducing tolerance. However, these advantages are balanced with shortcomings of solely targeting B cells owing to their regulatory role and interaction with other immune cells.
Investigations into preventing and treating ABMR have gained traction in the transplant community for this leading cause of graft failure. Rituximab (anti-CD20 monoclonal antibody) has been used prominently in renal transplantation for the treatment of ABMR, often in conjunction with IVIG, plasmapheresis (PP), or other agents, to prevent and reduce the incidence of ABMR [139,140]. Other solid organ transplant specialists have followed suit [141,142]. Traditionally, desensitization regimens relied on the removal of DSAs through PP, which is costly, invasive, and difficult to apply pretransplant in deceased donor transplantation [143]. B-cell targeting provides a noninvasive alternative approach. In a randomized controlled study of rituximab by van den Hoogen et al. [143], sensitized patients experienced less rejection episodes compared to the placebo group. The role of B cells extends to the controversial topic of transplantation tolerance. Liu et al. [117] showed that B-cell depletion therapy might provoke a bias toward a regulatory phenotype and promote long-term islet allograft survival in nonhuman primates. A subsequent study showed that a possible explanation for this earlier finding might be that Bregs promote Treg cell development via TGF-b production [144]. There are ongoing investigations to explain these findings.
Limitations of B-cell-targeted therapies warrant a discussion to optimize their use in the clinical setting. Several studies have found that B-cell-focused therapies ignore the importance of T-cell-directed immune responses, resulting in allograft rejection and tolerance failure [118,119]. Clatworthy et al. [121] examined the benefit of B-cell depletion by using rituximab as a sole induction therapy in a randomized controlled trial, but the study was halted after a high incidence of acute cellular rejection was noted in the rituximab group. The authors hypothesized that depletion led to a loss of Breg activity and subsequent cytokine storm that activated T cells. However, the study’s enrollment was small and the findings would need to be replicated with larger cohorts to confirm these findings. Furthermore, B-cell depletion with rituximab does not address PCs. Rituximab has been added to desensitization regimens to deplete B cells, reduce PC generation, and prevent antibody production in sensitized patients [145]. However, B cells no longer express CD20 once they differentiate into PCs and become “out of reach” of rituximab [146,147]. Clinically, depletional therapies with rituximab have many side effects, such as heightened risk of infection leading to sepsis [148,149] and cardiovascular disease (CVD) [150–152]. CVD is emerging as a highly morbid complication of rituximab-treated patients. Tyden and colleagues showed in their randomized controlled study that the rituximab group experienced higher mortality due to CVD with no difference in the incidence of AMR, T-cell-mediated rejection, or de novo DSA production. Kyaw et al. [151] showed that a possible explanation of such finding may be due to the depletion of atheroprotective B-lymphocyte populations with rituximab. Notably, the incidence of CVD in rituximab-treated patients is not limited to transplant patients; many cases have been reported in clinical trials in non-Hodgkin’s lymphoma, chronic lymphocytic leukemia, and rheumatoid arthritis, among others [152].
Concluding remarks
Clearly, alloantibody is detrimental for long-term graft survival. The plethora of new agents targeting either the B cell, plasma cell, related cytokines, or complement has led to new transplantation research and clinical trials. B cells fall into several functionally distinct subpopulations, and broad B-cell depletion may bring undesired immunologic side effects including increased rejection. Understanding how to selectively control B cells that contribute to the humoral response against donor antigens will continue to be an important and challenging area of research and potentially lead to improved long-term transplant outcomes.
Acknowledgment
We would like to gratefully acknowledge Lauren Halligan (Department of Surgery, Duke University) for her contribution in creating the illustrations in the manuscript.
Funding
This work was partially supported by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health as part of the Nonhuman Primate Transplantation Tolerance Cooperative Study Group under U19AI131471 (awarded to S.J.K.) and Opportunities Pool Round 13 (awarded to J.K.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Footnotes
Conflicts of interest
The authors have declared no conflicts of interest.
REFERENCES
- 1.OPTN/SRTR 2017 annual data report: introduction. Am J Transplant 2019; 19 (Suppl. 2): 11. [DOI] [PubMed] [Google Scholar]
- 2.Hart A, Smith JM, Skeans MA, et al. OPTN/SRTR 2017 annual data report: kidney. Am J Transplant 2019; 19 (Suppl. 2): 19. [DOI] [PubMed] [Google Scholar]
- 3.Wang JH, Skeans MA, Israni AK. Current status of kidney transplant outcomes: dying to survive. Adv Chronic Kidney Dis 2016; 23: 281. [DOI] [PubMed] [Google Scholar]
- 4.Kwun J, Knechtle SJ. Overcoming chronic rejection-can it B? Transplantation 2009; 88: 955. [DOI] [PubMed] [Google Scholar]
- 5.Chong AS, Rothstein DM, Safa K, Riella LV. Outstanding questions in transplantation: B cells, alloantibodies, and humoral rejection. Am J Transplant 2019; 19: 2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bohmig GA, Eskandary F, Doberer K, Halloran PF. The therapeutic challenge of late antibody-mediated kidney allograft rejection. Transpl Int 2019; 32: 775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Terasaki PI. The review by Kwun and Knechtle-“can it B?”-asks whether B cells are responsible for chronic rejection of transplants. Transplantation 2009; 88: 978. [DOI] [PubMed] [Google Scholar]
- 8.Patel R, Terasaki PI. Significance of the positive crossmatch test in kidney transplantation. N Engl J Med 1969; 280: 735. [DOI] [PubMed] [Google Scholar]
- 9.Terasaki PI, Cai J. Human leukocyte antigen antibodies and chronic rejection: from association to causation. Transplantation 2008; 86: 377. [DOI] [PubMed] [Google Scholar]
- 10.Terasaki P, Lachmann N, Cai J. Summary of the effect of de novo HLA antibodies on chronic kidney graft failure. Clin Transpl 2006: 455. [PubMed] [Google Scholar]
- 11.Billingham RE, Krohn PL, Medawar PB. Effect of locally applied cortisone acetate on survival of skin homografts in rabbits. Br Med J 1951; 2: 1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Benichou G, Yamada Y, Yun SH, Lin C, Fray M, Tocco G. Immune recognition and rejection of allogeneic skin grafts. Immunotherapy 2011; 3: 757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Pouliquen E, Koenig A, Chen CC, et al. Recent advances in renal transplantation: antibody-mediated rejection takes center stage. F1000Prime Rep 2015; 7: 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zhang R Donor-specific antibodies in kidney transplant recipients. Clin J Am Soc Nephrol 2018; 13: 182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Barten MJ, Schulz U, Beiras Fernandez A, et al. The clinical impact of donor-specific antibodies in heart transplantation. Transplant Rev 2018; 32: 207. [DOI] [PubMed] [Google Scholar]
- 16.Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Rates and determinants of progression to graft failure in kidney allograft recipients with de novo donor-specific antibody. Am J Transplant 2015; 15: 2921. [DOI] [PubMed] [Google Scholar]
- 17.Amico P, Honger G, Mayr M, Steiger J, Hopfer H, Schaub S. Clinical relevance of pretransplant donor specific HLA antibodies detected by single-antigen flow-beads. Transplantation 2009; 87: 1681. [DOI] [PubMed] [Google Scholar]
- 18.Wahrmann M, Bartel G, Exner M, et al. Clinical relevance of preformed C4d-fixing and non-C4d-fixing HLA single antigen reactivity in renal allograft recipients. Transplant Int 2009; 22: 982. [DOI] [PubMed] [Google Scholar]
- 19.Lefaucheur C, Loupy A, Hill GS, et al. Preexisting donor-specific HLA antibodies predict outcome in kidney transplantation. J Am Soc Nephrol 2010; 21: 1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Clatworthy MR, Targeting B. Cells and antibody in transplantation. Am J Transplant 2011; 11: 1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kurosaki T, Kometani K, Ise W. Memory B cells. Nat Rev Immunol 2015; 15: 149. [DOI] [PubMed] [Google Scholar]
- 22.Freischlag K, Pearl MH, Chambers ET. The clinical impact of non-HLA Antibodies in solid organ transplantation. Clin Transpl 2016; 32: 31. [PubMed] [Google Scholar]
- 23.Taner T, Gandhi MJ, Sanderson SO, et al. Prevalence, course and impact of HLA donor-specific antibodies in liver transplantation in the first year. Am J Transplant 2012; 12: 1504. [DOI] [PubMed] [Google Scholar]
- 24.Smith JD, Ibrahim MW, Newell H, et al. Pre-transplant donor HLA-specific antibodies: characteristics causing detrimental effects on survival after lung transplantation. J Heart Lung Transplant 2014; 33: 1074. [DOI] [PubMed] [Google Scholar]
- 25.Haarberg KM, Tambur AR. Detection of donor-specific antibodies in kidney transplantation. Br Med Bull 2014; 110: 23. [DOI] [PubMed] [Google Scholar]
- 26.Zhang Q, Reed EF. The importance of non-HLA antibodies in transplantation. Nat Rev Nephrol 2016; 12: 484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zito A, Schena A, Grandaliano G, Gesualdo L, Schena FP. Increasing relevance of donor-specific antibodies in antibody-mediated rejection. J Nephrol 2013; 26: 237. [DOI] [PubMed] [Google Scholar]
- 28.McKenna RM, Takemoto SK, Terasaki PI. Anti-HLA antibodies after solid organ transplantation. Transplantation 2000; 69: 319. [DOI] [PubMed] [Google Scholar]
- 29.Newell KA, Asare A, Kirk AD, et al. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest 2010; 120: 1836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Plotkin SA. Correlates of protection induced by vaccination. Clin Vaccine Immunol 2010; 17: 1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Rickert RC. Regulation of B lymphocyte activation by complement C3 and the B cell coreceptor complex. Curr Opin Immunol 2005; 17: 237. [DOI] [PubMed] [Google Scholar]
- 32.Mauri C, Menon M. Human regulatory B cells in health and disease: therapeutic potential. J Clin Invest 2017; 127: 772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang RX, Yu CR, Dambuza IM, et al. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med 2014; 20: 633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yoshizaki A, Miyagaki T, DiLillo DJ, et al. Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature 2012; 491: 264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immunity 2015; 42: 607. [DOI] [PubMed] [Google Scholar]
- 36.Williams GM, DePlanque B, Lower R, Hume D. Antibodies and human transplant rejection. Ann Surg 1969; 170: 603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Adebiyi OO, Gralla J, Klem P, et al. Clinical significance of pretransplant donor-specific antibodies in the setting of negative cell-based flow cytometry crossmatching in kidney transplant recipients. Am J Transplant 2016; 16: 3458. [DOI] [PubMed] [Google Scholar]
- 38.Higgins R, Lowe D, Hathaway M, et al. Human leukocyte antigen antibody-incompatible renal transplantation: excellent mediumterm outcomes with negative cytotoxic crossmatch. Transplantation 2011; 92: 900. [DOI] [PubMed] [Google Scholar]
- 39.Schinstock CA, Gandhi M, Cheungpasitporn W, et al. Kidney transplant with low levels of DSA or low positive B-flow crossmatch: an underappreciated option for highly sensitized transplant candidates. Transplantation 2017; 101: 2429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Eng HS, Bennett G, Chang SH, et al. Donor human leukocyte antigen specific antibodies predict development and define prognosis in transplant glomerulopathy. Hum Immunol 2011; 72: 386. [DOI] [PubMed] [Google Scholar]
- 41.Raess M, Frohlich G, Roos M, et al. Donor-specific anti-HLA antibodies detected by Luminex: predictive for short-term but not long-term survival after heart transplantation. Transplant Int 2013; 26: 1097. [DOI] [PubMed] [Google Scholar]
- 42.Musat AI, Agni RM, Wai PY, et al. The significance of donor-specific HLA antibodies in rejection and ductopenia development in ABO compatible liver transplantation. Am J Transplant 2011; 11: 500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kraus ES, Parekh RS, Oberai P, et al. Subclinical rejection in stable positive crossmatch kidney transplant patients: incidence and correlations. Am J Transplant 2009; 9: 1826. [DOI] [PubMed] [Google Scholar]
- 44.Halloran PF, Famulski KS, Reeve J. Molecular assessment of disease states in kidney transplant biopsy samples. Nat Rev Nephrol 2016; 12: 534. [DOI] [PubMed] [Google Scholar]
- 45.Agbor-Enoh S, Jackson AM, Tunc I, et al. Late manifestation of alloantibody-associated injury and clinical pulmonary antibody-mediated rejection: evidence from cell-free DNA analysis. J Heart Lung Transplant 2018; 37: 925. [DOI] [PubMed] [Google Scholar]
- 46.Loupy A, Lefaucheur C. Antibody mediated rejection of solid-organ allografts. N Engl J Med 2018; 379: 1150. [DOI] [PubMed] [Google Scholar]
- 47.Valenzuela NM, Reed EF. Antibodymediated rejection across solid organ transplants: manifestations, mechanisms, and therapies. J Clin Invest 2017; 127: 2492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Vo AA, Sinha A, Haas M, et al. Factors predicting risk for antibody-mediated rejection and graft loss in highly human leukocyte antigen sensitized patients transplanted after desensitization. Transplantation 2015; 99: 1423. [DOI] [PubMed] [Google Scholar]
- 49.Orandi BJ, Luo X, Massie AB, et al. Survival benefit with kidney transplants from HLA-incompatible live donors. N Engl J Med 2016; 374: 940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zeevi A, Lunz J, Feingold B, et al. Persistent strong anti-HLA antibody at high titer is complement binding and associated with increased risk of antibody-mediated rejection in heart transplant recipients. J Heart Lung Transplant 2013; 32: 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Tinckam KJ, Keshavjee S, Chaparro C, et al. Survival in sensitized lung transplant recipients with perioperative desensitization. Am J Transplant 2015; 15: 417. [DOI] [PubMed] [Google Scholar]
- 52.Loupy A, Lefaucheur C, Vernerey D, et al. Complement-binding anti-HLA antibodies and kidney-allograft survival. N Engl J Med 2013; 369: 1215. [DOI] [PubMed] [Google Scholar]
- 53.Lefaucheur C, Viglietti D, Bentlejewski C, et al. IgG donor-specific anti-human HLA antibody subclasses and kidney allograft antibody-mediated injury. J Am Soc Nephrol 2016; 27: 293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhang Q, Hickey M, Drogalis-Kim D, et al. Understanding the correlation between DSA, complement activation, and antibody-mediated rejection in heart transplant recipients. Transplantation 2018; 102: e431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.O’Leary JG, Kaneku H, Banuelos N, Jennings LW, Klintmalm GB, Terasaki PI. Impact of IgG3 subclass and C1q-fixing donor-specific HLA alloantibodies on rejection and survival in liver transplantation. Am J Transplant 2015; 15: 1003. [DOI] [PubMed] [Google Scholar]
- 56.Heidt S, Roelen DL, de Vaal YJ, et al. A NOVel ELISPOT assay to quantify HLA-specific B cells in HLA-immunized individuals. Am J Transplant 2012; 12: 1469. [DOI] [PubMed] [Google Scholar]
- 57.Luque S, Lucia M, Melilli E, et al. Value of monitoring circulating donor-reactive memory B cells to characterize antibody-mediated rejection after kidney transplantation. Am J Transplant 2019; 19: 368. [DOI] [PubMed] [Google Scholar]
- 58.Lucia M, Luque S, Crespo E, et al. Preformed circulating HLA-specific memory B cells predict high risk of humoral rejection in kidney transplantation. Kidney Int 2015; 88: 874. [DOI] [PubMed] [Google Scholar]
- 59.Zachary AA, Kopchaliiska D, Montgomery RA, Melancon JK, Leffell MS. HLA-specific B cells: II. Application to transplantation. Transplantation 2007; 83: 989. [DOI] [PubMed] [Google Scholar]
- 60.Tambur AR, Campbell P, Claas FH, et al. Sensitization in transplantation: assessment of risk (STAR) 2017 working group meeting report. Am J Transplant 2018; 18: 1604. [DOI] [PubMed] [Google Scholar]
- 61.Houp JA, Schillinger KP, Eckstein AJ, et al. Casting a smaller net into a bigger donor pool: a single center’s experience with the new kidney allocation system. Hum Immunol 2017; 78: 49. [DOI] [PubMed] [Google Scholar]
- 62.Stewart DE, Wilk AR, Toll AE, et al. Measuring and monitoring equity in access to deceased donor kidney transplantation. Am J Transplant 2018; 18: 1924. [DOI] [PubMed] [Google Scholar]
- 63.Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Evolution and clinical pathologic correlations of de novo donor-specific HLA antibody post kidney transplant. Am J Transplant 2012; 12: 1157. [DOI] [PubMed] [Google Scholar]
- 64.Wiebe C, Rush DN, Nevins TE, et al. Class II eplet mismatch modulates tacrolimus trough levels required to prevent donor-specific antibody development. J Am Soc Nephrol 2017; 28: 3353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Schinstock CA, Cosio F, Cheungpasitporn W, et al. The value of protocol biopsies to identify patients with de novo donor-specific antibody at high risk for allograft loss. Am J Transplant 2017; 17: 1574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Morrell MR, Pilewski JM, Gries CJ, et al. De novo donor-specific HLA antibodies are associated with early and high-grade bronchiolitis obliterans syndrome and death after lung transplantation. J Heart Lung Transplant 2014; 33: 1288. [DOI] [PubMed] [Google Scholar]
- 67.Smith JD, Banner NR, Hamour IM, et al. De novo donor HLA-specific antibodies after heart transplantation are an independent predictor of poor patient survival. Am J Transplant 2011; 11: 312. [DOI] [PubMed] [Google Scholar]
- 68.Kaneku H, O’Leary JG, Banuelos N, et al. De novo donor-specific HLA antibodies decrease patient and graft survival in liver transplant recipients. Am J Transplant 2013; 13: 1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cheng EY, Everly MJ, Kaneku H, et al. Prevalence and clinical impact of donor-specific alloantibody among intestinal transplant recipients. Transplantation 2017; 101: 873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kubal CA, Mangus R, Ekser B, et al. Class II human leukocyte antigen epitope mismatch predicts de novo donor-specific antibody formation after liver transplantation. Liver Transpl 2018; 24: 1101. [DOI] [PubMed] [Google Scholar]
- 71.Lim WH, Wong G, Heidt S, Claas FHJ. Novel aspects of epitope matching and practical application in kidney transplantation. Kidney Int 2018; 93: 314. [DOI] [PubMed] [Google Scholar]
- 72.Tran A, Fixler D, Huang R, Meza T, Lacelle C, Das BB. Donor-specific HLA alloantibodies: impact on cardiac allograft vasculopathy, rejection, and survival after pediatric heart transplantation. J Heart Lung Transplant 2016; 35: 87. [DOI] [PubMed] [Google Scholar]
- 73.Snyder LD, Wang Z, Chen DF, et al. Implications for human leukocyte antigen antibodies after lung transplantation: a 10-year experience in 441 patients. Chest 2013; 144: 226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wiebe C, Tambur A, Nickerson PW. A call to action-the transplant recipient’s expectation of precision in transplant medicine. Am J Transplant 2018; 18: 2845. [DOI] [PubMed] [Google Scholar]
- 75.Gunasekaran M, Sharma M, Hachem R, Bremner R, Smith MA, Mohanakumar T. Circulating exosomes with distinct properties during chronic lung allograft rejection. J Immunol 2018; 200: 2535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Tiriveedhi V, Gautam B, Sarma NJ, et al. Pre-transplant antibodies to Kalpha1 tubulin and collagen-V in lung transplantation: clinical correlations. J Heart Lung Transplant 2013; 32: 807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gill JS, Landsberg D, Johnston O, et al. Screening for de novo anti-human leukocyte antigen antibodies in nonsensitized kidney transplant recipients does not predict acute rejection. Transplantation 2010; 89: 178. [DOI] [PubMed] [Google Scholar]
- 78.Platt JL, Vercellotti GM, Dalmasso AP, et al. Transplantation of discordant xenografts: a review of progress. Immunol Today 1990; 11: 450; discussion 6–7. [DOI] [PubMed] [Google Scholar]
- 79.Bach FH, Turman MA, Vercellotti GM, Platt JL, Dalmasso AP. Accommodation: a working paradigm for progressing toward clinical discordant xenografting. Transplant Proc 1991; 23: 205. [PubMed] [Google Scholar]
- 80.Koch CA, Khalpey ZI, Platt JL. Accommodation: preventing injury in transplantation and disease. J Immunol 2004; 172: 5143. [DOI] [PubMed] [Google Scholar]
- 81.Alexandre GP, Squifflet JP, De Bruyere M, et al. Present experiences in a series of 26 ABO-incompatible living donor renal allografts. Transplant Proc 1987; 19: 4538. [PubMed] [Google Scholar]
- 82.Dorling A Transplant accommodation–are the lessons learned from xenotransplantation pertinent for clinical allotransplantation? Am J Transplant 2012; 12: 545. [DOI] [PubMed] [Google Scholar]
- 83.Yu PB, Holzknecht ZE, Bruno D, Parker W, Platt JL. Modulation of natural IgM binding and complement activation by natural IgG antibodies: a role for IgG anti-Gal alpha1–3Gal antibodies. J Immunol 1996; 157: 5163. [PubMed] [Google Scholar]
- 84.Dalmasso AP, He T, Benson BA. Inhibition of complement-mediated porcine endothelial cell cytotoxicity by human IgM natural antibody. Transplant Proc 1996; 28: 535. [PubMed] [Google Scholar]
- 85.Hasan R, Van den Bogaerde J, Forty J, Wright L, Wallwork J, White DJ. Xenograft adaptation is dependent on the presence of antispecies antibody, not prolonged residence in the recipient. Transplant Proc 1992; 24: 531. [PubMed] [Google Scholar]
- 86.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: 1471. [DOI] [PubMed] [Google Scholar]
- 87.Wang H, Arp J, Liu W, et al. Inhibition of terminal complement components in presensitized transplant recipients prevents antibody-mediated rejection leading to long-term graft survival and accommodation. J Immunol 2007; 179: 4451. [DOI] [PubMed] [Google Scholar]
- 88.Rother RP, Arp J, Jiang J, et al. C5 blockade with conventional immunosuppression induces long-term graft survival in presensitized recipients. Am J Transplant 2008; 8: 1129. [DOI] [PubMed] [Google Scholar]
- 89.Ross CN, Gaskin G, Gregor-Macgregor S, et al. Renal transplantation following immunoadsorption in highly sensitized recipients. Transplantation 1993; 55: 785. [DOI] [PubMed] [Google Scholar]
- 90.Salama AD, Delikouras A, Pusey CD, et al. Transplant accommodation in highly sensitized patients: a potential role for Bcl-xL and alloantibody. Am J Transplant 2001; 1: 260. [DOI] [PubMed] [Google Scholar]
- 91.Nocera A, Tagliamacco A, Cioni M, et al. Kidney intragraft homing of de novo donor-specific HLA antibodies is an essential step of antibody-mediated damage but not per se predictive of graft loss. Am J Transplant 2017; 17: 692. [DOI] [PubMed] [Google Scholar]
- 92.Garcia de Mattos Barbosa M, Cascalho M, Platt JL. Accommodation in ABO incompatible organ transplants. Xenotransplantation 2018; 25: e12418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Racanelli V, Rehermann B. Hepatitis C virus infection: when silence is deception. Trends Immunol 2003; 24: 456. [DOI] [PubMed] [Google Scholar]
- 94.Junnikkala S, Hakulinen J, Jarva H, et al. Secretion of soluble complement inhibitors factor H and factor H-like protein (FHL-1) by ovarian tumour cells. Br J Cancer 2002; 87: 1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Donin N, Jurianz K, Ziporen L, Schultz S, Kirschfink M, Fishelson Z. Complement resistance of human carcinoma cells depends on membrane regulatory proteins, protein kinases and sialic acid. Clin Exp Immunol 2003; 131: 254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Lonze BE, Dagher NN, Simpkins CE, et al. Eculizumab, bortezomib and kidney paired donation facilitate transplantation of a highly sensitized patient without vascular access. Am J Transplant 2010; 10: 2154. [DOI] [PubMed] [Google Scholar]
- 97.Zeng Q, Ng YH, Singh T, et al. B cells mediate chronic allograft rejection independently of antibody production. J Clin Invest 2014; 124: 1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Noorchashm H, Reed AJ, Rostami SY, et al. B cell-mediated antigen presentation is required for the pathogenesis of acute cardiac allograft rejection. J Immunol 2006; 177: 7715. [DOI] [PubMed] [Google Scholar]
- 99.Ng YH, Oberbarnscheidt MH, Chandramoorthy HC, Hoffman R, Chalasani G. B cells help alloreactive T cells differentiate into memory T cells. Am J Transplant 2010; 10: 1970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Fu YX, Huang G, Wang Y, Chaplin DD. B lymphocytes induce the formation of follicular dendritic cell clusters in a lymphotoxin alpha-dependent fashion. J Exp Med 1998; 187: 1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ngo VN, Cornall RJ, Cyster JG. Splenic T zone development is B cell dependent. J Exp Med 2001; 194: 1649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Nolte MA, Arens R, Kraus M, et al. B cells are crucial for both development and maintenance of the splenic marginal zone. J Immunol 2004; 172: 3620. [DOI] [PubMed] [Google Scholar]
- 103.Lund FE, Randall TD. Effector and regulatory B cells: modulators of CD4+ T cell immunity. Nat Rev Immunol 2010; 10: 236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Cochain C, Auvynet C, Poupel L, et al. The chemokine decoy receptor D6 prevents excessive inflammation and adverse ventricular remodeling after myocardial infarction. Arterioscler Thromb Vasc Biol 2012; 32: 2206. [DOI] [PubMed] [Google Scholar]
- 105.Chen J, Crispin JC, Tedder TF, Dalle Lucca J, Tsokos GC. B cells contribute to ischemia/reperfusion mediated tissue injury. J Autoimmun 2009; 32: 195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Jang HR, Gandolfo MT, Ko GJ, Satpute SR, Racusen L, Rabb H. B cells limit repair after ischemic acute kidney injury. J Am Soc Nephrol 2010; 21: 654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Cippa PE, Liu J, Sun B, Kumar S,Naesens M, McMahon AP. A late B lymphocyte action in dysfunctional tissue repair following kidney injury and transplantation. Nat Commun 2019; 10: 1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bouaziz JD, Yanaba K, Tedder TF.Regulatory B cells as inhibitors of immune responses and inflammation. Immunol Rev 2008; 224: 201. [DOI] [PubMed] [Google Scholar]
- 109.Yanaba K, Bouaziz JD, Matsushita T,Magro CM, St Clair EW, Tedder TF. B-lymphocyte contributions to human autoimmune disease. Immunol Rev 2008; 223: 284. [DOI] [PubMed] [Google Scholar]
- 110.Chong AS, Sciammas R.Matchmaking the B-cell signature of tolerance to regulatory B cells. Am J Transplant 2011; 11: 2555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Mizoguchi A, Bhan AK. A case for regulatory B cells. J Immunol 2006; 176: 705. [DOI] [PubMed] [Google Scholar]
- 112.Lund FE, Garvy BA, Randall TD,Harris DP. Regulatory roles for cytokine-producing B cells in infection and autoimmune disease. Curr Dir Autoimmun 2005; 8: 25. [DOI] [PubMed] [Google Scholar]
- 113.Peng B, Ming Y, Yang C. Regulatory B cells: the cutting edge of immune tolerance in kidney transplantation. Cell Death Dis 2018; 9: 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Yanaba K, Bouaziz JD, Haas KM, Poe JC, Fujimoto M, Tedder TF. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity 2008; 28: 639. [DOI] [PubMed] [Google Scholar]
- 115.Ding Q, Yeung M, Camirand G, et al. Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice. J Clin Invest 2011; 121: 3645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Lykken JM, Candando KM, Tedder TF. Regulatory B10 cell development and function. Int Immunol 2015; 27: 471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.DiLillo DJ, Griffiths R, Seshan SV, et al. B lymphocytes differentially influence acute and chronic allograft rejection in mice. J Immunol 2011; 186: 2643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lal G, Nakayama Y, Sethi A, et al. Interleukin-10 from marginal zone precursor B-cell subset is required for costimulatory blockade-induced transplantation tolerance. Transplantation 2015; 99: 1817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Marino J, Paster JT, Trowell A, et al. B cell depletion with an anti-CD20 antibody enhances alloreactive memory T cell responses after transplantation. Am J Transplant 2016; 16: 672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Starling RC, Armstrong B, Bridges ND, et al. Accelerated allograft vasculopathy with rituximab after cardiac transplantation. J Am Coll Cardiol 2019; 74: 36. [DOI] [PubMed] [Google Scholar]
- 121.Clatworthy MR, Watson CJ, Plotnek G, et al. B-cell-depleting induction therapy and acute cellular rejection. N Engl J Med 2009; 360: 2683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Krejcik J, Casneuf T, Nijhof IS, et al. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood 2016; 128: 384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kwun J, Matignon M, Manook M, et al. Daratumumab in sensitized kidney transplantation: potentials and limitations of experimental and clinical use. J Am Soc Nephrol 2019; 30: 1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Blair PA, Norena LY, Flores-Borja F, et al. CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic lupus erythematosus patients. Immunity 2010; 32: 129. [DOI] [PubMed] [Google Scholar]
- 125.Iwata Y, Matsushita T, Horikawa M, et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood 2011; 117: 530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lino AC, Dang VD, Lampropoulou V, et al. LAG-3 inhibitory receptor expression identifies immunosuppressive natural regulatory plasma cells. Immunity 2018; 49: 120e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Dalai SK, Mirshahidi S, Morrot A, Zavala F, Sadegh-Nasseri S. Anergy in memory CD4+ T cells is induced by B cells. J Immunol 2008; 181: 3221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Sagoo P, Perucha E, Sawitzki B, et al. Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans. J Clin Invest 2010; 120: 1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Pallier A, Hillion S, Danger R, et al. Patients with drug-free long-term graft function display increased numbers of peripheral B cells with a memory and inhibitory phenotype. Kidney Int 2010; 78: 503. [DOI] [PubMed] [Google Scholar]
- 130.Chesneau M, Michel L, Dugast E, et al. Tolerant kidney transplant patients produce B cells with regulatory properties. J Am Soc Nephrol 2015; 26: 2588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Chesneau M, Pallier A, Braza F, et al. Unique B cell differentiation profile in tolerant kidney transplant patients. Am J Transplant 2014; 14: 144. [DOI] [PubMed] [Google Scholar]
- 132.Bigot J, Pilon C, Matignon M, et al. Transcriptomic signature of the CD24(hi) CD38(hi) transitional B cells associated with an immunoregulatory phenotype in renal transplant recipients. Am J Transplant 2016; 16: 3430. [DOI] [PubMed] [Google Scholar]
- 133.Lee KM, Kim JI, Stott R, et al. Anti-CD45RB/anti-TIM-1-induced tolerance requires regulatory B cells. Am J Transplant 2012; 12: 2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Deng S, Moore DJ, Huang X, et al. Cutting edge: transplant tolerance induced by anti-CD45RB requires B lymphocytes. J Immunol 2007; 178: 6028. [DOI] [PubMed] [Google Scholar]
- 135.Newell KA, Asare A, Sanz I, et al. Longitudinal studies of a B cell-derived signature of tolerance in renal transplant recipients. Am J Transplant 2015; 15: 2908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Moreso F, Torres IB, Martinez-Gallo M, et al. Gene expression signature of tolerance and lymphocyte subsets in stable renal transplants: results of a cross-sectional study. Transplant Immunol 2014; 31: 11. [DOI] [PubMed] [Google Scholar]
- 137.Asare A, Kanaparthi S, Lim N, et al. B cell receptor genes associated with tolerance identify a cohort of immunosuppressed patients with improved renal allograft graft function. Am J Transplant 2017; 17: 2627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Baron D, Ramstein G, Chesneau M, et al. A common gene signature across multiple studies relate biomarkers and functional regulation in tolerance to renal allograft. Kidney Int 2015; 87: 984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Vo AA, Peng A, Toyoda M, et al. Use of intravenous immune globulin and rituximab for desensitization of highly HLA-sensitized patients awaiting kidney transplantation. Transplantation 2010; 89: 1095. [DOI] [PubMed] [Google Scholar]
- 140.Loupy A, Suberbielle-Boissel C, Zuber J, et al. Combined posttransplant prophylactic IVIg/anti-CD 20/plasmapheresis in kidney recipients with preformed donor-specific antibodies: a pilot study. Transplantation 2010; 89: 1403. [DOI] [PubMed] [Google Scholar]
- 141.Kobashigawa JA, Patel JK, Kittleson MM, et al. The long-term outcome of treated sensitized patients who undergo heart transplantation. Clin Transplant 2011; 25: E61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Snyder LD, Gray AL, Reynolds JM, et al. Antibody desensitization therapy in highly sensitized lung transplant candidates. Am J Transplant 2014; 14: 849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.van den Hoogen MW, Kamburova EG, Baas MC, et al. Rituximab as induction therapy after renal transplantation: a randomized, double-blind, placebo-controlled study of efficacy and safety. Am J Transplant 2015; 15: 407. [DOI] [PubMed] [Google Scholar]
- 144.Lee KM, Stott RT, Zhao G, et al. TGF-beta-producing regulatory B cells induce regulatory T cells and promote transplantation tolerance. Eur J Immunol 2014; 44: 1728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Vo AA, Lukovsky M, Toyoda M, et al. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N Engl J Med 2008; 359: 242. [DOI] [PubMed] [Google Scholar]
- 146.Martin F, Chan AC. B cell immunobiology in disease: evolving concepts from the clinic. Annu Rev Immunol 2006; 24: 467. [DOI] [PubMed] [Google Scholar]
- 147.Hoyer BF, Manz RA, Radbruch A, Hiepe F. Long-lived plasma cells and their contribution to autoimmunity. Ann NY Acad Sci 2005; 1050: 124. [DOI] [PubMed] [Google Scholar]
- 148.Schrezenmeier E, Budde K, Staeck O, et al. Incidence of infectious disease and malignancies after rituximab therapy in kidney transplant recipients: results from a cohort in Germany. Transplant Proc 2017; 49: 2269. [DOI] [PubMed] [Google Scholar]
- 149.Nixon A, Ogden L, Woywodt A, Dhaygude A. Infectious complications of rituximab therapy in renal disease. Clin Kidney J 2017; 10: 455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Tyden G, Ekberg H, Tufveson G, Mjornstedt L. A randomized, double-blind, placebo-controlled study of single dose rituximab as induction in renal transplantation: a 3-year follow-up. Transplantation 2012; 94: e21. [DOI] [PubMed] [Google Scholar]
- 151.Kyaw T, Tay C, Krishnamurthi S, et al. B1a B lymphocytes are atheroprotective by secreting natural IgM that increases IgM deposits and reduces necrotic cores in atherosclerotic lesions. Circ Res 2011; 109: 830. [DOI] [PubMed] [Google Scholar]
- 152.Barnett AN, Hadjianastassiou VG, Mamode N. Rituximab in renal transplantation. Transplant Int 2013; 26: 563. [DOI] [PubMed] [Google Scholar]
