Abstract
We have attempted here to provide an up-to-date review of the collaboration between helper T cells and B cells in response to protein and glycoprotein antigens. This collaboration is essential as it not only protects from many pathogens but also contributes to a litany of autoimmune and immune-mediated diseases.
OVERVIEW
Sequential collaborative interactions between helper T cells and B cells are a central aspect of adaptive immune responses initiated by protein and glycoprotein antigens. Many functions of B cells are dependent on CD4+ T cells. Similarly, numerous T cell–mediated processes, both physiologic and pathologic, appear to depend on the ability of activated B cells to nurture and maintain CD4+ T cells. Consequently, approaches designed to prevent T-B collaboration are an important part of our therapeutic armamentarium, even for diseases that appear to be driven by CD4+ T cells. A conceptual understanding of T-B collaboration is essential to appreciate how cellular and humoral immunity is developed and maintained and how alterations within 1 cell population propagate throughout the system. Knowledge of the mechanisms of T-B collaboration informs the pathogenesis, prevention, and treatment of numerous immune-mediated human diseases, including pathogen responses, autoimmunity, and transplant rejection.
The immune system evolved in part to mount pathogen responses. Indeed, its importance to human physiology was first appreciated in this setting. Therefore, the initial description of the biology of T-B collaboration in this review will largely be considered from the viewpoint of pathogen responses. We will separately consider the relevance of T-B collaboration in autoimmunity and transplantation.
LINKED RECOGNITION, B-CELL ACTIVATION, AND THE INDUCTION OF T-B COLLABORATION
Antigens and antigen complexes contain 2 categories of physically linked antigenic determinants (epitopes). Protrusions on the surface of an antigen serve as B-cell epitopes that are recognized by the antigen receptor on 1 or more B-cell clones. B-cell antigen receptors directly bind native conformational epitopes on antigens that can be chemically diverse; these antigens include small molecules, nucleic acids, and lipids but are most commonly carbohydrates and proteins.1 In contrast, T-cell antigen receptors on CD4+ T cells recognize peptide epitopes derived from protein and glycoprotein antigens when presented on major histocompatibility complex (MHC) class II molecules.2 To generate MHC class II–restricted T-cell epitopes, antigens are processed into linear stretches of approximately 15 to 25 amino acids via proteolytic degradation in late endosomes or lysosomes. These linear peptides may be capable of binding tightly to the groove of a host MHC class II molecule (Figure 1). The exposed residues of a peptide bound to an MHC class II molecule may be recognized by a T-cell receptor (TCR) on ≥1 T-cell clones.
FIGURE 1.

Distinct epitopes on the same protein are recognized by B and T cells. A, Two conformational epitopes, a “green triangle,” and a “yellow square” represent B-cell epitopes in the protein and a linear “black peptide” represents the T-cell epitope in this protein. B, The BCR specific for a “yellow square” epitope on a B cell binds specifically to and endocytoses the protein antigen. C, In the late endosomal/lysosomal compartment, the endocytosed antigen is processed to peptides and the “black peptide” binds the MHC class II groove and is transported to the cell surface of the B cell. D, An activated CD4+ T cell specific for the black peptide (previously activated by DCs presenting the same black peptide on an identical MHC class II molecule) now recognizes the B cell that has processed and presented the black peptide on its MHC class II molecules. BCR, B-cell receptor; DC, dendritic cell; MHC, major histocompatibility complex.
Importantly, B cells process and present T-cell epitopes contained within the specific antigen recognized by the individual B-cell clone. Given the restricted use of peptide epitopes by T cells, T-B interactions necessarily involve cells that recognize epitopes derived from a shared protein, glycoprotein, or protein complex antigen.3,4 Therefore, T-B collaboration requires the colocalization of a B-cell clone that recognizes a conformational epitope on the surface of an antigen and a T-cell clone that recognizes a different linear peptide derived from the same antigen and bound to an MHC class II molecule on the B-cell clone.
Cognate T-B interactions are required for most antibody isotype switching, antibody affinity maturation, plasma cell development, and memory B-cell formation.5 These processes, and the interactions that drive them, are spatially and temporally restricted. The initial antigenic stimulation of B cells leads to partial activation with limited proliferation and differentiation. Activated B cells endocytose, process, and present antigen. They downregulate C-X-C chemokine receptor type 5 (CXCR5), a chemokine receptor that promotes localization to B-cell follicles, and upregulate C-C chemokine receptor type 7 (CCR7), the ligand for which is enriched in the T-cell zone, allowing migration to the T-B boundary.5 Successful engagement with a CD4+ T cell that recognizes a shared epitope initiates a sequence of events leading to both short-term and long-lasting humoral immunity. A broad overview of T-B collaboration is provided in Figure 2.
FIGURE 2.

An overview of T-B collaboration. A DC that has captured, processed, and presented a specific peptide on a specific MHC class II protein migrates into the T-cell zone in a draining lymph node and selects and activates a specific CD4+ T cell. The selected T cells proliferate, alter the expression of chemokine receptors, and migrate toward the interface between T and B cells. A B-cell clone that recognizes a B-cell epitope on the same protein antigen in the B-cell zone or follicle alters its chemokine receptor expression and migrates toward the T-B interface. At the interface, activated CD4+ T cells recognize B cells that present their specific peptide–MHC class II complexes and the B cells activate, proliferate, undergo class switching, and differentiate into extrafollicular plasmablasts. These activated B cells and plasmablasts in the T-B interface are called extrafollicular foci. Activated B cells may then activate previously activated CD4+ T cells and induce their differentiation into T follicular helper cells, which in turn contribute to the formation of GCs and the selection of higher affinity B cells that will initially form memory B cells and later give rise to long-lived plasma cells. Details in the text. DC, dendritic cell; GC, germinal center; MHC, major histocompatibility complex; Tfh, follicular helper T cell.
T-CELL ACTIVATION AND THE INDUCTION OF T-B COLLABORATION
At sites of inflammation, pathogen-derived molecules, such as lipopolysaccharide, flagellin, or nucleic acids, or molecules released by damaged host cells bind pattern recognition receptors on dendritic cells (DCs), leading to their activation. Activated DCs capture, process, and present protein antigens more efficiently, produce higher levels of MHC and costimulatory molecules, and express CCR7.6
After arriving in the T-cell zone, DCs activate rare naive CD4+ T cells whose TCRs recognize a relevant MHC class II–peptide complex.7 Activated T cells initiate diverse developmental programs and undergo clonal expansion. These primed, but not yet mature, cells begin to alter chemokine expression. Downregulation of CCR7 permits movement away from the T-cell zone. The ultimate destination and function of each cell depends on its specific developmental program. Cells developing into peripheral effector subsets (Th1, Th2, Th17, etc) upregulate sphingosine 1-posphate receptor 1 to permit egress from lymphoid tissue and additional receptors that guide them to peripheral sites.8 Cells developing along the follicular helper pathway upregulate CXCR5 and migrate toward the B-cell zone.9 Functional maturation requires reexposure to antigen at the site of inflammation or at the T-B boundary.
THE FORMATION OF AN EXTRAFOLLICULAR FOCUS
Interactions between developing cognate T and B cells lead to the formation of an extrafollicular B-cell focus. Ligation of CD40 on B cells by CD40L on T cells increases B-cell activation, promotes proliferation, and initiates class switch recombination to most human isotypes. The vast majority of isotype switching occurs at extrafollicular foci.10 Limited somatic hypermutation, and resulting affinity maturation, can occur in extrafollicular foci but does not reach the levels seen in germinal center (GC) B cells, primarily because of a lack of architectural and cellular components required for the iterative process of mutation and selection seen in GCs.11
Extrafollicular foci can occur in isolation or may result in the development of GCs. The progeny of individual B-cell clones can develop into extrafollicular, GC, and memory B cells.12 High-affinity interactions predispose B cells toward extrafollicular responses, whereas lower-affinity B cells are more likely to enter GCs.13,14 Extrafollicular plasmablasts are short lived, whereas GCs produce long-lived plasma cells. Memory B cells arise both outside the GC (extrafollicular) and in the early stages of the GC response.15 Memory B cells thus generally exhibit a reduced degree of somatic hypermutation with an affinity for foreign antigen greater than that of naive B cells but lower than long-lived plasma cells. The diminished affinity maturation seen in memory B cells may allow for broad specificity with few restrictions on the potential direction of subsequent rounds of maturation, permitting rapid, flexible, and effective responses to antigenic variants produced by successive generations of pathogens.
Although follicular helper T (Tfh) cells are responsible for providing help to GC B cells,16,17 the T cells involved in the initial T-B collaboration that drives the vast majority of isotype switching have not been conclusively identified. In humans and mice, CD4+ T cells in secondary lymphoid tissues exhibit a range of expression of Tfh markers, yet only cells with the highest expression of programed cell death protein 1 (PD1), CXCR5, and B-cell lymphoma 6 (BCL6), commonly called GC-Tfh or Tgc, migrate into GCs and provide help to GC B cells.18 Many, and often most, Tfh-like cells express lower levels of PD1 and CXCR5 and are not found within GCs. In humans, these cells are typically negative for BCL6.19–21 They may represent precursors to GC-Tfh and are commonly called pre-Tfh, but their developmental relationship with GC-Tfh has not been formally tested. As such, we prefer to call PD1-low CXCR5-low cells extrafollicular helper T cells. The specific contributions of extrafollicular versus Tfh cells to isotype switching remains unclear. The gaps in our understanding of the phenotypic and functional heterogeneity within Tfh-like cells will be addressed in more detail in the section below on helper T cells.
THE GC RESPONSE
The histological structures described as GCs were discovered by anatomists and pathologists before any notion of the existence of immunity. Walther Flemming, the great German anatomist, is widely recognized for his description in 1882 of the doubling of the chromosomes in the nuclei of cells that occurs as a prelude to cell division, a process he called “mitosis.” Although Flemming is best remembered today for his studies on cytogenetics and mitosis, in 1885 he described structures in lymph nodes that he called “GCs,” and he speculated that these were the sites at which lymphocytes were generated. The function of lymphocytes in adaptive immunity was only established in 1957 by Gowans,22 and although indeed some lymphocytes do divide in GCs, they are not the sites of B or T lymphopoiesis. Rather, they are incubators that foster the evolution of activated B cells into high-affinity class-switched antibody-producing plasma cells.23 In the true Darwinian sense, this process involves genetic diversification followed by natural selection and the eventual survival of only the fittest clones.
At its peak, the GC can be divided into a dark zone, in which B cells proliferate and undergo somatic hypermutation, and a light zone, in which the selection of high-affinity B cells occurs. It is the combination of somatic hypermutation and the selection of high-affinity B cells that forms the basis for the affinity maturation of antibodies. Repeated rounds of proliferation and mutation in the dark zone, followed by selection in the light zone, allow for the accumulation of mutations in both the heavy and light chain genes. These mutations are found primarily in the regions that bind B-cell epitopes because competition for limited antigen imparts a selective advantage on cells with the highest affinity.
The initiation and maintenance of GC reactions requires the reciprocal induction of the transcription factor BCL6, which functions as a repressor-of-repressors in both GC B cells and cognate Tfh cells.24 Expression of an enzyme called activation-induced cytidine deaminase (AID) in B cells initiates the process of somatic hypermutation, which largely targets the rearranged variable region (VDJ or VJ) exons of Ig genes.25 Most somatic hypermutation occurs while B cells are dividing in the dark zone. There are 3 possible consequences of ongoing somatic hypermutation: mutations may alter the affinity for foreign antigen, they may alter affinity to self-antigen, or they may induce an overwhelming degree of DNA damage leading to apoptosis.
After multiple divisions, surviving dark zone B cells downregulate the expression of the CXCR4 chemokine receptor that originally restricted them to the dark zone, upregulate CXCR5, and drift into the light zone of the GC.26 In the light zone, B cells encounter follicular DCs that exhibit antigen in immune complexes on Fc and complement receptors.1 They also encounter Tfh cells, which only populate the light zone. B cells with the highest affinity to a foreign antigen most efficiently capture it, enhancing their ability to present linear peptides on MHC molecules and increasing the probability that they form a stable interaction with a cognate Tfh cell. This interaction depends on ligation of CD40 on B cells by CD40L on T cells and is required for survival.27 Importantly, many pathogens produce epitopes with substantial overlap to human self-antigens, a phenomenon known as molecular mimicry, and mutations that reduce affinity to self-antigen potentiate T-cell collaboration and optimal affinity maturation trajectories.28
Positively selected B cells that receive additional CD40 signals induce c-Myc, reexpress CXCR4, and return to the dark zone to proliferate and undergo additional somatic hypermutation.29 Repeated rounds of genetic diversification and proliferation in the dark zone followed by competition for antigen and T cell help in the light zone to create a selective pressure that increases the relative frequency of cells with higher-affinity B-cell receptors. Thus, the fitness of a GC B cell is a function of its affinity for antigen, and “survival of the fittest” is the engine that drives affinity maturation.
What exactly induces the earlier emergence of memory B cells and the later emergence of the precursors of long-lived plasma cells is not known. Regardless, while memory cells exhibit only modest increases in affinity, additional rounds of selection generate plasmablasts with high-affinity B-cell receptors. These cells migrate to the bone marrow, where they differentiate further into long-lived plasma cells that secrete high-affinity antibodies for many years, often for decades.
T CELLS THAT HELP B CELLS: TFH, PRE-TFH, GC-TFH, AND PERIPHERAL HELPER T CELLS
Naive CD4+ T cells are pluripotent precursors that differentiate into phenotypically and functionally distinct subsets uniquely tailored to specific inflammatory contexts. The diversity of potential outcomes can be conceptually organized along 2 functional dimensions. With few exceptions, CD4+ T cells either promote or suppress inflammation and modulate either cellular or humoral responses. Effector T cells, including Th1, Th2, and Th17 cells, promote cellular responses at the site of inflammation. Regulatory T cells (Treg) suppress them. Tfh-like cells promote humoral responses in secondary (and possibly tertiary) lymphoid tissues, whereas follicular Treg (Tfr) cells oppose them. The heterogeneity seen among effector T cells is mirrored in Treg, Tfh, and Tfr cells, with subsets of each displaying phenotypic and functional overlap with classical effector lineages.30
Differentiation begins after initial activation by professional antigen-presenting cells (APCs) in secondary lymphoid tissues. The quantity and quality of TCR, costimulatory, and cytokine signals impart specific developmental trajectories.31 Potent TCR activation and CD28 costimulation favor Tfh development.32,33 The activation marker PD1 is elevated on Tfh cells, likely due in part to the strength of stimulation required to promote their differentiation.34 Sequential antigen presentation and ligation of inducible T-cell costimulatory (ICOS) first by DCs and then by cognate B cells is required for sustained expression of CXCR5 and BCL6 and maturation into Tfh.35–38 The cytokines interleukin (IL)-6 and IL-21 also promote Tfh development, whereas additional cytokines are thought to impart functional heterogeneity.39
Cognate B cells are primarily located within secondary lymphoid tissues, but T-B collaboration can occur at peripheral sites of inflammation, leading to the generation of peripheral Tfh-like cells and formation of tertiary (ectopic) lymphoid structures (ELSs).40–42 ELSs provide ongoing support for the production of class-switched antibodies in multiple inflammatory contexts, including renal transplant rejection.43 They can contain mature PD1+ CXCR5+ BCL6+ Tfh cells and fully formed GCs or may exhibit limited spatial organization characterized by extrafollicular B-cell responses and the presence of PD1+ CXCR5− BCL6− CD4+ Tfh-like cells known as peripheral helper T (Tph) cells.44,45
There is substantial heterogeneity within Tfh-like cells.46 They can adopt partial phenotypes associated with effector subsets, and the frequency of these cells correlates with the abundance of their effector equivalent. This overlapping heterogeneity allows for coordinated and selective modulation of distinct aspects of cellular and humoral responses. Cytokines produced by CD4+ T cells promote cellular responses optimized for different pathogen types and also regulate humoral responses.47 For example, IL-4 and IL-21 are important for the formation of GCs and the induction of affinity maturation. IL-4 and IL-13 promote, whereas IL-21 opposes, switching to IgE and TGF-β promotes switching to IgA.48–52 However, the precise relationships between different cytokines and antibody isotypes remain poorly understood.
Examination of T-B conjugates in mice suggested GC-Tfh cells are the source of cytokines that guide affinity maturation and isotype switching.48 However, more recent data indicated that the vast majority of isotype switching occurs in pre-GC or extrafollicular B cells after initial cognate interaction with T cells.10 As previously mentioned, many CD4+ T cells in the secondary lymphoid tissues and ELS of humans express reduced levels of PD1 and CXCR5 and are found outside of B-cell follicles.53 These cells may represent developmental precursors of Tfh and central memory cells or they may be comprise cells that failed to obtain sufficient B-cell help, leading to phenotypic decay.44,54,55 Alternatively, they may represent an overlapping but independent and functionally mature lineage that drives class switching in extrafollicular and pre-GC B cells.
In mice, PD1-low, CXCR5-low, CD4+ T cells phenotypically similar to human extrafollicular helpers can be found at the T-B border. These cells express BCL6 and are required for extrafollicular B-cell class switching to IgG1 and IgG2c in some models.19 This suggests that extrafollicular helper T cells may collaborate with B cells to drive class switching. In support of this, signaling lymphocytic activation molecule-associated protein (SAP) is required for GC formation and long-term humoral responses but appears dispensable for class-switched extrafollicular responses.56,57 In humans, PD1-low, CXCR5-low, BCL6+ tonsil CD4+ cells can induce proliferation and differentiation of naive B cells into class-switched antibody-producing cells in vitro but are unable to provide help to GC B cells.58
However, most human extrafollicular helper T cells do not express BCL6.20,21 Interestingly, extrafollicular class switching events may not necessarily require BCL6. Although deletion of BCL6 in murine CD4+ T cells leads to complete loss of Tfh cells and GCs, its effect on class switching is less pronounced, and the magnitude of this effect varies by isotype and by inflammatory context. The dependence of IgG1 and IgE appears to be absolute in rodents, but deletion of BCL6 leads to inconsistent, modest, or absent reductions in T cell–dependent isotype switching to IgG2b, IgG2c, IgG3, and IgA.16,56,57,59–64 There is evidence that BCL6-negative T cells can drive class switching in humans. CXCR5+ BCL6− extrafollicular CD4+ T cells can provide help to B cells in vitro.53 Tph cells have been observed adjacent to B cells and are capable of driving IgG plasma cell differentiation in vitro.44 Patients with severe coronavirus disease 2019 (COVID-19) exhibited abundant class-switched antibodies despite losing BCL6+ Tfh cells.20,65,66 Interestingly, IgG1 was the most abundant class-switched isotype in these patients, and IgG1 antibodies harbored the fewest number of somatic mutations, indicating that class switching to IgG1 may not require BCL6 in humans.
As with B cells, the progeny of individual naive CD4+ T-cell clones can develop into multiple lineages, and the affinity of a TCR biases these fate outcomes.32,33 High affinity for antigen predisposes cells to develop into Tfh versus effector T cells. The role of affinity in driving extrafollicular helper differentiation remains unexplored. Does increased affinity promote extrafollicular versus follicular responses, as in B cells, or is the established relationship between affinity and Tfh development paramount? The former is supported by observations that cognate interactions are long-lasting at the T-B border and shorter-lived in the follicle and GC.67 Additional studies are needed to identify the cells responsible for driving extrafollicular help to B cells, both central and peripheral, their developmental relationship with other cell lineages, the role of specific antigenic determinants in guiding their differentiation, and their contribution to health and disease. Immunophenotypes T helper cell subsets of relevance are listed in Table 1.
TABLE 1.
T-cell subsets important in T-B collaboration
| T-cell subsets | Key markers | Location | Key cytokines |
|---|---|---|---|
|
| |||
| Pre-Tfh | CD4+ ICOS+ PD1 + CXCR5+ | SLO | ? |
| GC-Tfh | CD4+ ICOS+ PD1+ CXCR5++ BCL6+ | SLO, GC | IL-4, IL-21, CXCL13 |
| Tph | CD4+ICOS+ PD1+ CXCR5− BCL6− | Inflamed tissue | CXCL13 |
| cTfh | CD4+ ICOS+/− PD1+/− CXCR5−/− BCL6− | Inflamed tissue | IL-21 |
| Tfr | CD4+ICOS+ PD1+ CXCR5+ BCL6+ Foxp3+ | SLO | IL-10, TGF-β |
cTfh, circulating Tfh cell; GC, germinal center; ICOS, inducible T cell costimulatory; IL, interleukin; PD1, programed cell death protein 1; SLO, secondary lymphoid organ; Tfh, follicular helper T cell; Tfr, follicular regulatory T cell; Tph, peripheral helper T cell.
THE REGULATION OF GC RESPONSES: ANTIGEN CLEARANCE, TREG, AND TFR
Tolerance must be maintained during GC reactions. Additionally, eventually, GC reactions must recede and, ultimately, dissolve. These processes are mediated in part by the suppressive effects of Tfr cells.68,69 Self-reactive natural Treg cells (nTreg) are generated in the thymus from developing T-cell precursors and maintain tolerance to self-antigen. Peripheral Treg (pTreg) are generated in secondary lymphoid tissues from naive precursors in response to both self and foreign epitopes but function primarily to maintain tolerance to foreign antigens of the commensal microbiota.70 Tfr exhibit phenotypic and functional overlap with Treg and Tfh cells and exert suppressive effects on GC reactions. They are predominantly derived from thymic natural Treg (nTreg) and can also develop from naive precursors, possibly via transdifferentiation of pTreg or Tfh cells.71–73
Regulatory populations expand in response to inflammation. nTreg prevent loss of T-cell tolerance to self-antigen. Tfr cells derived from nTreg constrain GC reactions, preventing the development of autoreactive GC B cells and the production of autoantibodies. In contrast, Tfr derived from naive precursors, whether pTreg or Tfh, appear to oppose GC reactions to foreign antigens.71–73 Interestingly, this opposition is required for optimal antibody affinity. As the supply of antigen diminishes, and the positive feedback loops driving inflammation are reduced, the suppressive effects of Tfr cells come to predominate, leading to resolution of GCs.
THE LOSS OF GCS IN SEVERE INTRACELLULAR INFECTIONS
Disruption of the delicate interplay of T and B cells can have devastating consequences. For example, severe COVID-19 infections appear linked to a failure in TFH cells and GC development.20,65 Although thoracic lymph node from age-matched controls contained abundant GCs, we observed a dramatic loss of BCL6+ TFH cells and GC B cells in patients with severe COVID-19.20 However, T-B conjugates and AID+ B cells were frequent and consistent with extrafollicular T-B collaboration. In support of this, vigorous extrafollicular B-cell responses have been observed in patients with severe COVID-19.68
This phenomenon has been observed in other severe intracellular infections and may be related to dysregulated production of type 1 inflammatory cytokines. Loss of GCs was reported in severe acute respiratory syndrome.74 GCs failed to develop in response to the murine rickettsia Ehrlichia muris. This phenomenon was reversed by tumor necrosis factor (TNF)-α blockade and by deletion of the gene encoding TNF-α.75 The loss of TFH cells and GCs observed in a murine malaria model was reversed by blockade of TNF-α or interferon (IFN)-β. Deletion of TBX21, which encodes the transcription factor T-box expression in T cells and is required for the differentiation of IFN-γ producing TH1 cells, also prevented GC loss.76 In a Salmonella infection model, IL-12 was responsible for both a block in Tfh cell differentiation and a loss of GCs.77 IL-12 contributes to TH1 cell differentiation. Both IL-12 and TH1 cells increase production of TNF-α in the lymph node. Finally, mice immunized with lymphocytic choriomeningitis peptide and then subsequently infected with lymphocytic choriomeningitis clone 13 developed a severe viral infection involving the lungs and other organs, pronounced lymphopenia, and the loss of GCs in lymph nodes, a syndrome eerily similar to that seen in severe COVID-19. Very high levels of circulating IL-12 were documented in these mice.78
Interestingly, examination of thoracic lymph nodes from patients with severe COVID-19 revealed high levels of TNF-α and increased numbers of TH1 cells.20 Based on these findings, we suspect that high levels of TNF-α and IL-12 may promote TH1 differentiation and block TFH development, leading to a loss of GCs. Thus, the sequential induction of IL-12, IFN-γ and TNF-α could be linked to the phenotype seen in severe COVID-19, a phenomenon that may be relevant to other human diseases, and murine infectious models may afford an opportunity to understand the mechanisms underlying it.
T-B COLLABORATION IN AUTOIMMUNE AND CHRONIC INFLAMMATORY DISEASES
Most common autoimmune diseases are complex multigene disorders. Some genetic variants linked to autoimmunity result in a breakdown in B- or T-cell tolerance. Others enhance activation of adaptive or innate immunity or alter cellular processes that indirectly impinge on immune function. Environmental challenges can exacerbate underlying genetic susceptibilities by inducing autoreactive responses because of molecular mimicry.
Very few autoimmune diseases are entirely dependent on only T cells or only B cells. In many diseases, including systemic lupus erythematosus, pemphigus vulgaris, and myasthenia gravis to name but a few, autoantibodies are pathophysiologically relevant (as opposed to representing an epiphenomenon within a disease). Almost all disease-causing autoantibodies are IgG antibodies.79 Therefore, T-B collaboration is of obvious relevance and is known to occur at both central and peripheral sites.80
In lupus, extrafollicular IgD− CD27− CD11c+ CXCR5− DN2 B cells have been suggested to be the key precursors of autoantibody-secreting cells.81 As a result of this finding and other studies in animal models, it is thought that many autoantibodies are generated at extrafollicular sites.82 A widely held and related view, not substantiated by very strong evidence to date, posits that this may be because it is easier to break tolerance at extrafollicular sites than in GCs. It may be theoretically argued that although Treg function at both follicular and extrafollicular sites, it is only in the GC that successive rounds of selection iteratively reduce affinity to self-antigen and increase affinity to foreign antigen. In contrast, somatic hypermutation in extrafollicular sites is not accompanied by selection and thus may produce a greater propensity for autoreactivity.
The role of T-B collaboration in autoimmunity extends beyond autoantibody production. Depletion of B cells using monoclonal antibodies to CD20 has been shown to be therapeutically beneficial in a number of autoimmune and inflammatory disease contexts, although these diseases are often considered to be largely T-cell driven.83–85 Activated B cells may play an important role in presenting antigen to CD4+ T cells, or provide these T cells with essential cytokines or costimulation, in these disorders. In support of this view, it has been argued on the basis of studies in rodents that in some circumstances, CD4+ T-cell memory may be sustained by B cells.86 Other possible mechanisms need to be considered. For instance, B-cell depletion is a very effective therapy in multiple sclerosis. Although there may be many possible contributions of B cells to the pathogenesis of multiple sclerosis, recent evidence shows that the induction of T cell–dependent B-cell responses to an antigen from Epstein-Barr virus may induce cross-reactive B-cell responses that drive the disease process.87,88
B CELLS AND ANTI-HLA ANTIBODIES IN TRANSPLANT REJECTION
Solid organ transplant serves as a lifesaving therapy for end-stage organ dysfunction. However, long-term allograft survival is limited despite potent, broad-based immune suppression. Chronic antibody-mediated rejection (AMR) is a major barrier to improving long-term graft survival. Donor-specific anti-HLA antibodies (DSAs) are one of the diagnostic criteria of AMR and play a key role in its pathophysiology.89 Seminal studies by Paul Terasaki’s group established the importance of anti-HLA antibodies in AMR.90 It is thought that DSAs bind to antigen, primarily HLA molecules, on endothelial cells in glomerular and peritubular capillaries, leading to complement activation and endothelial injury.91 Studies show that 8% to 10% of kidney transplant recipients develop de novo DSA within 1 y of transplant.92 Acquisition of anti-HLA antibodies within 1 y posttransplant is associated with reduced allograft survival.93 Inhibition of DSA production prevents graft rejection in nonhuman primate kidney transplant models.94,95
Alloreactive B cells can be derived from naive and memory B cells. Historically, IgM DSAs were considered harmless but may be associated with worse allograft survival.96 Nevertheless, most clinically relevant alloreactive antibodies are IgG. Their pathogenicity varies by isotype; data suggest that complement-fixing IgG subclasses are associated with acute AMR, whereas responses dominated by IgG4, which cannot bind FC receptors, are associated with subclinical AMR.97,98 IgG1 is the most common DSA isotype, but most responses are mixed and IgG3, which has the highest affinity for Fc gamma receptor IIIA (FCγRIIIA), correlates best with AMR frequency and severity.99,100
The role of extrafollicular versus GC responses in DSA production and AMR remains unclear. In mice, although deletion of SAP in host CD4+ T cells failed to abrogate early, low-affinity antibody production in a cardiac transplant model, late alloantibody responses were abolished and grafts survived indefinitely.101 Secretion of GC-dependent class-switched, high-affinity antibodies was required for rejection. A separate study found that graft-versus-host allorecognition by tissue-resident donor CD4+ T cells was required to initiate alloimmune humoral responses.102 This process was not dependent on SAP, indicating that induction of DSA may depend on extrafollicular T-B interactions. Maximal DSA levels were achieved in the absence of SAP in host CD4+ T cells, but epitope spreading, vasculopathy, and graft rejection were dependent on germinal reactions. Finally, GC B-cell numbers correlated with DSA levels and kidney allograft rejection.103
Both extrafollicular and GC alloreactive B cells are seen in human transplant recipients. Although DSA levels broadly correlate with AMR, many patients with high DSA titers do not exhibit graft inflammation or vasculopathy.104,105 Notably, epitope spreading appears to correlate with coronary artery vasculopathy in human cardiac transplant recipients.106 Somatic hypermutation is an important driver of epitope spreading and, once tolerance is broken, GCs can recruit additional autoreactive clones.107,108 Furthermore, pretransplant IgG4, which is indicative of a preexisting, advanced GC-dependent humoral response, predicts posttransplant IgG3 production and acute AMR.109 However, extrafollicular responses are a major source of autoreactive antibodies in multiple diseases.82,110,111 Given these findings, it is possible that DSA production and epitope spreading may begin with extrafollicular T-B interactions but that GC dependent, high-affinity antibodies preferentially drive AMR. Differential reliance on extrafollicular versus follicular T-cell help may help explain why DSA alone is insufficient to predict rejection.
Antigen presentation by B cells is critical for DSA and AMR. Studies suggest that it may drive other aspects of allo responses as well. Chronic cardiac allograft vasculopathy was absent in mice lacking B cells but unchanged in mice deficient for antibodies but not B cells.112 Although effector cell numbers were unaffected, IFN-γ-producing memory CD4+ and CD8+ T-cell numbers were dramatically reduced in the absence of B cells in a mouse skin transplant model.113 Interestingly, B cells may be important for more than just antigen presentation and antibody production. In a mouse model of cardiac transplant tolerization, alloantigen-specific B cells were identified in transplant-tolerant recipients that supported TFH development but were unable themselves to become GC B cells.114 These tolerized B cells inhibited DSA production by naive B cells. Thus, B cell–intrinsic properties may impact alloimmune responses in unexpected ways.
TFH, CTFH, AND TFR IN TRANSPLANT REJECTION
T cells are important mediators of both cellular and humoral alloreactions. Loss of CD8+ T cells by anti-CD8 mAb impairs cellular responses and delays, but does not prevent, rejection in a mouse model of heart transplantation.115 Depletion of CD4+ T cells, or costimulatory blockade (eg, CD154 cytotoxic T lymphocyte–associated antigen-4 immunoglobulin fusion protein [CTLA4-Ig]), suppresses both cellular and humoral immunity and can achieve long-term tolerance.116–118 Blockade of CD40 or CD40L prevents DSA production and dramatically extends graft survival in nonhuman primate kidney transplant models, suggesting T-cell help to B cells is a key driver of transplant rejection.94,95
T-cell responses begin with alloantigen presentation by professional APC in secondary lymphoid tissues. Direct and indirect allorecognitions have long been understood to be major pathways of alloantigen presentation. In direct allorecognition, recipient T cells recognize intact donor MHC molecules expressed on the donor cells, whereas in the indirect pathway, recipient T cells recognize donor-derived peptides presented in the context of recipient MHC expressed on the recipient APCs. The semidirect pathway (cross-dressing) was more recently described as an additional and critical mechanism of alloantigen recognition. In the semidirect pathway, intact donor peptide–MHC complex is transferred from donor DCs to recipient DCs and presented to recipient T cells by the recipient DCs. Fadi Lakkis’ group showed that cross-dressed DCs stably engaged TCR-transgenic effector CD8+ T cells in the allograft immediately after transplantation and for up to 8 wk in mouse models of islet and kidney transplantation.119 Mouse models suggest that the indirect pathway is the primary contributor to AMR, although donor-derived T cells may play some role in initiating DSA production via “inverted direct allorecognition.”102,120–122
Regardless of the source of initial antigenic stimulation, development of IgG DSA requires T-B collaboration.94,95,101–103,123 In secondary lymphoid tissues, T cells primed by APCs presenting alloantigens proliferate and differentiate. Some cells migrate to the graft and modulate cellular inflammation, whereas others move to the T-B boundary and provide help to B cells. Additionally, T and B cells sometimes form organized tertiary lymphoid structures in graft tissue. The role of these structures in transplant rejection is not fully understood.124 B cells can be found in close proximity to Tfh-like T cells in kidney allograft biopsies not only in patients with mixed cellular rejection and AMR but also in patients with isolated cellular rejection.43,125,126 Intragraft tertiary lymphoid structures were reported to be associated with worse long-term allograft function, but tissue-resident class-switched innate-like B cells appear to bind renal self-antigens and are not enriched for reactivity against alloantigen.127,128 Furthermore, although B cells have been shown to expand and undergo limited somatic hypermutation in situ, the relative abundance of silent mutations in complementarity determining regions and the absence of follicular DCs suggested that these cells were not part of functional GCs.105
Tfh cells are thought to be critical for T-B collaboration during allo responses. Tfh and GCs develop in mouse models of skin, heart, and kidney transplantation. Using Tfh-deleter mice (CD4-cre; CXCR5-IRES-loxp-STOP-loxp-DTR), Sage et al showed that IgG DSA and AMR were virtually eliminated in the absence of CXCR5+ CD4+ T cells in a model of kidney transplantation.129 Graft survival was dramatically enhanced, but cell-mediated allograft responses were increased and ultimately drove rejection, indicating that abrogation of T-B collaboration is not sufficient to prevent disease. These data may suggest that DSA production in kidney transplantation is dependent on Tfh, but CXCR5+ CD4+ T cells also drive extrafollicular responses.19,53,58,130 Although GCs appear to preferentially drive AMR, extrafollicular T-B interactions are sufficient for DSA production and may be required to initiate follicular responses. The precise role of specific subsets of Tfh-like cells, their influence on GC versus extrafollicular B-cell responses, and their role in graft failure remain incompletely defined.
In transplant patients, circulating Tfh (cTfh) cells, defined as CXCR5+ or ICOS+, are studied as a surrogate marker for humoral alloimmune responses. cTfh cells are a distinct population of T cells found in peripheral blood that exhibits functional overlap with Tfh cells; they secrete IL-21 and can induce naive B cells to proliferate and differentiate into antibody-producing cells in vitro.131,132 The proportion of cTfh cells is higher in kidney transplant recipients with AMR and lower in patients who achieve operational tolerance or who are at low risk of rejection.133,134 La Muraglia et al showed that cTfh cells are an early biomarker of humoral alloreactivity and precede development of DSAs after transplantation, using a mouse skin transplant model.135 However, cTfh cells are heterogenous, do not express BCL6, and their ontogeny is poorly understood. The manner and degree to which these cells reflect the nuances of T-B collaboration is unclear.
The role of Tfr cells in transplant is also poorly understood. Tfr-deleter mice (Foxp3-YFP-cre; CXCR5-IRES-loxp-STOP-loxp-DTR) revealed that Tfr modestly inhibited DSA production in response to allogeneic splenocyte challenge but did not alter DSA levels or graft survival in a model of kidney transplant.129 The relevance of Tfr to human transplant biology is unknown.
THERAPEUTIC TARGETING OF T-B INTERACTION
Several therapies have been tested to treat or prevent allo-specific antibody production: anti-CD20 antibodies to deplete B cells (eg, Rituximab), proteasome inhibitors to deplete plasma cells (eg, Bortezomib), plasmapheresis to remove circulating antibodies, enzymatic digestion by IgG endopeptidase, and anti-IL-6 receptor monoclonal antibodies to modulate B-cell differentiation.136–139 However, their efficacy is limited and, given their lack of specificity, they carry substantial risks of infections.
More recent efforts have focused on the ability of T cells to help B cells. Nonhuman primate transplant models indicate that blockade of CD40/CD40L interactions prevents AMR.118,140 Early antibodies targeting CD40L led to thromboembolic complications and were discontinued. Newer agents engineered to avoid platelet activation, and monoclonals targeting CD40, are under investigation. CD28 blockade also leads to a sharp reduction in Tfh in a mouse model of skin transplantation, and the Belatacept Evaluation of Nephroprotection and Efficacy as First-line Immunosuppression Trial and Belatacept Evaluation of Nephroprotection and Efficacy as First-line Immunosuppression Trial-EXTended criteria donors trials found that fewer patients receiving belatacept, a CTLA4-Ig that interferes with CD28 costimulation, developed DSAs compared with those receiving cyclosporine.141–143 Similarly, although proteasome inhibitor monotherapy frequently leads to DSA rebound, combined pretransplant costimulation blockade and proteasome inhibition reduced Tfh numbers and DSA levels, delayed DSA rebound, and prolonged graft survival in nonhuman primate–sensitized kidney and skin transplant models and in highly sensitized human cardiac transplant candidates.144–146 Combined costimulation blockade and proteasome inhibition were also able to reverse acute AMR in a sensitized mouse skin transplant model and in human kidney transplant recipients, and prolonged costimulation blockade prevented DSA rebound.147 These data indicate that T-B collaboration is required for the initiation, maintenance, and rebound of DSA-mediated AMR. The therapeutics targeting T-B collaboration are summarized in Table 2. However, CD28 costimulation is required for all T-cell inflammatory responses, and prospective cohort studies have consistently shown that belatacept and antimetabolite (eg, mycophenolic mofetil and azathioprine) therapy is associated with impaired seroconversion after COVID-19 vaccination.148–150 Caution is warranted when wielding such blunt tools.
TABLE 2.
Therapies targeting T-B collaboration
| Categories | Target | Reagent | Target | Clinical trial and indications | Target disease | NCT number |
|---|---|---|---|---|---|---|
|
| ||||||
| Costimulatory blockade | CD28 | FR104/VEL-101 | Pegylated anti-CD28 Fab’ fragment | Phase I/II; phase I | Solid organ transplant | NCT05238493; NCT04837092 |
| Lulizumab | Pegylated anti-CD28 specific domain antibody | Phase IIa | Solid organ transplant, SLE | NCT04903054; NCT04066114; NCT-2265744 | ||
| CD80/86 | Abatacept | CTLA4-Ig | FDA approved for rheumatoid arthritis and prophylaxis of acute GVHD | Rheumatoid arthritis, acute GVHD | ||
| Belatacept | CTLA4-Ig | FDA approved for kidney transplant patients | Solid organ transplant | |||
| CD40L | Ruplizumab (hu5C8)a | Anti-CD40L mAb | Discontinued because of thromboembolic events | Solid organ transplant | ||
| Toralizumab (IDEC-131)a | Anti-CD40L mAb | Discontinued because of thromboembolic events | Solid organ transplant | |||
| Toralizumab (IDEC-131)a | Anti-CD40L mAb | Discontinued because of thromboembolic events | Solid organ transplant | |||
| Dapirolizumab | Anti-CD40L mAb | Phase I | SLE | NCT01764594 | ||
| CD40 | Iscalimab | Anti-CD40 mAb | Halted because of nonsuperiority compared with CNI | Solid organ transplant | NCT03663335 | |
| Bleselumab | Anti-CD40 mAb | Phase I | Solid organ transplant | NCT01780844; NCT01279538 | ||
| Syk tylosine kinase | Fostamatinib | Tylosine kinase inhibitor | Phase II | Solid organ transplant | NCT03991780 | |
| BAFF | Atacicept | TACI-Ig | Phase II | SLE | NCT01972568 | |
| Plasma cell–targeted therapy | Proteasome | Bortezomib | Proteasome inhibitor | FDA approved for multiple myeloma | Multiple myeloma | |
| Carfilzomib | Proteasome inhibitor | FDA approved for multiple myeloma | Multiple myeloma | |||
| CD38 | Daratumumab | Anti-CD38 mAb | FDA approved for multiple myeloma | Multiple myeloma, solid organ transplant | ||
| Felzartamab | Anti-CD38 mAb | Phase III | Antibody-mediated rejection | NCT05021484 | ||
| BCMA; CD3 | REGN5458/9 | Anti-BCMA+ anti-CD3 bispecific mAb | Phase I/II | Multiple myeloma | NCT05137054 | |
| CD19 | Inebilizumab | Humanized anti-CD19 mAb | Phase III | Neuromyelitis optic spectrum disorder | NCT02200770 | |
| CD20 | Rituximab | Chimeric anti-CD20 mAb | FDA approved for non-Hodgkin’s lymphoma, CLL, rheumatoid arthritis, granulomatous with polyangiitis, microscopic polyangiitis, pemphigus vulgaris | |||
| Ofatumumab | Humanized anti-CD20 mAb (IgG1k) | FDA approved for CLL, multiple sclerosis | ||||
| Obinutuzumab | Humanized anti-CD20 mAb (defucosylated, IgG1) | FDA approved for CLL | ||||
| Cytokine | IL-2 | Aldesleukin | IL-2 (low dose) | Phase I/II | HCV vasculitis, GVHD, Type 1 diabetes, SLE, Solid organ transplant | NCT00574652; NCT00529035; NCT00525889; NCT04077684; NCT02417870 |
| Efavaleukin alfa | Mutant IL-2 Fc | Phase I/II | GVHD, SLE, Rheumatoid arthritis | NCT03422627; NCT03451422; NCT03410056 | ||
| IL-21 | NNC0114-0006 | Anti-IL-21 mAb | Phase II | Type 1 diabetes, Rheumatoid arthritis, Crohn’s disease | NCT02443155; NCT01208506 | |
| BOS161721 | Anti-IL-21 mAb | Phase I/II | SLE | NCT03371251 | ||
| ATR-107 | Anti-IL-21R mAb | Phase I | PK/PD study in healthy subjects | NCT01162889 | ||
| IL-6 | Clazakizumab | Anti-IL-6 mAb (humanized IgG1) | Phase III | Solid organ transplant | NCT03744910 | |
| Tocilizumab | Anti-IL-6Ra mAb | Phase III | Solid organ transplant | NCT04561986 | ||
| IL-7 | RN168 | Anti-IL-7Ra mAb | Phase I | Type 1 diabetes | NCT02038764 | |
| OSE-127 | Anti-IL-7Ra mAb | Phase II | Ulcerative colitis | NCT04882007 | ||
Discontinued because of side-effect profile.
CLL, chronic lymphocytic leukemia; CNI, calcineurin inhibitor; CTLA4, cytotoxic T lymphocyte–associated protein 4; FDA, Food and Drug Administration; GVHD, graft-versus-host disease; HCV, hepatitis C virus; IL, interleukin; mAb, monoclonal antibody; PK/PD, pharmacokinetics/ pharmacodynamics; SLE, systemic lupus erythematosus; TACI, transmembrane activator and CAML interactor.
CONCLUDING REMARKS
T-B collaboration initiates a complicated network of sequential interactions, both direct and indirect, that positively reinforce alloimmune antibody responses. Disruption or modulation of this collaboration may aid efforts to treat or prevent AMR. Selective manipulation of isotype switching could improve outcomes, but the mechanisms driving isotype rearrangements are poorly understood and given their roles in other aspects of immune responses, efforts targeting the relevant cytokines may lack specificity. The ability to prevent allo-specific antibody production without disrupting normal humoral response would revolutionize organ transplantation. However, a more sophisticated understanding of humoral responses to alloantigen is needed before this goal can be achieved. The relative contribution of GC versus extrafollicular antibody production to AMR is ill-defined and may be context dependent. Furthermore, antigen-specific interventions are required to prevent alloantibody production without compromising beneficial T-B collaboration or to augment desired T-B collaboration without driving rejection. B-cell allo-epitopes are well described, but little is known about the T-cell epitope landscape. What types of mismatches generate T-cell epitopes? Which ones are responsible for AMR? What types of T-B collaboration do they lead to? T-cell epitopes are difficult to identify, but linked recognition may offer a roadmap forward.
Acknowledgments
N.M. is supported by K08DK120868. S.P. was supported by National Institutes of Health grant U19 AI110495.
Footnotes
S.P. is on the Scientific Advisory Boards of Abpro, BeBiopharma, Paratus Sciences, and Octagon Therapeutics, but these positions are unrelated to the topic of this review. The other authors declare no conflicts of interest.
REFERENCES
- 1.Heesters BA, van der Poel CE, Das A, et al. Antigen Presentation to B Cells. Trends Immunol. 2016;37:844–854. [DOI] [PubMed] [Google Scholar]
- 2.Davis MM, Bjorkman PJ. T-cell antigen receptor genes and T-cell recognition [published correction appears in Nature 1988 Oct 20;335(6192):744]. Nature. 1988;334:395–402. [DOI] [PubMed] [Google Scholar]
- 3.Klinman NR. The secondary immune response to a hapten in vitro. Antigen concentration and the carrier effect. J Exp Med. 1971;133:963–972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Abbas AK, Haber S, Rock KL. Antigen presentation by hapten-specific B lymphocytes. IISpecificity and properties of antigen-presenting B lymphocytes, and function of immunoglobulin receptors. J Immunol. 1985;135:1661–1667. [PubMed] [Google Scholar]
- 5.Cyster JG, Allen CDC. B Cell Responses: Cell Interaction Dynamics and Decisions. Cell. 2019;177:524–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Reis e Sousa C Activation of dendritic cells: translating innate into adaptive immunity. Curr Opin Immunol. 2004;16:21–25. [DOI] [PubMed] [Google Scholar]
- 7.Jenkins MK, Khoruts A, Ingulli E, et al. In vivo activation of antigen-specific CD4 T cells. Annu Rev Immunol. 2001;19:23–45. [DOI] [PubMed] [Google Scholar]
- 8.Cyster JG. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu Rev Immunol. 2005;23:127–159. [DOI] [PubMed] [Google Scholar]
- 9.Haynes NM, Allen CD, Lesley R, et al. Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation. J Immunol. 2007;179:5099–5108. [DOI] [PubMed] [Google Scholar]
- 10.Roco JA, Mesin L, Binder SC, et al. Class-Switch Recombination Occurs Infrequently in Germinal Centers. Immunity. 2019;51:337–350.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Elsner RA, Shlomchik MJ. Germinal Center and Extrafollicular B Cell Responses in Vaccination, Immunity, and Autoimmunity. Immunity. 2020;53:1136–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Taylor JJ, Pape KA, Steach HR, et al. Humoral immunity. Apoptosis and antigen affinity limit effector cell differentiation of a single naïve B cell. Science. 2015;347:784–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Paus D, Phan TG, Chan TD, et al. Antigen recognition strength regulates the choice between extrafollicular plasma cell and germinal center B cell differentiation. J Exp Med. 2006;203:1081–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Benson MJ, Erickson LD, Gleeson MW, et al. Affinity of antigen encounter and other early B-cell signals determine B-cell fate. Curr Opin Immunol. 2007;19:275–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Weisel F, Shlomchik M. Memory B Cells of Mice and Humans. Annu Rev Immunol. 2017;35:255–284. [DOI] [PubMed] [Google Scholar]
- 16.Johnston RJ, Poholek AC, DiToro D, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science. 2009;325:1006–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Crotty S T Follicular Helper Cell Biology: A Decade of Discovery and Diseases. Immunity. 2019;50:1132–1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yusuf I, Kageyama R, Monticelli L, et al. Germinal center T follicular helper cell IL-4 production is dependent on signaling lymphocytic activation molecule receptor (CD150). J Immunol. 2010;185:190–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lee SK, Rigby RJ, Zotos D, et al. B cell priming for extrafollicular antibody responses requires Bcl-6 expression by T cells. J Exp Med. 2011;208:1377–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kaneko N, Kuo HH, Boucau J, et al. ; Massachusetts Consortium on Pathogen Readiness Specimen Working Group. Loss of Bcl-6-Expressing T Follicular Helper Cells and Germinal Centers in COVID-19. Cell. 2020;183:143–157.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ioannidou K, Ndiaye DR, Noto A, et al. In Situ Characterization of Follicular Helper CD4 T Cells Using Multiplexed Imaging. Front Immunol. 2021;11:607626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Gowans JL. The effect of the continuous re-infusion of lymph and lymphocytes on the output of lymphocytes from the thoracic duct of unanaesthetized rats. Br J Exp Pathol. 1957;38:67–78. [PMC free article] [PubMed] [Google Scholar]
- 23.Victora GD, Nussenzweig MC. Germinal Centers. Annu Rev Immunol. 2022;40:413–442. [DOI] [PubMed] [Google Scholar]
- 24.Choi J, Diao H, Faliti CE, et al. Bcl-6 is the nexus transcription factor of T follicular helper cells via repressor-of-repressor circuits. Nat Immunol. 2020;21:777–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Muramatsu M, Kinoshita K, Fagarasan S, et al. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell. 2000;102:553–563. [DOI] [PubMed] [Google Scholar]
- 26.Allen CD, Ansel KM, Low C, et al. Germinal center dark and light zone organization is mediated by CXCR4 and CXCR5. Nat Immunol. 2004;5:943–952. [DOI] [PubMed] [Google Scholar]
- 27.Liu YJ, Joshua DE, Williams GT, et al. Mechanism of antigen-driven selection in germinal centres. Nature. 1989;342:929–931. [DOI] [PubMed] [Google Scholar]
- 28.Burnett DL, Langley DB, Schofield P, et al. Germinal center antibody mutation trajectories are determined by rapid self/foreign discrimination. Science. 2018;360:223–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dominguez-Sola D, Victora GD, Ying CY, et al. The proto-oncogene MYC is required for selection in the germinal center and cyclic reentry. Nat Immunol. 2012;13:1083–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.DiToro D, Basu R. Emerging Complexity in CD4+T Lineage Programming and Its Implications in Colorectal Cancer. Front Immunol. 2021;12:694833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations (*). Annu Rev Immunol. 2010;28:445–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tubo NJ, Pagán AJ, Taylor JJ, et al. Single naive CD4+ T cells from a diverse repertoire produce different effector cell types during infection. Cell. 2013;153:785–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.DiToro D, Winstead CJ, Pham D, et al. Differential IL-2 expression defines developmental fates of follicular versus nonfollicular helper T cells. Science. 2018;361:eaao2933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Oestreich KJ, Yoon H, Ahmed R, et al. NFATc1 regulates PD-1 expression upon T cell activation. J Immunol. 2008;181:4832–4839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Akiba H, Takeda K, Kojima Y, et al. The role of ICOS in the CXCR5+ follicular B helper T cell maintenance in vivo. J Immunol. 2005;175:2340–2348. [DOI] [PubMed] [Google Scholar]
- 36.Breitfeld D, Ohl L, Kremmer E, et al. Follicular B helper T cells express CXC chemokine receptor 5, localize to B cell follicles, and support immunoglobulin production. J Exp Med. 2000;192:1545–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Choi YS, Kageyama R, Eto D, et al. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity. 2011;34:932–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Weber JP, Fuhrmann F, Feist RK, et al. ICOS maintains the T follicular helper cell phenotype by down-regulating Krüppel-like factor 2. J Exp Med. 2015;212:217–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Eto D, Lao C, DiToro D, et al. IL-21 and IL-6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS One. 2011;6:e17739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hutloff A T Follicular Helper-Like Cells in Inflamed Non-Lymphoid Tissues. Front Immunol. 2018;9:1707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Pitzalis C, Jones GW, Bombardieri M, et al. Ectopic lymphoid-like structures in infection, cancer and autoimmunity. Nat Rev Immunol. 2014;14:447–462. [DOI] [PubMed] [Google Scholar]
- 42.Gu-Trantien C, Willard-Gallo K. PD-1hiCXCR5−CD4+ TFH Cells Play Defense in Cancer and Offense in Arthritis. Trends Immunol. 2017;38:875–878. [DOI] [PubMed] [Google Scholar]
- 43.Liarski VM, Kaverina N, Chang A, et al. Cell distance mapping identifies functional T follicular helper cells in inflamed human renal tissue. Sci Transl Med. 2014;6:230–ra46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rao DA, Gurish MF, Marshall JL, et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature. 2017;542:110–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Marks KE, Rao DA. T peripheral helper cells in autoimmune diseases. Immunol Rev. 2022;307:191–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Song W, Craft J. T follicular helper cell heterogeneity: Time, space, and function. Immunol Rev. 2019;288:85–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Stavnezer J, Guikema JE, Schrader CE. Mechanism and regulation of class switch recombination. Annu Rev Immunol. 2008;26:261–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Reinhardt RL, Liang HE, Locksley RM. Cytokine-secreting follicular T cells shape the antibody repertoire. Nat Immunol. 2009;10:385–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bryant VL, Ma CS, Avery DT, et al. Cytokine-mediated regulation of human B cell differentiation into Ig-secreting cells: predominant role of IL-21 produced by CXCR5+ T follicular helper cells. J Immunol. 2007;179:8180–8190. [DOI] [PubMed] [Google Scholar]
- 50.Bossie A, Brooks KH, Krammer PH, et al. Activation of murine B cells from different tissues with different mitogens. IIIsotype distribution of secreted immunoglobulins in the presence and absence of IL-4-containing T cell supernatants. J Mol Cell Immunol. 1987;3:221–226. [PubMed] [Google Scholar]
- 51.Yang Z, Wu CM, Targ S, et al. IL-21 is a broad negative regulator of IgE class switch recombination in mouse and human B cells. J Exp Med. 2020;217:e20190472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gowthaman U, Chen JS, Zhang B, et al. Identification of a T follicular helper cell subset that drives anaphylactic IgE. Science. 2019;365:eaaw6433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kim ST, Choi JY, Lainez B, et al. Human Extrafollicular CD4+ Th Cells Help Memory B Cells Produce Igs. J Immunol. 2018;201:1359–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Choi YS, Yang JA, Yusuf I, et al. Bcl6 expressing follicular helper CD4 T cells are fate committed early and have the capacity to form memory. J Immunol. 2013;190:4014–4026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Pepper M, Pagán AJ, Igyártó BZ, et al. Opposing signals from the Bcl6 transcription factor and the interleukin-2 receptor generate T helper 1 central and effector memory cells. Immunity. 2011;35:583–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Crotty S, Kersh EN, Cannons J, et al. SAP is required for generating long-term humoral immunity. Nature. 2003;421:282–287. [DOI] [PubMed] [Google Scholar]
- 57.Morra M, Barrington RA, Abadia-Molina AC, et al. Defective B cell responses in the absence of SH2D1A. Proc Natl Acad Sci U S A. 2005;102:4819–4823. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bentebibel SE, Schmitt N, Banchereau J, et al. Human tonsil B-cell lymphoma 6 (BCL6)-expressing CD4+ T-cell subset specialized for B-cell help outside germinal centers. Proc Natl Acad Sci U S A. 2011;108:E488–E497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kitayama D, Sakamoto A, Arima M, et al. A role for Bcl6 in sequential class switch recombination to IgE in B cells stimulated with IL-4 and IL-21. Mol Immunol. 2008;45:1337–1345. [DOI] [PubMed] [Google Scholar]
- 60.Kobayashi T, Iijima K, Dent AL, et al. Follicular helper T cells mediate IgE antibody response to airborne allergens. J Allergy Clin Immunol. 2017;139:300–313.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Koh B, Ulrich BJ, Nelson AS, et al. Bcl6 and Blimp1 reciprocally regulate ST2+ Treg-cell development in the context of allergic airway inflammation. J Allergy Clin Immunol. 2020;146:1121–1136.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhang B, Liu E, Gertie JA, et al. Divergent T follicular helper cell requirement for IgA and IgE production to peanut during allergic sensitization. Sci Immunol. 2020;5:eaay2754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chen JS, Chow RD, Song E, et al. High-affinity, neutralizing antibodies to SARS-CoV-2 can be made without T follicular helper cells. Sci Immunol. 2022;7:eabl5652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cannons JL, Yu LJ, Jankovic D, et al. SAP regulates T cell-mediated help for humoral immunity by a mechanism distinct from cytokine regulation. J Exp Med. 2006;203:1551–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Duan YQ, Xia MH, Ren L, et al. Deficiency of Tfh Cells and Germinal Center in Deceased COVID-19 Patients. Curr Med Sci. 2020;40:618–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Woodruff MC, Ramonell RP, Nguyen DC, et al. Extrafollicular B cell responses correlate with neutralizing antibodies and morbidity in COVID-19. Nat Immunol. 2020;21:1506–1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Qi H, Cannons JL, Klauschen F, et al. SAP-controlled T-B cell interactions underlie germinal centre formation. Nature. 2008;455:764–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Linterman MA, Pierson W, Lee SK, et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 2011;17:975–982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sage PT, Sharpe AH. The multifaceted functions of follicular regulatory T cells. Curr Opin Immunol. 2020;67:68–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Sakaguchi S, Yamaguchi T, Nomura T, et al. Regulatory T cells and immune tolerance. Cell. 2008;133:775–787. [DOI] [PubMed] [Google Scholar]
- 71.Aloulou M, Carr EJ, Gador M, et al. Follicular regulatory T cells can be specific for the immunizing antigen and derive from naive T cells. Nat Commun. 2016;7:10579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Fazilleau N, Aloulou M. Several Follicular Regulatory T Cell Subsets With Distinct Phenotype and Function Emerge During Germinal Center Reactions. Front Immunol. 2018;9:1792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Jacobsen JT, Hu W, Castro TB R, et al. Expression of Foxp3 by T follicular helper cells in end-stage germinal centers. Science. 2021;373:eabe5146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gu J, Gong E, Zhang B, et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med. 2005;202:415–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Popescu M, Cabrera-Martinez B, Winslow GM. TNF-α Contributes to Lymphoid Tissue Disorganization and Germinal Center B Cell Suppression during Intracellular Bacterial Infection. J Immunol. 2019;203:2415–2424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ryg-Cornejo V, Ioannidis LJ, Ly A, et al. Severe Malaria Infections Impair Germinal Center Responses by Inhibiting T Follicular Helper Cell Differentiation. Cell Rep. 2016;14:68–81. [DOI] [PubMed] [Google Scholar]
- 77.Elsner RA, Shlomchik MJ. IL-12 Blocks Tfh Cell Differentiation during Salmonella Infection, thereby Contributing to Germinal Center Suppression. Cell Rep. 2019;29:2796–2809.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Penaloza-MacMaster P, Barber DL, Wherry EJ, et al. Vaccine-elicited CD4 T cells induce immunopathology after chronic LCMV infection. Science. 2015;347:278–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Elkon K, Casali P. Nature and functions of autoantibodies. Nat Clin Pract Rheumatol. 2008;4:491–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Crotty S T follicular helper cell differentiation, function, and roles in disease. Immunity. 2014;41:529–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Jenks SA, Cashman KS, Zumaquero E, et al. Distinct Effector B Cells Induced by Unregulated Toll-like Receptor 7 Contribute to Pathogenic Responses in Systemic Lupus Erythematosus [published correction appears in Immunity. 2020 Jan 14;52(1):203]. Immunity. 2018;49:725–739.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Jenks SA, Cashman KS, Woodruff MC, et al. Extrafollicular responses in humans and SLE. Immunol Rev. 2019;288:136–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hauser SL, Waubant E, Arnold DL, et al. ; HERMES Trial Group. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358:676–688. [DOI] [PubMed] [Google Scholar]
- 84.Gelfand JM, Cree BAC, Hauser SL. Ocrelizumab and Other CD20+ B-Cell-Depleting Therapies in Multiple Sclerosis. Neurotherapeutics. 2017;14:835–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pitashny M, Shoenfeld Y. B cell depletion in autoimmune rheumatic diseases. Autoimmun Rev. 2005;4:436–441. [DOI] [PubMed] [Google Scholar]
- 86.Whitmire JK, Asano MS, Kaech SM, et al. Requirement of B cells for generating CD4+ T cell memory. J Immunol. 2009;182:1868–1876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bjornevik K, Cortese M, Healy BC, et al. Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science. 2022;375:296–301. [DOI] [PubMed] [Google Scholar]
- 88.Lanz TV, Brewer RC, Ho PP, et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature. 2022;603:321–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Loupy A, Haas M, Roufosse C, et al. The Banff 2019 Kidney Meeting Report (I): Updates on and clarification of criteria for T cell- and antibody-mediated rejection. Am J Transplant. 2020;20:2318–2331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Terasaki PI, Cai J. Human leukocyte antigen antibodies and chronic rejection: from association to causation. Transplantation. 2008;86:377–383. [DOI] [PubMed] [Google Scholar]
- 91.Loupy A, Lefaucheur C. Antibody-Mediated Rejection of Solid-Organ Allografts. N Engl J Med. 2018;379:1150–1160. [DOI] [PubMed] [Google Scholar]
- 92.Everly MJ, Rebellato LM, Haisch CE, et al. Incidence and impact of de novo donor-specific alloantibody in primary renal allografts. Transplantation. 2013;95:410–417. [DOI] [PubMed] [Google Scholar]
- 93.Lee PC, Zhu L, Terasaki PI, et al. HLA-specific antibodies developed in the first year posttransplant are predictive of chronic rejection and renal graft loss. Transplantation. 2009;88:568–574. [DOI] [PubMed] [Google Scholar]
- 94.Kirk AD, Burkly LC, Batty DS, et al. Treatment with humanized monoclonal antibody against CD154 prevents acute renal allograft rejection in nonhuman primates. Nat Med. 1999;5:686–693. [DOI] [PubMed] [Google Scholar]
- 95.Perrin S, Magill M. The Inhibition of CD40/CD154 Costimulatory Signaling in the Prevention of Renal Transplant Rejection in Nonhuman Primates: A Systematic Review and Meta Analysis. Front Immunol. 2022;13:861471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Babu A, Andreou A, Briggs D, et al. Clinical Relevance of Donor-Specific IgM Antibodies in HLA Incompatible Renal Transplantation: A Retrospective Single-Center Study. Clin Transpl. 2016;32:173–179. [PubMed] [Google Scholar]
- 97.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;27:293–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Valenzuela NM, Hickey MJ, Reed EF. Antibody Subclass Repertoire and Graft Outcome Following Solid Organ Transplantation. Front Immunol. 2016;7:433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bruhns P, Iannascoli B, England P, et al. Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses. Blood. 2009;113:3716–3725. [DOI] [PubMed] [Google Scholar]
- 100.Pernin V, Beyze A, Szwarc I, et al. Distribution of de novo Donor-Specific Antibody Subclasses Quantified by Mass Spectrometry: High IgG3 Proportion Is Associated With Antibody-Mediated Rejection Occurrence and Severity. Front Immunol. 2020;11:919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chhabra M, Alsughayyir J, Qureshi MS, et al. Germinal Center Alloantibody Responses Mediate Progression of Chronic Allograft Injury. Front Immunol. 2019;9:3038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Qureshi MS, Alsughayyir J, Chhabra M, et al. Germinal center humoral autoimmunity independently mediates progression of allograft vasculopathy. J Autoimmun. 2019;98:44–58. [DOI] [PubMed] [Google Scholar]
- 103.Steines L, Poth H, Schuster A, et al. Disruption of Tfh:B Cell Interactions Prevents Antibody-Mediated Rejection in a Kidney Transplant Model in Rats: Impact of Calcineurin Inhibitor Dose. Front Immunol. 2021;12:657894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Louis K, Bailly E, Macedo C, et al. T-bet+CD27+CD21- B cells poised for plasma cell differentiation during antibody-mediated rejection of kidney transplants. JCI Insight. 2021;6:e148881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ferdman J, Porcheray F, Gao B, et al. Expansion and somatic hypermutation of B-cell clones in rejected human kidney grafts. Transplantation. 2014;98:766–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ciubotariu R, Liu Z, Colovai AI, et al. Persistent allopeptide reactivity and epitope spreading in chronic rejection of organ allografts. J Clin Invest. 1998;101:398–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Cornaby C, Gibbons L, Mayhew V, et al. B cell epitope spreading: mechanisms and contribution to autoimmune diseases. Immunol Lett. 2015;163:56–68. [DOI] [PubMed] [Google Scholar]
- 108.Degn SE, van der Poel CE, Firl DJ, et al. Clonal Evolution of Autoreactive Germinal Centers. Cell. 2017;170:913–926.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Khovanova N, Daga S, Shaikhina T, et al. Subclass analysis of donor HLA-specific IgG in antibody-incompatible renal transplantation reveals a significant association of IgG4 with rejection and graft failure. Transpl Int. 2015;28:1405–1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ols ML, Cullen JL, Turqueti-Neves A, et al. Dendritic Cells Regulate Extrafollicular Autoreactive B Cells via T Cells Expressing Fas and Fas Ligand. Immunity. 2016;45:1052–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Deng R, Hurtz C, Song Q, et al. Extrafollicular CD4+ T-B interactions are sufficient for inducing autoimmune-like chronic graft-versus-host disease. Nat Commun. 2017;8:978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Zeng Q, Ng YH, Singh T, et al. B cells mediate chronic allograft rejection independently of antibody production. J Clin Invest. 2014;124:1052–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Ng YH, Oberbarnscheidt MH, Chandramoorthy HC, et al. B cells help alloreactive T cells differentiate into memory T cells. Am J Transplant. 2010;10:1970–1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Khiew SH, Jain D, Chen J, et al. Transplantation tolerance modifies donor-specific B cell fate to suppress de novo alloreactive B cells. J Clin Invest. 2020;130:3453–3466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Campos L, Naji A, Deli BC, et al. Survival of MHC-deficient mouse heterotopic cardiac allografts. Transplantation. 1995;59:187–191. [PubMed] [Google Scholar]
- 116.Han WR, Murray-Segal LJ, Mottram PL. Assessment of peripheral tolerance in anti-CD4 treated C57BL/6 mouse heart transplants recipients. Transpl Immunol. 1999;7:37–44. [DOI] [PubMed] [Google Scholar]
- 117.Mottram PL, Raisanen-Sokolowski A, Glysing-Jensen T, et al. Redefining peripheral tolerance in the BALB/c to CBA mouse cardiac allograft model: vascular and cytokine analysis after transient CD4 T cell depletion. Transplantation. 1998;66:1510–1518. [DOI] [PubMed] [Google Scholar]
- 118.Kim EJ, Kwun J, Gibby AC, et al. Costimulation blockade alters germinal center responses and prevents antibody-mediated rejection. Am J Transplant. 2014;14:59–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Hughes AD, Zhao D, Dai H, et al. Cross-dressed dendritic cells sustain effector T cell responses in islet and kidney allografts. J Clin Invest. 2020;130:287–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lin CM, Gill RG. Direct and indirect allograft recognition: pathways dictating graft rejection mechanisms. Curr Opin Organ Transplant. 2016;21:40–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Conlon TM, Saeb-Parsy K, Cole JL, et al. Germinal center alloantibody responses are mediated exclusively by indirect-pathway CD4 T follicular helper cells. J Immunol. 2012;188:2643–2652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Charmetant X, Chen CC, Hamada S, et al. Inverted direct allorecognition triggers early donor-specific antibody responses after transplantation. Sci Transl Med. 2022;14:eabg1046. [DOI] [PubMed] [Google Scholar]
- 123.Chen CC, Koenig A, Saison C, et al. CD4+ T Cell Help Is Mandatory for Naive and Memory Donor-Specific Antibody Responses: Impact of Therapeutic Immunosuppression. Front Immunol. 2018;9:275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kwun J, Manook M, Page E, et al. Crosstalk Between T and B Cells in the Germinal Center After Transplantation. Transplantation. 2017;101:704–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sarwal M, Chua MS, Kambham N, et al. Molecular heterogeneity in acute renal allograft rejection identified by DNA microarray profiling. N Engl J Med. 2003;349:125–138. [DOI] [PubMed] [Google Scholar]
- 126.de Leur K, Clahsen-van Groningen MC, van den Bosch TPP, et al. Characterization of ectopic lymphoid structures in different types of acute renal allograft rejection. Clin Exp Immunol. 2018;192:224–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Lee YH, Sato Y, Saito M, et al. Advanced Tertiary Lymphoid Tissues in Protocol Biopsies are Associated with Progressive Graft Dysfunction in Kidney Transplant Recipients. J Am Soc Nephrol. 2022;33:186–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Asano Y, Daccache J, Jain D, et al. Innate-like self-reactive B cells infiltrate human renal allografts during transplant rejection. Nat Commun. 2021;12:4372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Mohammed MT, Cai S, Hanson BL, et al. Follicular T cells mediate donor-specific antibody and rejection after solid organ transplantation. Am J Transplant. 2021;21:1893–1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Odegard JM, Marks BR, DiPlacido LD, et al. ICOS-dependent extrafollicular helper T cells elicit IgG production via IL-21 in systemic autoimmunity. J Exp Med. 2008;205:2873–2886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Morita R, Schmitt N, Bentebibel SE, et al. Human blood CXCR5(+) CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion [published correction appears in Immunity. 2011 Jan 28;34(1):135]. Immunity. 2011;34:108–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.de Leur K, Dor FJ, Dieterich M, et al. IL-21 Receptor Antagonist Inhibits Differentiation of B Cells toward Plasmablasts upon Alloantigen Stimulation. Front Immunol. 2017;8:306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Chenouard A, Chesneau M, Bui Nguyen L, et al. Renal Operational Tolerance Is Associated With a Defect of Blood Tfh Cells That Exhibit Impaired B Cell Help. Am J Transplant. 2017;17:1490–1501. [DOI] [PubMed] [Google Scholar]
- 134.Dujardin A, Chesneau M, Dubois F, et al. Clinical and immunological follow-up of very long-term kidney transplant recipients treated with calcineurin inhibitors indicates dual phenotypes. Kidney Int. 2021;99:1418–1429. [DOI] [PubMed] [Google Scholar]
- 135.La Muraglia GM 2nd, Wagener ME, Ford ML, et al. Circulating T follicular helper cells are a biomarker of humoral alloreactivity and predict donor-specific antibody formation after transplantation. Am J Transplant. 2020;20:75–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Moreso F, Crespo M, Ruiz JC, et al. Treatment of chronic antibody mediated rejection with intravenous immunoglobulins and rituximab: A multicenter, prospective, randomized, double-blind clinical trial. Am J Transplant. 2018;18:927–935. [DOI] [PubMed] [Google Scholar]
- 137.Eskandary F, Regele H, Baumann L, et al. A Randomized Trial of Bortezomib in Late Antibody-Mediated Kidney Transplant Rejection. J Am Soc Nephrol. 2018;29:591–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Woodle ES, Tremblay S, Rossi A, et al. Plasma cell targeting to prevent antibody-mediated rejection. Am J Transplant. 2020;20(Suppl 4):33–41. [DOI] [PubMed] [Google Scholar]
- 139.Jordan SC, Lorant T, Choi J, et al. IgG Endopeptidase in Highly Sensitized Patients Undergoing Transplantation [published correction appears in N Engl J Med. 2017 Oct 26;377(17):1700]. N Engl J Med. 2017;377:442–453. [DOI] [PubMed] [Google Scholar]
- 140.Schroder PM, Fitch ZW, Schmitz R, et al. The past, present, and future of costimulation blockade in organ transplantation. Curr Opin Organ Transplant. 2019;24:391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Badell IR, La Muraglia GM 2nd, Liu D, et al. Selective CD28 Blockade Results in Superior Inhibition of Donor-Specific T Follicular Helper Cell and Antibody Responses Relative to CTLA4-Ig. Am J Transplant. 2018;18:89–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Vincenti F, Rostaing L, Grinyo J, et al. Belatacept and Long-Term Outcomes in Kidney Transplantation [published correction appears in N Engl J Med. 2016 Feb 18;374(7):698]. N Engl J Med. 2016;374:333–343. [DOI] [PubMed] [Google Scholar]
- 143.Bray RA, Gebel HM, Townsend R, et al. De novo donor-specific antibodies in belatacept-treated vs cyclosporine-treated kidney-transplant recipients: Post hoc analyses of the randomized phase III BENEFIT and BENEFIT-EXT studies. Am J Transplant. 2018;18:1783–1789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ezekian B, Schroder PM, Mulvihill MS, et al. Pretransplant Desensitization with Costimulation Blockade and Proteasome Inhibitor Reduces DSA and Delays Antibody-Mediated Rejection in Highly Sensitized Nonhuman Primate Kidney Transplant Recipients. J Am Soc Nephrol. 2019;30:2399–2411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Schroder PM, Schmitz R, Fitch ZW, et al. Preoperative carfilzomib and lulizumab based desensitization prolongs graft survival in a sensitized non-human primate model. Kidney Int. 2021;99:161–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Alishetti S, Farr M, Jennings D, et al. Desensitizing highly sensitized heart transplant candidates with the combination of belatacept and proteasome inhibition. Am J Transplant. 2020;20:3620–3630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Jain D, Rajab A, Young JS, et al. Reversing donor-specific antibody responses and antibody-mediated rejection with bortezomib and belatacept in mice and kidney transplant recipients. Am J Transplant. 2020;20:2675–2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Ou MT, Boyarsky BJ, Chiang TPY, et al. Immunogenicity and Reactogenicity After SARS-CoV-2 mRNA Vaccination in Kidney Transplant Recipients Taking Belatacept. Transplantation. 2021;105:2119–2123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Boyarsky BJ, Werbel WA, Avery RK, et al. Immunogenicity of a Single Dose of SARS-CoV-2 Messenger RNA Vaccine in Solid Organ Transplant Recipients. JAMA. 2021;325:1784–1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Boyarsky BJ, Werbel WA, Avery RK, et al. Antibody Response to 2-Dose SARS-CoV-2 mRNA Vaccine Series in Solid Organ Transplant Recipients. JAMA. 2021;325:2204–2206. [DOI] [PMC free article] [PubMed] [Google Scholar]
