Fig 2 - uploaded by Nathan Pennock
Content may be subject to copyright.
T cell differentiation. Top : APC [i.e., dendritic cell (DC)] recognition of a spectrum of pathogens through various patho- gen-associated molecular pattern receptors results in cytokine release from the APC. Along with TCR engagement, milieu cytokines initiate ( top middle ) differentiation to one of a variety T cell subsets programmed by transcription factors to specifically respond to the spectrum of the instigating pathogen [pathogen and T helper (Th) subset color coordinated]. Upon differentiation, T cells themselves produce cytokines, which feed back into the cellular milieu, amplifying and balancing the immune response to promote specific pathogen clearance ( bottom middle ) and host survival. Finally, sustained, ill-timed, or otherwise exaggerated T cell immune responses from any of the T cell subsets results in a range of immunopathol- ogies from autoimmunity to allergy and cancer ( bottom ). IFN, interferon; TGF-  , transforming growth factor-  ; Gz, granzyme.
Source publication
Contexts in source publication
Context 1
... the differential control of a transcription factor called ROR ␥ T (91). As the name suggests, this T cell subset is characterized by its ability to produce the cytokine IL-17. IL-17 potently activates neutrophils and, along with IL-8 and other chemokines generated by Th17s, strongly recruits neutrophils to the site of fungal and bacterial invasion. With their release of potent oxidative chemical species, neutrophils are able to directly kill many bacteria and fungi. The importance of Th17s is highlighted in individuals with genetic alteration resulting in diminished IL-17. These individuals suffer from recurrent, severe bacterial and fungal infections (73). While the inflammatory environment is heavily influenced by the specific nature of the invading pathogen, not all T cell differentiation is pathogenic specific. Some T cell differentiation occurs to support immune functions common to all infection responses. The cytokines IL-21 and IL-27 are generated in response to a variety of pathogens and serve to polarize naïve T cells, via the induction of transcription factor Bcl-6 (54), to specifically home to B cell follicles in SLOs (i.e., the spleen and tonsils). Once there, these Tfh cells express a variety of cytokines and costimulatory molecules to assist in the germinal center reaction of B cells, promoting the robust generation of high-affinity antibodies (9). As a testament to the breadth of Tfh influence, mice genetically deficient in molecules necessary for Tfh differentiation fail to produce germinal centers or high-affinity antibodies, a condition that makes them more susceptible to a broad spectrum of infectious agents (16). Unchecked, persistent, or overexuberant immune responses carry with them the danger of immunopathology. To avert this disaster, the immune system produces Tregs, a regulatory subset of T cells that puts the brakes on a variety of inflammatory processes (51, 75). Unlike other T cell subtypes, Tregs can be produced directly from thymic selection [natural Tregs (nTregs)] as well as differentiated [induced Tregs (iTregs)] under the influence of environmental factors such as TGF-  and retinoic acid (12). In either type of Treg, suppressive activity is mediated by active expression of the transcription factor FOXP3 (22, 33). The hallmark effects of Tregs are the impairment of T cell proliferation and cytokine production from other T cell subsets despite their engagement of their antigen-specific TCR. Tregs use a diverse repertoire of mechanisms to achieve these means, such as production of the suppressive cytokines IL-10 (63), TGF-  , or IL-35. These cytokines appear to be instrumental in reestablishing immune quiescence at the elimination of the invading pathogen as well as maintaining immune tolerance to self. In the genetic absence of FOXP3 , and thus the absence of regulatory T cells, both people and mice exhibit broad multiorgan autoimmunity (4, 10). Given their capacity for cytolytic activity, the other major subset of T cells, CD8 T cells, specialize in the eradication of intracellular pathogens and even cancer. When CD8 T cells recognize their antigens in the presence of IFN- ␣ /  and IL-12 cytokines, they differentiate into cytotoxic T cells. Like Th1 cells, cytotoxic T cells generate robust amounts of IFN- ␥ and TNF- ␣ . In addition, these activated and polarized CD8 T cells generate large amounts of secretory vesicles that, when re- leased in close contact to other cells, directly lyse neighboring cells. This activity is mediated through the perforin and granzyme protein families contained within the vesicles. Since cytotoxic CD8 T cells recognize antigen presented on the more ubiquitously expressed MHC class I molecule, CD8 T cells can interact with virtually every cell in the body. Cells presenting antigen in the form of peptide/MHC class I on their surface are identified by the T cell and directly lysed by interactions of their pathogen peptide/MHC with the TCR of CD8 T cells. TCR engagement directs lytic vesicles to the region of interaction, releasing the molecule into the synapse between the cells, thereby lysing the neighboring cell. In this way, CD8 T cells recognizing tumor-associated antigens can lyse cancer- ous cells upon a competent encounter. Like CD4 Th1 cells, cytotoxic CD8 T cells are programmed through the transcription factor T-bet, but their differentiation is also supported through the transcription factor Eomesodermin (70). It bears repeating that while cytotoxic function seems to be a common feature of CD8 T cells regardless of the cytokine milieu, the inflammatory environment can also influence their cytokine production profile in a similar fashion as CD4 T cells, producing such documented subsets referred to as Tc2s and Tc17s (35). Finally, it is important to note that immunological disease occurs when any of these processes occur in an unyielding or overly robust manner or in the absence of a traditional immu- nological/pathogenic trigger (Fig. 2). Distinct roles have been elucidated for both Th1 and Th17s in multiple autoimmune conditions in humans [type 1 diabetes (47) and multiple sclerosis (48, 58)]. Additionally, robust Th2/Th9 responses have been clearly linked to asthma (52, 55) and allergy (60). Unre- solved Th2 and Th17 inflammation results in tissue fibrosis and loss of functional organ architecture (i.e., pulmonary fibrosis) (73, 89). Cytotoxic and Th1 responses have lethal conse- quences when the natural host response or therapeutic inter- vention goes too far and elicits a massive bolus of inflammatory cytokines (i.e., cytokine storm), sending the host into pyrogenic shock. Highlighting the exquisite balance of the immune system, too much regulatory T cell activity is also detrimental to the host as it impairs host tumor immune surveillance, permitting the persistence of oncogenic cells (57, 95). Thus, appropriate immune homeostasis for the host depends on the coordinated temporal regulation of immune activation and immune suppression. As discussed above, T cells have an amazing capacity to proliferate and adopt functional roles aimed at clearing a host of an infectious agent. Just as remarkably, although less un- derstood, is the drastic decline in the T cell population once the primary response is over and the infection is terminated. What remains afterward is a population of T cells with a “memory” for the pathogen they had just taken part in controlling. These remaining T cells, after the collapse of the primary response, are altered in their functional abilities. Compared with their naïve counterparts, these memory T cells have less stringent requirements for subsequent activation via antigenic and costimulatory receptors, an increased proliferative potential, and a more rapid effector response. In addition, memory cells can traffic through both SLOs and peripheral tissues, giving them access to tissues poorly accessed by naïve (peripheral tissues) or effector (SLO) T cells. Collectively, these functions produce an in situ response to reinfection in a fraction of the time taken by the primary response. A T cell response typically peaks ϳ 7–15 days after initial antigen stimulation. For a productive response, this peak cor- responds roughly to the eradication of the pathogen. Over the next few days, 90 –95% of antigen-specific T cells then die off, leaving behind a pool of memory cells with a range of phenotypes and functionalities. For both CD4 and CD8 T cells, there are two main subclasses of memory cells: central-memory (T CM ) and effector-memory (T EM ) T cells. T CM cells are commonly defined phenotypically as expressing high levels of the IL-7 receptor (CD127), high levels of adhesion markers like CD44 and CD62L, low levels of the surface marker killer cell lectin-like receptor subfamily G member 1 (KLRG-1), and high levels of the chemokine/homing receptor C-C chemokine receptor type 7 (CCR7). Furthermore, T CM cells are functionally characterized by their increased potential for proliferation after antigen reencounter. T EM cells phenotypically contrast with T CM cells in that they generally express low levels of CD62L, low levels of CD127, high levels of KLRG-1, and deficiency in CCR7. As their name implies, T EM cells display rapid effector function (granzyme B and IFN- ␥ production) but a limited proliferative potential. The high expression of CD62L and CCR7 by T CM cells allow for preferential homing to SLOs (which constitutively produce the CCR7 ligands CCL19 and CCL21), where they are well situated to protect from a systemic infection and seed the peripheral tissues with new effector cells after stimulation. In contrast, their lack of CCR7 and CD62L expression results in preferential T EM cell trafficking through nonlymphoid tissues. This trafficking pattern, in conjunction with their increased cytolytic capacity, marks them as “first responders” at the peripheral site where reinfection could occur. Taken together, these phenotypic and functional characteristics favor a model where T EM cells control the initial exposure to a pathogen at the site of infection, affording T CM cells the time required to proliferate and create a new round of effectors, ultimately promoting the final elimination of the pathogen. That said, it is safe to say that T CM and T EM cells occupy opposite ends of an everdiversifying spectrum of T cell memory subsets. Many more surface and functional markers than those described above have been identified, painting a much more nuanced view of memory T cell subsets than the simplified T CM /T EM dichotomy described above. For example, the cell surface marker KLRG-1 highly correlates with effector and T EM cell types, yet a specific function for this molecule has yet to be defined. Furthermore, populations of high-KLRG1, high-CD127 cells can be found, the function of which may be more related to T CM than T EM cells by virtue of their respon- siveness to IL-7. Thus, the very act of ...
Context 2
... of T cell activation. However, numerous lines of evidence have suggested that, although CD28 ligation is a necessary second signal, other membrane-bound and/or membrane-soluble inflammatory signals are necessary to achieve complete T cell activation, paving the way for “three-signal” and “four-signal” models as well (see below). Collectively, the data seem to indicate a role for inflammatory cytokine mediators in directing the differentiation of the stimulated T cell into an effector appropriate for the immunological insult being addressed (17, 50). Likewise, the data reflect a general role for members of the TNF receptor superfamily (CD27, OX-40, 41BB, and CD30) when interacting with their appropriate ligand on APCs (CD70, OX-40L, 41BBL, and CD30L, respectively) to promote the survival of proliferating cells through their differentiation process and on into memory (59, 77). The identification of these costimulatory signals has also provided mechanistic insights as to the connection between the innate and adaptive arms of immunity. Few (and sometimes none) of the costimulatory ligands described above are found on the surface of resting, immature APCs, i.e., an APC un- stimulated by microbes or by any proinflammatory mediators typically made by innate immune cells responding to infectious challenge (32). Thus, in the steady state, T cell interactions with a specific antigen on these resting APCs results in anergy and immune tolerance, a process that appears to be responsible for eliminating self-reactive T cells to antigens expressed only in the periphery and thereby preventing autoimmunity (7). However, when an APC becomes activated by sensing pathogens or inflammation through one or more cytokine and/or innate pattern recognition receptors, the various costimulatory ligands are expressed, allowing T cell activation, proliferation, and differentiation (40). Thus, the production of innate inflammatory signals and mediators is a necessary prelude to the effective transition to an adaptive response. Finally, T cells also express an array of inhibitory receptors, helping to fine tune the eventual response of the T cell to fit the inflammatory milieu where it was stimulated. These inhibitory receptors can act to both limit costimulatory signaling as well as costimulatory molecule ligation. A good example is cytotoxic T lymphocyte antigen (CTLA)-4, an inhibitory molecule expressed on activated T cells that both produces intracellular phosphatase activity that dampens downstream signaling of TCRs and CD28 and also acts as a competing receptor for CD80 and CD86 (indeed, CTLA-4 actually has higher affinity for CD86 binding than does CD28) (53). As a result, depending on its level of cell surface expression, CTLA-4 can directly interfere with CD28 associating with CD80/CD86. A number of other inhibitory receptors have been identified (programmed cell death-1, lymphocyte activation gene 3, and V-domain Ig suppressor of T cell activation), and blockade of their function using monoclonal antibodies is being successfully exploited clinically for the purposes of augmenting immunity against various cancers (66). T cell support of immune responses comes in two broad categories: generation of “helper” T cells and generation of “cytotoxic” T cells. A broad generalization segregates helper function to CD4 T cells and cytotoxic functionality to CD8 T cells. Other less prominent, although not necessarily less important, T cell subsets exist ( ␥␦ T cells and NK T cells) but will not be specifically addressed in this review. However, many of the principles of T cell differentiation and cytokine production described below can also apply to these other subsets, and other reviews have been directed toward their function and importance (8, 28). Helper CD4 T cell responses support the immune response by the robust generation of cytokines and chemokines that either activate neighboring cells to perform specific functions (cytokines) or recruit (chemokines) new immune cell subsets to sites of pathogen encounter. While CD8 T cells also are capable of a diverse array of cytokine production, their function appears to be largely focused on the elimination of pathogen-infected host cells by cytotoxic means. This is most commonly accomplished by the delivery of cytotoxic granules into the cytosol of the infected cell (recog- nized by TCR binding to peptide/MHC on the target cell) by a CD8 T cell. It is important to note that while these are the canonical functions of CD4 and CD8 T cells (helper cytokine production and cytotoxic activity, respectively), numerous ex- ceptions to these rules have been documented, and, in any setting, the potential of cytokine-producing helper CD8 T cells and/or cytotoxic CD4 T cells must be considered. In the face of the diverse spectrum of pathogens encountered by the host (viruses, bacteria, and parasites), the host produces a spectrum of specialized T cell responses uniquely suited to the invading pathogen (Fig. 2). Interactions between pathogens and pattern recognition receptors on cells of the innate immune system results in the production of various inflammatory cytokines. Naïve T cells retain their specificity by expression of their unique TCR but remain uncommitted to their helper fate until engagement of their TCR is accompanied by the integra- tion of molecular signals downstream of their cytokine receptors. In response to the specific cytokine environment (cytokine milieu), antigen-stimulated T cells will be genetically programmed into a variety of potential subsets that possess effector mechanisms appropriate for eliminating the pathogen. Helper T cell responses are thus classified into T helper (Th) subsets, with the major ones (although not all) designated as Th1, Th2, Th17, Th9, Tfh, and Tregs. The Th1 and Th2 subsets were so named because they were the first two subsets discov- ered (64). In recent years, a convention has emerged of naming the T cell subset based on its cytokine production profile (Th17 and Th9) or biological significance [folicular helper (Tfh) and regulatory (Treg)], which is preferred since it carries with it relevant functional, rather than historical, information about the subset in question. Figure 2 shows a visual summary for each subset described in further detail below. The generation of interferon (IFN)- ␣ /  and IL-12 cytokines in response to an intracellular pathogen (i.e., viruses and mycobacterium tuberculosis) stimulates responding T cells to induce the expression of the transcription factor T-bet (65). As a transcription factor, T-bet then docks in the promoter regions of genes, promoting T cells to differentiate into the Th1 subset (82). The Th1 subset is characterized by its generation of large and persistent amounts of IFN- ␥ and TNF- ␣ . These cytokines then feed back into the general cytokine milieu, activating neighboring cells like macrophages to elevate their phagocytic and antigen-presenting properties. Additionally, genes turned on in neighboring cells stimulated with either type of IFN will shut down host proteins commonly hijacked by viruses, thereby restricting viral replication and quarantining the virus. When naïve T cells are activated in the presence of IL-4, produced by a variety of innate cell types in response to parasites (84), it induces T cell differentiation into the Th2 subset (20). Th2 cells generate large amounts of IL-4, IL-5, and IL-13. These cytokines disseminate, activating neighboring eosinophils, mast cells, and basophils, which specialize in the elimination of parasites (83). Additionally, Th2-generated cytokines promote B cells to produce IgE and IgA isoforms of antigen-specific antibody, which circulate to mucosal surfaces and neutralize future threats of parasitic encounter. Interactions with other parasites that generate milieus rich in IL-4 and transforming growth factor (TGF)- result in the generation of a similar but developmentally distinct Th subset, Th9 (18, 87). Like Th2 cells, Th9 cells thwart parasites through the production of IL-4 and IL-13 but also produce, as their name implies, IL-9 (29). IL-9 supports CD4 T cell expansion and survival but additionally has potent effects on mast cells, promoting their activation and expansion. Both Th2 and Th9 subsets achieve their unique helper characteristics through the upregulation of genes activated by the transcription factor GATA-3 (94). GATA-3, in turn, is upregulated by IL-4 driven from the innate response. In response to extracellular bacteria and fungi (34), innate immune cells generate large amounts of both TGF-  and IL-6 (39). When naïve T cells receive these signals with additional and sustained IL-21 and IL-23 stimulation, they become “Th17” helper T cells (56) under the differential control of a transcription factor called ROR ␥ T (91). As the name suggests, this T cell subset is characterized by its ability to produce the cytokine IL-17. IL-17 potently activates neutrophils and, along with IL-8 and other chemokines generated by Th17s, strongly recruits neutrophils to the site of fungal and bacterial invasion. With their release of potent oxidative chemical species, neutrophils are able to directly kill many bacteria and fungi. The importance of Th17s is highlighted in individuals with genetic alteration resulting in diminished IL-17. These individuals suffer from recurrent, severe bacterial and fungal infections (73). While the inflammatory environment is heavily influenced by the specific nature of the invading pathogen, not all T cell differentiation is pathogenic specific. Some T cell differentiation occurs to support immune functions common to all infection responses. The cytokines IL-21 and IL-27 are generated in response to a variety of pathogens and serve to polarize naïve T cells, via the induction of transcription factor Bcl-6 (54), to specifically home to B cell follicles in SLOs (i.e., the spleen and tonsils). Once there, these Tfh cells express ...
Context 3
... typically made by innate immune cells responding to infectious challenge (32). Thus, in the steady state, T cell interactions with a specific antigen on these resting APCs results in anergy and immune tolerance, a process that appears to be responsible for eliminating self-reactive T cells to antigens expressed only in the periphery and thereby preventing autoimmunity (7). However, when an APC becomes activated by sensing pathogens or inflammation through one or more cytokine and/or innate pattern recognition receptors, the various costimulatory ligands are expressed, allowing T cell activation, proliferation, and differentiation (40). Thus, the production of innate inflammatory signals and mediators is a necessary prelude to the effective transition to an adaptive response. Finally, T cells also express an array of inhibitory receptors, helping to fine tune the eventual response of the T cell to fit the inflammatory milieu where it was stimulated. These inhibitory receptors can act to both limit costimulatory signaling as well as costimulatory molecule ligation. A good example is cytotoxic T lymphocyte antigen (CTLA)-4, an inhibitory molecule expressed on activated T cells that both produces intracellular phosphatase activity that dampens downstream signaling of TCRs and CD28 and also acts as a competing receptor for CD80 and CD86 (indeed, CTLA-4 actually has higher affinity for CD86 binding than does CD28) (53). As a result, depending on its level of cell surface expression, CTLA-4 can directly interfere with CD28 associating with CD80/CD86. A number of other inhibitory receptors have been identified (programmed cell death-1, lymphocyte activation gene 3, and V-domain Ig suppressor of T cell activation), and blockade of their function using monoclonal antibodies is being successfully exploited clinically for the purposes of augmenting immunity against various cancers (66). T cell support of immune responses comes in two broad categories: generation of “helper” T cells and generation of “cytotoxic” T cells. A broad generalization segregates helper function to CD4 T cells and cytotoxic functionality to CD8 T cells. Other less prominent, although not necessarily less important, T cell subsets exist ( ␥␦ T cells and NK T cells) but will not be specifically addressed in this review. However, many of the principles of T cell differentiation and cytokine production described below can also apply to these other subsets, and other reviews have been directed toward their function and importance (8, 28). Helper CD4 T cell responses support the immune response by the robust generation of cytokines and chemokines that either activate neighboring cells to perform specific functions (cytokines) or recruit (chemokines) new immune cell subsets to sites of pathogen encounter. While CD8 T cells also are capable of a diverse array of cytokine production, their function appears to be largely focused on the elimination of pathogen-infected host cells by cytotoxic means. This is most commonly accomplished by the delivery of cytotoxic granules into the cytosol of the infected cell (recog- nized by TCR binding to peptide/MHC on the target cell) by a CD8 T cell. It is important to note that while these are the canonical functions of CD4 and CD8 T cells (helper cytokine production and cytotoxic activity, respectively), numerous ex- ceptions to these rules have been documented, and, in any setting, the potential of cytokine-producing helper CD8 T cells and/or cytotoxic CD4 T cells must be considered. In the face of the diverse spectrum of pathogens encountered by the host (viruses, bacteria, and parasites), the host produces a spectrum of specialized T cell responses uniquely suited to the invading pathogen (Fig. 2). Interactions between pathogens and pattern recognition receptors on cells of the innate immune system results in the production of various inflammatory cytokines. Naïve T cells retain their specificity by expression of their unique TCR but remain uncommitted to their helper fate until engagement of their TCR is accompanied by the integra- tion of molecular signals downstream of their cytokine receptors. In response to the specific cytokine environment (cytokine milieu), antigen-stimulated T cells will be genetically programmed into a variety of potential subsets that possess effector mechanisms appropriate for eliminating the pathogen. Helper T cell responses are thus classified into T helper (Th) subsets, with the major ones (although not all) designated as Th1, Th2, Th17, Th9, Tfh, and Tregs. The Th1 and Th2 subsets were so named because they were the first two subsets discov- ered (64). In recent years, a convention has emerged of naming the T cell subset based on its cytokine production profile (Th17 and Th9) or biological significance [folicular helper (Tfh) and regulatory (Treg)], which is preferred since it carries with it relevant functional, rather than historical, information about the subset in question. Figure 2 shows a visual summary for each subset described in further detail below. The generation of interferon (IFN)- ␣ /  and IL-12 cytokines in response to an intracellular pathogen (i.e., viruses and mycobacterium tuberculosis) stimulates responding T cells to induce the expression of the transcription factor T-bet (65). As a transcription factor, T-bet then docks in the promoter regions of genes, promoting T cells to differentiate into the Th1 subset (82). The Th1 subset is characterized by its generation of large and persistent amounts of IFN- ␥ and TNF- ␣ . These cytokines then feed back into the general cytokine milieu, activating neighboring cells like macrophages to elevate their phagocytic and antigen-presenting properties. Additionally, genes turned on in neighboring cells stimulated with either type of IFN will shut down host proteins commonly hijacked by viruses, thereby restricting viral replication and quarantining the virus. When naïve T cells are activated in the presence of IL-4, produced by a variety of innate cell types in response to parasites (84), it induces T cell differentiation into the Th2 subset (20). Th2 cells generate large amounts of IL-4, IL-5, and IL-13. These cytokines disseminate, activating neighboring eosinophils, mast cells, and basophils, which specialize in the elimination of parasites (83). Additionally, Th2-generated cytokines promote B cells to produce IgE and IgA isoforms of antigen-specific antibody, which circulate to mucosal surfaces and neutralize future threats of parasitic encounter. Interactions with other parasites that generate milieus rich in IL-4 and transforming growth factor (TGF)- result in the generation of a similar but developmentally distinct Th subset, Th9 (18, 87). Like Th2 cells, Th9 cells thwart parasites through the production of IL-4 and IL-13 but also produce, as their name implies, IL-9 (29). IL-9 supports CD4 T cell expansion and survival but additionally has potent effects on mast cells, promoting their activation and expansion. Both Th2 and Th9 subsets achieve their unique helper characteristics through the upregulation of genes activated by the transcription factor GATA-3 (94). GATA-3, in turn, is upregulated by IL-4 driven from the innate response. In response to extracellular bacteria and fungi (34), innate immune cells generate large amounts of both TGF-  and IL-6 (39). When naïve T cells receive these signals with additional and sustained IL-21 and IL-23 stimulation, they become “Th17” helper T cells (56) under the differential control of a transcription factor called ROR ␥ T (91). As the name suggests, this T cell subset is characterized by its ability to produce the cytokine IL-17. IL-17 potently activates neutrophils and, along with IL-8 and other chemokines generated by Th17s, strongly recruits neutrophils to the site of fungal and bacterial invasion. With their release of potent oxidative chemical species, neutrophils are able to directly kill many bacteria and fungi. The importance of Th17s is highlighted in individuals with genetic alteration resulting in diminished IL-17. These individuals suffer from recurrent, severe bacterial and fungal infections (73). While the inflammatory environment is heavily influenced by the specific nature of the invading pathogen, not all T cell differentiation is pathogenic specific. Some T cell differentiation occurs to support immune functions common to all infection responses. The cytokines IL-21 and IL-27 are generated in response to a variety of pathogens and serve to polarize naïve T cells, via the induction of transcription factor Bcl-6 (54), to specifically home to B cell follicles in SLOs (i.e., the spleen and tonsils). Once there, these Tfh cells express a variety of cytokines and costimulatory molecules to assist in the germinal center reaction of B cells, promoting the robust generation of high-affinity antibodies (9). As a testament to the breadth of Tfh influence, mice genetically deficient in molecules necessary for Tfh differentiation fail to produce germinal centers or high-affinity antibodies, a condition that makes them more susceptible to a broad spectrum of infectious agents (16). Unchecked, persistent, or overexuberant immune responses carry with them the danger of immunopathology. To avert this disaster, the immune system produces Tregs, a regulatory subset of T cells that puts the brakes on a variety of inflammatory processes (51, 75). Unlike other T cell subtypes, Tregs can be produced directly from thymic selection [natural Tregs (nTregs)] as well as differentiated [induced Tregs (iTregs)] under the influence of environmental factors such as TGF-  and retinoic acid (12). In either type of Treg, suppressive activity is mediated by active expression of the transcription factor FOXP3 (22, 33). The hallmark effects of Tregs are the impairment of T cell proliferation and cytokine production from other T cell subsets despite their engagement of their ...
Similar publications
Germinal center (GC) B cells cycle between two states, the light zone (LZ) and the dark zone (DZ), and in the latter they proliferate and hypermutate their immunoglobulin genes. How this functional transition takes place is still controversial. In this study, we demonstrate that ablation of Foxo1 after GC development led to the loss of the DZ GC B...
Lymphocyte costimulation plays a central role in immunology, inflammation, and immunotherapy. The inducible T cell costimulator (ICOS) is expressed on T cells following peptide: MHC engagement with CD28 costimulation. The interaction of ICOS with its sole ligand, the inducible T cell costimulatory ligand (ICOSL; also known as B7-related protein-1),...
We investigated the basis for partial reactivity of naive CD8 T cells expressing an alloreactive transgenic TCR in response to a mutant alloantigen. When unstimulated APCs were used, IFN-gamma as well as IL-2 and cell proliferation were observed in response to wild-type Ag, whereas mutant Ag induced only IFN-gamma DNA binding and reporter gene assa...
The development and activation of MHC class II (MHC-II)-restricted CD4(+) T cells are distinct immunological processes that are strictly MHC-II-dependent. To address their relative dependence on MHC-II, we established a novel ENU-induced mutant mouse on the C57BL/6 background, named I-A(12%), with ∼8-fold reduced I-A expression on the surface of B...
Background:
-High fat diet (HFD) promotes endothelial dysfunction and pro-inflammatory monocyte activation, which contribute to atherosclerosis in obesity. We investigated whether HFD also induces dysfunction of red blood cells (RBC), which serve as a reservoir for chemokines via binding to Duffy antigen receptor for chemokines (DARC).
Methods an...
Citations
... Another vital function is the generation of immunological memory. Upon encountering a pathogen for the first time, certain lymphocytes differentiate into memory cells, which can recognize and mount a rapid and robust response upon subsequent exposure to the same pathogen (Pennock et al., 2013). This process forms the basis of immunization and provides long-lasting protection. ...
... This process forms the basis of immunization and provides long-lasting protection. Additionally, the adaptive immune system exhibits immunological tolerance to prevent immune reactions against the body's own tissues (Pennock et al., 2013). Furthermore, the adaptive immune response involves the production of antibodies by B cells, which can neutralize pathogens or mark them for destruction by other immune cells (LeBien and Tedder, 2008). ...
Cellular senescence has been implicated in the pathophysiology of many age-related diseases. However, it also plays an important protective role in the context of tumor suppression and wound healing. Reducing senescence burden through treatment with senolytic drugs or the use of genetically targeted models of senescent cell elimination in animals has shown positive results in the context of mitigating disease and age-associated inflammation. Despite positive, albeit heterogenous, outcomes in clinical trials, very little is known about the short-term and long-term immunological consequences of using senolytics as a treatment for age-related conditions. Further, many studies examining cellular senescence and senolytic treatment have been demonstrated in non-infectious disease models. Several recent reports suggest that senescent cell elimination may have benefits in COVID-19 and influenza resolution and disease prognosis. In this review, we discuss the current clinical trials and pre-clinical studies that are exploring the impact of senolytics on cellular immunity. We propose that while eliminating senescent cells may have an acute beneficial impact on primary immune responses, immunological memory may be negatively impacted. Closer investigation of senolytics on immune function and memory generation would provide insight as to whether senolytics could be used to enhance the aging immune system and have potential to be used as therapeutics or prophylactics in populations that are severely and disproportionately affected by infections such as the elderly and immunocompromised.
... An effective T cell response typically involves transitioning from a naïve state to an activated state that culminates in the formation of long-lasting memory. When stimulated by an antigen, naïve T cells become activated and undergo subsequent expansion (11). Subsequently, a subset of these activated cells undergoes maturation, giving rise to various memory subsets, including central memory T cells, effector memory T cells and tissue-resident memory T cells (12). ...
IntroductionSeveral efforts have been made to describe the complexity of T cell heterogeneity during the COVID-19 disease; however, there remain gaps in our understanding in terms of the granularity within.Methods
For this attempt, we performed a single-cell transcriptomic analysis of 33 individuals (4 healthy, 16 COVID-19 positive patients, and 13 COVID-19 recovered individuals).ResultsWe found CD8+ T cell-biased lymphopenia in COVID-19 patients compared to healthy and recovered individuals. We also found an optimal Th1/Th2 ratio, indicating an effective immune response during COVID-19. Expansion of activated CD4+ T and NK T was detected in the COVID-19-positive individuals. Surprisingly, we found cellular and metal ion homeostasis pathways enriched in the COVID-19-positive individuals compared to the healthy and recovered in the CD8+ T cell populations (CD8+ TCM and CD8+ TEM) as well as activated CD4+ T cells.DiscussionIn summary, the COVID-19-positive individuals exhibit a dynamic T cell mediated response. This response may have a possible association with the dysregulation of non-canonical pathways, including housekeeping functions in addition to the conventional antiviral immune response mediated by the T cell subpopulation. These findings considerably extend our insights into the heterogeneity of T cell response during and post-SARS-CoV-2 infection.
... An effective T cell response typically involves transitioning from a naïve state to an activated state that culminates in the formation of long-lasting memory. When stimulated by an antigen, naïve T cells become activated and undergo subsequent expansion (11). Subsequently, a subset of these activated cells undergoes maturation, giving rise to various memory subsets, including central memory T cells, effector memory T cells and tissue-resident memory T cells (12). ...
Introduction
Several efforts have been made to describe the complexity of T cell heterogeneity during the COVID-19 disease; however, there remain gaps in our understanding in terms of the granularity within.
Methods
For this attempt, we performed a single-cell transcriptomic analysis of 33 individuals (4 healthy, 16 COVID-19 positive patients, and 13 COVID-19 recovered individuals).
Results
We found CD8+ T cell-biased lymphopenia in COVID-19 patients compared to healthy and recovered individuals. We also found an optimal Th1/Th2 ratio, indicating an effective immune response during COVID-19. Expansion of activated CD4+ T and NK T was detected in the COVID-19-positive individuals. Surprisingly, we found cellular and metal ion homeostasis pathways enriched in the COVID-19-positive individuals compared to the healthy and recovered in the CD8+ T cell populations (CD8+ TCM and CD8+ TEM) as well as activated CD4+ T cells.
Discussion
In summary, the COVID-19-positive individuals exhibit a dynamic T cell mediated response. This response may have a possible association with the dysregulation of non-canonical pathways, including housekeeping functions in addition to the conventional antiviral immune response mediated by the T cell subpopulation. These findings considerably extend our insights into the heterogeneity of T cell response during and post-SARS-CoV-2 infection.
Keywords: single cell RNA-seq, COVID-19, T cell heterogeneity, non-canonical, metal ion
... CD4 + T cells constitutively express TCR αβ and CD4 molecules, representing an integral element of the adaptive immune response with different immune functions [1,2]. CD4 + T cells at different stages of development have different metabolic characteristics that are compatible with their immune function [3]. ...
Background
Bcl-3 is a member of the IκB protein family and an essential modulator of NF-κB activity. It is well established that Bcl-3 is critical for the normal development, survival and differentiation of adaptive immune cells, especially T cells. However, the regulation of immune cell function by Bcl-3 through metabolic pathways has rarely been studied.
Results
In this study, we explored the role of Bcl-3 in the metabolism and function of T cells via the mTOR pathway. We verified that the proliferation of Bcl-3-deficient Jurkat T cells was inhibited, but their activation was promoted, and Bcl-3 depletion regulated cellular energy metabolism by reducing intracellular ATP and ROS production levels and mitochondrial membrane potential. Bcl-3 also regulates cellular energy metabolism in naive CD4⁺ T cells. In addition, the knockout of Bcl-3 altered the expression of mTOR, Akt, and Raptor, which are metabolism-related genes, in Jurkat cells.
Conclusions
This finding indicates that Bcl-3 may mediate the energy metabolism of T cells through the mTOR pathway, thereby affecting their function. Overall, we provide novel insights into the regulatory role of Bcl-3 in T-cell energy metabolism for the prevention and treatment of immune diseases.
... In this case, induction of IFN-γ and IL-12 cytokines may activate cytotoxic T-cell and Th1-responses. IL-6 has a standing for the gaps concerning innate as well as adoptive immune response, and helps T cells to secrete IFN-γ and IL-2 [47,48]. IFN-γ, TNF-α also activates macrophages for antibacterial activity. ...
Typhoid and emerging paratyphoid fever are a severe enteric disease worldwide with high morbidity and mortality. Licensed typhoid vaccines are in the market, but no paratyphoid vaccine is currently available. In the present study we developed a bivalent vaccine against Salmonella Typhi and Paratyphi A using a bacterial ghost platform. Bacterial ghost cells (BGs) are bacteria-derived cell membranes without cytoplasmic contents that retain their cellular morphology, including all cell surface features. Furthermore, BGs have inherent adjuvant properties that promote an enhanced humoral and cellular immune reaction to the target antigen. Sodium hy-droxide was used to prepare ghost cells of Salmonella Typhi and Paratyphi A. The bacterial ghost cells were characterised using electron microscopy. Then BALB/c mice were immunized three times (0 th , 14 th and 28 th day) with the bivalent typhoidal bacterial ghost cells. Haematological study of adult mice throughout immunization period reflected that the immunogen was safe to administer and does not affect the animals' health. After complete immunization, we found significant serum antibody titter against whole cell lysate, outer membrane protein and lipopolysaccharide of both bacteria, and cell-mediated immunity was observed in an ex-vivo experiment. CD4+, CD8a+ and CD19+ splenic cell populations were increased in immunized animals. Biva-lent Typhoidal ghost cell immunized mice showed better survival, less bacterial colonization in systemic organs, and less inflammation and/or destruction of tissue in histopathological analysis than non-immunized control mice. Serum antibodies of immunized animals can significantly inhibit bacterial motility and mucin penetration ability with better killing properties against Salmonella Typhi and Paratyphi A. Furthermore, significant passive protection was observed through the adoptive transfer of serum antibody and lymphocytes of immunized animals to naïve animals after bacterial infection. In summary, Bivalent Typhoidal Bacterial Ghost cells (BTBGs) enhances immunogenic properties and serves as a safe and effective prevention strategy against Salmonella Typhi and Paratyphi A.
... According to the ICD model, this could be elucidated by exploring changes in subsets of T cells, which are a major component of adaptive immunity. Considering the peak response of antigen-exposed T cells within 1-2 weeks and early T-cell changes observed a week post PD-1 inhibitor administration in a previous study, it is possible that chemotherapy's impact on systemic immunity via the tumoricidal effect could be detected early in T cells [29,39,40]. ...
... From this point onward, neoantigens produced by the tumor interact with immune cells in the TME. Considering that the T-cell response generally peaks within 1-2 weeks following antigen exposure, the changes in immune cells observed on day 7 seem to occur in a sufficient timeframe for cancer cell response to chemotherapy to manifest and be reflected in systemic immune cells [39,40]. ...
The clinical significance of PD-1 expression in circulating CD8+ T cells in patients with gastric cancer (GC) receiving chemotherapy remains unelucidated. Therefore, we aimed to examine its prognostic significance in blood samples of 68 patients with advanced GC who received platinum-based chemotherapy. The correlation between peripheral blood mononuclear cells, measured using fluorescence-activated cell sorting, was evaluated. Patients were divided into two groups according to the changes in PD-1+CD8+ T-cell frequencies between day 0 and 7. They were categorized as increased or decreased PD-1+CD8+ T-cell groups. The increased PD-1+CD8+ T-cell group showed longer progression-free survival (PFS) and overall survival (OS) than the decreased PD-1+CD8+ T-cell group (PFS: 8.7 months vs. 6.1 months, p = 0.007; OS: 20.7 months vs. 10.8 months, p = 0.003). The mean duration of response was significantly different between the groups (5.7 months vs. 2.5 months, p = 0.041). Multivariate analysis revealed that an increase in PD-1+CD8+ T-cell frequency was an independent prognostic factor. We concluded that the early increase in PD-1+CD8+ T-cell frequency is a potential predictor of favorable prognoses and durable responses in patients with advanced GC receiving chemotherapy.
... T cells are an essential component of the adaptive immune response, and can be divided into two main types: CD4+ T cells, also called "helper" cells, and CD8+ T cells, also known as "cytotoxic" cells [56]. Another type of cytotoxic immune cell found in the innate immune response is an NK cell; these are well-known for their ability to destroy virally infected cells, and to detect and control early cancer cells [57]. ...
Millions globally suffer from myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). The inflammatory symptoms, illness onset, recorded outbreak events, and physiological variations provide strong indications that ME/CFS, at least sometimes, has an infectious origin, possibly resulting in a chronic unidentified viral infection. Meanwhile, studies exposing generalized metabolic disruptions in ME/CFS have stimulated interest in isolated immune cells with an altered metabolic state. As the metabolism dictates the cellular function, dissecting the biomechanics of dysfunctional immune cells in ME/CFS can uncover states such as exhaustion, senescence, or anergy, providing insights into the consequences of these phenotypes in this disease. Despite the similarities that are seen metabolically between ME/CFS and other chronic viral infections that result in an exhausted immune cell state, immune cell exhaustion has not yet been verified in ME/CFS. This review explores the evidence for immunometabolic dysfunction in ME/CFS T cell and natural killer (NK) cell populations, comparing ME/CFS metabolic and functional features to dysfunctional immune cell states, and positing whether anergy, exhaustion, or senescence could be occurring in distinct immune cell populations in ME/CFS, which is consistent with the hypothesis that ME/CFS is a chronic viral disease. This comprehensive review of the ME/CFS immunometabolic literature identifies CD8+ T cell exhaustion as a probable contender, underscores the need for further investigation into the dysfunctional state of CD4+ T cells and NK cells, and explores the functional implications of molecular findings in these immune-cell types. Comprehending the cause and impact of ME/CFS immune cell dysfunction is critical to understanding the physiological mechanisms of ME/CFS, and developing effective treatments to alleviate the burden of this disabling condition.
... The fundamental mechanism of intestinal involvement in X-SCID is little understood. CD4+ T cells coordinate effective immune responses, retain immune tolerance, and participate in memory cell differentiation [12,13]. According to mouse model studies, the colonic microbiota is critical for recruiting an appropriate level of CD4 Foxp3-expressing regulatory T cells (Treg) to the colon to inhibit inflammation [14]. ...
Background
X-linked severe combined immunodeficiency is caused by IL2RG gene mutation. Several variations have been identified in the IL2RG gene, which potentially can prevent the production of nonfunctional proteins. Herein, a novel X-linked variant in the IL2RG gene is reported in twin brothers, associated with inflammatory bowel symptoms.
Case presentation
The patients were 26-month-old monozygotic twin middle-eastern males with failure to thrive and several inpatient admissions due to severe chronic nonbloody diarrhea that started at the age of 12 months. Pancolitis was revealed after performing upper and lower gastrointestinal endoscopies on the twin with more severe gastrointestinal symptoms. Flow cytometric evaluation of the peripheral blood cells showed low levels of CD4+ cells in both patients. Next generation sequencing-based gene panel test results of the two patients proved a novel heterozygous missense X-linked IL2RG mutation (70330011 A > G, p.Trp197Arg) in one of the patients, which was predicted to be deleterious (CADD score of 28), which soon after was confirmed by Sanger segregation in his twin brother. Both parents were wild types and had never experienced similar symptoms. The patients received an human leukocyte antigen (HLA)-matched cord blood transplant. The twin with more severe gastrointestinal symptoms died 1 month after transplantation. In his brother, watery diarrhea eventually subsided after transplantation.
Conclusion
Intestinal involvement in X-linked severe combined immunodeficiency is a rare presentation that might be neglected. The increasing availability of genetic screening tests worldwide could be helpful for early detection of such lethal primary immunodeficiency diseases and in implementing effective interventions to handle the severe outcomes.
... Our study showed that the levels of B cells were significantly lower in patients with distant metastatic (33,34). Memory CD4 + T cells represent adaptive immune memory (35), which may be induced by memory CD4 + T cells during a second encounter with pathogens. CD28 is a critical costimulatory molecule that participates as a secondary signal for activating CD4 + and CD8 + T cells to produce an antitumor response. ...
Objective
The prognosis of patients with advanced cancers of the upper gastrointestinal (UGI) tract is poor. Systemic chemotherapy forms the basis for their treatment, with limited efficacy. Biomarkers have been introduced into clinical practice for cancer management. This study aimed to investigate the predictive and prognostic values of circulating biomarkers in patients with advanced esophageal and gastric cancers receiving chemotherapy.
Design
Overall, 92 patients with advanced esophageal squamous cell carcinoma (ESCC; n = 38) and gastric adenocarcinoma (GAC; n = 54) were enrolled. We analyzed the association of circulating lymphocyte subsets, inflammatory markers, and blood cell counts with treatment efficacy and patient survival.
Results
Significant differences were identified in peripheral blood parameters between the groups with different clinicopathological features. Hemoglobin (Hb, p = 0.014), eosinophil counts ( p = 0.028), CD4 ⁺ CD28 ⁺ T/CD4 ⁺ T percentage ( p = 0.049), CD8 ⁺ CD38 ⁺ T/CD8 ⁺ T percentage ( p = 0.044), memory CD4 ⁺ T ( p = 0.007), and CD4 ⁺ CD28 ⁺ T ( p = 0.007) were determined as predictors for achieving non-PD (progression disease) in the ESCC cohort. High levels of eosinophils ( p = 0.030) and memory CD4 ⁺ T cells ( p = 0.026) and high eosinophil-to-lymphocyte ratio (ELR, p = 0.013) were predictors of non-PD in patients with GAC. The combined detection models exhibited good ability to distinguish between partial response (PR)/non-PR and PD/non-PD in patients with ESCC and GAC, respectively. Using the multivariate Cox model, the Eastern Cooperative Oncology Group (ECOG) score status (hazard ratio [HR]: 4.818, 95% confidence intervals [CI]: 2.076–11.184, p < 0.001) and eosinophil count (HR: 0.276, 95% CI: 0.120–0.636, p = 0.003) were independent prognostic factors of progression-free survival (PFS) in patients with ESCC. Metastatic sites (HR: 2.092, 95% CI: 1.307–3.351, p = 0.002) and eosinophil-to-lymphocyte ratio (ELR; HR: 0.379, 95% CI: 0.161–0.893, p = 0.027) were independent prognostic factors for overall survival (OS) in patients with ESCC. Differentiation (HR: 0.041, 95% CI: 0.200–0.803, p = 0.010), memory CD4 ⁺ T (HR: 0.304, 95% CI: 0.137–0.675, p = 0.003), NK cells (HR: 2.302, 95% CI: 1.044–3.953, p = 0.037), and C-reactive protein-to-lymphocyte ratio (CLR; HR: 2.070, 95% CI: 1.024–4.186, p = 0.043) were independent prognostic factors for PFS in patients with GAC. Total lymphocyte counts (HR: 0.260, 95% CI: 0.086–0.783, p = 0.017), CD8 ⁺ T (HR: 0.405, 95% CI: 0.165–0.997, p = 0.049), NK cells (HR: 3.395, 95% CI: 1.592–7.238, p = 0.002), and monocyte-to-lymphocyte ratio (MLR; HR: 3.076, 95% CI: 1.488–6.360, p = 0.002) were identified as independent prognostic factors associated with OS of GAC.
Conclusion
Lymphocyte subsets, blood cell counts, and inflammatory parameters may predict the chemotherapeutic response and prognosis in ESCC and GAC. A combination of these markers can be used to stratify patients into risk groups, which could improve treatment strategies.
... The differentiation process of Th1 cells is tightly regulated by a feedback loop, with Th1 differentiation mainly regulated by TF T-bet, which can promote IFN-g secretion by upregulating IL-12Rb2 receptor expression and T-bet expression regulated by signal transducer and activator of transcription 1 (STAT1), which is in turn activated by IFN-g secreted by TH1 cells (133). When the feedback loop in activated T cells is disrupted, reduced IL-12 secretion decreases that of IFN-g and T-bet, thereby affecting the Th1 cell-mediated immune response (134)(135)(136)(137)(138)(139)(140). ...
Reactive oxygen species (ROS) are produced both enzymatically and non-enzymatically in vivo . Physiological concentrations of ROS act as signaling molecules that participate in various physiological and pathophysiological activities and play an important role in basic metabolic functions. Diseases related to metabolic disorders may be affected by changes in redox balance. This review details the common generation pathways of intracellular ROS and discusses the damage to physiological functions when the ROS concentration is too high to reach an oxidative stress state. We also summarize the main features and energy metabolism of CD4 ⁺ T-cell activation and differentiation and the effects of ROS produced during the oxidative metabolism of CD4 ⁺ T cells. Because the current treatment for autoimmune diseases damages other immune responses and functional cells in the body, inhibiting the activation and differentiation of autoreactive T cells by targeting oxidative metabolism or ROS production without damaging systemic immune function is a promising treatment option. Therefore, exploring the relationship between T-cell energy metabolism and ROS and the T-cell differentiation process provides theoretical support for discovering effective treatments for T cell-mediated autoimmune diseases.