
Citation: Daniil Shevyrev, Valeriy Tereshchenko, Olesya Manova, Vladimir kozlov. Homeostatic proliferation as a physiological process and a risk factor for autoimmune pathology[J]. AIMS Allergy and Immunology, 2021, 5(1): 18-32. doi: 10.3934/Allergy.2021002
[1] | Ryuta Muromoto, Kenji Oritani, Tadashi Matsuda . IL-17 signaling is regulated through intrinsic stability control of mRNA during inflammation. AIMS Allergy and Immunology, 2022, 6(3): 188-199. doi: 10.3934/Allergy.2022014 |
[2] | Izabela Krzemień, Angelika Szatan, Paulina Skalska, Martyna Cieślik, Angelika Domagała, Magdalena Gębicka, Krzysztof Bryniarski, Katarzyna Nazimek . Serum cytokine concentrations correlate with the severity of imiquimod and IL-23-induced psoriatic inflammation in mice. AIMS Allergy and Immunology, 2024, 8(4): 265-282. doi: 10.3934/Allergy.2024016 |
[3] | Catalina Marysol Carvajal Gonczi, Fadi Touma, Tina Daigneault, Chelsea Pozzebon, Kelly Burchell-Reyes, Peter J. Darlington . Modulation of IL-17A and IFNγ by β2-adrenergic agonist terbutaline and inverse-agonist nebivolol, influence of ADRB2 polymorphisms. AIMS Allergy and Immunology, 2021, 5(4): 222-239. doi: 10.3934/Allergy.2021017 |
[4] | Ryuta Muromoto, Kenji Oritani, Tadashi Matsuda . Tyk2-mediated homeostatic control by regulating the PGE2-PKA-IL-10 axis. AIMS Allergy and Immunology, 2021, 5(3): 175-183. doi: 10.3934/Allergy.2021013 |
[5] | Xi Luo, Hehong Li, Rongshan Chen, Yinhui Zeng, Wenlong Liu, Qingxiang Zeng . MicroRNA-155 is a critical regulator of regulatory T cells in OM-85 Broncho-Vaxom treated experimental models of allergic rhinitis. AIMS Allergy and Immunology, 2024, 8(1): 8-17. doi: 10.3934/Allergy.2024002 |
[6] | Chunyan Li, Wojciech Dawicki, Xiaobei Zhang, Chris Rudulier, John R. Gordon . IL-10- and retinoic acid-induced regulatory dendritic cells are therapeutically equivalent in mouse models of asthma and food allergy. AIMS Allergy and Immunology, 2021, 5(2): 73-91. doi: 10.3934/Allergy.2021007 |
[7] | Tuba Karakurt, Nurhan Kasap, Kübra Aslan, Hayrunnisa Bekis Bozkurt, Fatma Bal Cetinkaya, Gizem Uslu, Zafer Bicakci, Filiz Özen, Özlem Cavkaytar, Ahmet Eken, Mustafa Arga . Suspected ALPS with clinical and laboratory findings: Three patients—three different diagnoses. AIMS Allergy and Immunology, 2023, 7(4): 304-312. doi: 10.3934/Allergy.2023020 |
[8] | Masahiro Seike . Histamine suppresses T helper 17 responses mediated by transforming growth factor-β1 in murine chronic allergic contact dermatitis. AIMS Allergy and Immunology, 2018, 2(4): 180-189. doi: 10.3934/Allergy.2018.4.180 |
[9] | Sabrine Louhaichi, Mariem Salhi, Anissa Berraïes, Besma Hamdi, Jamel Ammar, Kamel Hamzaoui, Agnès Hamzaoui . Co-inhibitory receptors in female asthmatic patients: Correlation with IL-17 and IL-26. AIMS Allergy and Immunology, 2018, 2(1): 10-23. doi: 10.3934/Allergy.2018.1.10 |
[10] | Kamyar Nasiri, Zohreh Mortezania, Sanaz Oftadehbalani, Mohammad Khosousi Sani, Amin Daemi, Seyyedeh Touran Hosseini, Yusuf Döğüş, Zafer Yönden . Parasitology and Covid-19: epidemiological, immunological, pathological, and therapeutic aspects. AIMS Allergy and Immunology, 2023, 7(1): 92-103. doi: 10.3934/Allergy.2023007 |
To protect the organism from various antigen challenges (different infectious agents or altered self-antigens), the immune system has to maintain the diversity and size of the peripheral pool of T-lymphocytes at a constant level that is kept within a narrow range [1]. Before the involution of the thymus, restoration of the T-cell pool occurs due to the migration of naïve T-lymphocytes from the thymus, and with age, homeostatic proliferation (HP) starts to play the key role [2]. HP is a physiological process induced by lymphopenia and targeted for the restoration and maintenance of the peripheral pool of T-lymphocytes [3]–[5]. HP requires free lymphocyte niches that become free in the conditions of lymphopenia and occurs in the T-cell zones of lymph nodes and spleen [6], which is confirmed by the disturbance of the process of HP in mice deprived of secondary lymphoid organs [7]. The main factors that provide HP are the signal from the T-cell receptor (TCR), signals of co-stimulation, as well as IL-7 and IL-15 [8]–[12]. At the same time, the intensity of HP directly depends on the TCR signal strength, which is determined by the avidity of the interaction between TCR and peptide in the Major Histocompatibility Complex (MHC). For HP of low intensity (slow HP), it is enough to have a “tonic” TCR signal and elevated levels of IL-7 and IL-15. It is of a polyclonal character and, probably, does not lead to negative consequences by maintaining the diversity of TCR. HP of high intensity (fast HP) on the other hand primarily depends on a strong TCR signal, is of oligoclonal character, and leads to the changes in the TCR landscape and formation of lymphocytes with the memory-like T-cells, increasing the risk of the development of autoimmune pathologies [13]–[18]. We define the TCR landscape not only as the diversity of TCR but also as the frequency of occurrence of each specific TCR. Thus, the strength of TCR signal that T-lymphocytes receive in the conditions of lymphopenia not only influences the rate of restoration of the T-cell pool but also determines qualitative changes in the immune system that occur during HP.
It is well-known that during maturation, thymocytes go through the stages of positive and negative selection. Positive selection results in the selection of T-lymphocytes capable of recognizing antigenic determinants in complex with MHC. Negative selection results in the elimination of lymphocytes that have a high affinity to self-antigens in MHC complexes [19]. Lymphocytes with relatively high affinity to self-antigens die as a result of apoptosis. Lymphocytes with moderate affinity to self-antigens become T-regulatory cells (Tregs), and lymphocytes with relatively low affinity represent effector T-lymphocytes capable of recognizing foreign antigens. It should be noted that the affinity of TCR to cognate antigens differs in various populations of T-lymphocytes. The majority of self-reactive T-cells have a significantly lower absolute affinity to self-antigens than effector T-lymphocytes to foreign antigens, and the absolute affinity of TCRs of anti-tumor lymphocytes to neoantigens lies somewhere in the middle [20]. The mechanisms of central tolerance and, in particular, negative selection are not completely efficient and some potentially self-reactive T-lymphocytes with a high affinity of TCRs to self-antigens can get into the peripheral bloodstream [21]–[24]. This leads to the risk of the development of autoimmune diseases and requires the mechanisms of peripheral tolerance [25]. It should be noted that because of the limited size of the pool of T-cells, there is a compromise observed between the diversity of TCRs and the number of the existing cells of a given specificity. If the TCR repertoire is too diverse, the rate of occurrence of lymphocytes of a given specificity becomes too low for timely reaction to a pathogen. However, if the repertoire is not sufficiently diverse, foreign antigens may be missed. The minimal TCR repertoire that provides the optimal balance between the diversity and the number of T-lymphocytes of a given specificity is called protecton [19],[26]. According to some data, in humans, the general diversity of TCRs of naïve T-lymphocytes reaches 108 variants [27]. The mechanisms described above are involved in the formation of the TCR specificity landscape of naive T-lymphocytes (Figure 1).
After leaving the thymus, some effector T-lymphocytes are differentiated into peripherally-induced pTregs when they face antigens of symbiotic microflora. Thus, the general repertoire of TCRs of T-lymphocytes is characterized by the presence of thymic tTregs with relatively high affinity to self-antigens, effector T-lymphocytes with high affinity of TCRs to foreign antigens and low affinity to self-antigens, pTregs with high affinity to antigens of commensal microflora, as well as some self-reactive T-lymphocytes [28],[29]. It should be mentioned that TCR specificity spectra of the described subtypes of T-cells barely overlap [30],[31].
Well-known, a combination of genetic predisposition (including SNP mutations in the genes of MHC, enzymes, cytokines, or their receptors) with an action of unfavorable environmental factors is essential for autoimmune disease development [32],[33]. In this context, the escape of self-reactive T-lymphocytes from the negative selection in the thymus, as well as the presence of anti-tumor T-lymphocytes with intermediate affinity to self-antigens, create an additional risk of the development of autoimmune processes. At the same time, the realization of autoimmune pathology requires certain disturbances in the mechanisms that provide peripheral tolerance. The authors believe that such disturbances can occur in cases with lymphopenia during the process of HP, which is confirmed by the association between lymphopenia and different autoimmune diseases [34]–[37]. So, signs of autoimmunity are frequently observed in various pathologies that are accompanied by lymphopenia, such as rheumatoid arthritis, Sjogren's syndrome, insulin-dependent diabetes mellitus, Crohn's disease, systemic lupus erythematosus, HIV-infection, etc. [34],[38].
During life, the human organism is affected by numerous physical, chemical, and infectious factors that require a reaction from the immune system side, and often associated with loss of immune equilibrium and development of lymphopenia. Obviously, there are mechanisms aimed at returning to the initial state or attaining a new equilibrium state by the immune system. For example, it is observed during the resolution of the immune response when the excessive amounts of lymphocytes eliminate via apoptosis because of the lack of specific antigen stimulation, and the excessive amount of antibodies is corrected via idiotype–anti-idiotype interactions according to the immune network theory of Niels Jerne [39]. Another example is the process of restoration of the number of lymphocytes after lymphopenia, which occurs via the proliferation on the periphery and is limited by the number of vacant niches that provide the lymphocytes with the factors of survival. In both cases, homeostatic mechanisms get involved that target the quantitative return to the steady-state conditions of the immune system. However, at the same time, its qualitative characteristics can change. In general, a dynamic equilibrium in the immune system is controlled by universal mechanisms that include TCR, humoral, and co-stimulating molecule signals. As was mentioned above, these three groups of stimuli (TCR signals, humoral, and co-stimulating signals) play the most important role in the process of HP. Moreover, the contribution of one or another type of signals differs for slow and fast HP.
Slow HP occurs in the conditions of moderate lymphopenia. The division of cells is observed rarer than 1 time per day; at the same time, CD8+ lymphocytes are divided faster than CD4+ cells [13],[40]. In regular conditions, for the maintenance of homeostasis and basal activity, T-lymphocytes require a “tonic” TCR signal from the interaction between the TCR and peptide in the complex with the MHC on dendritic cells. As a result, partial phosphorylation of ζ-chain of TCR complexes occurs. [41]–[43]. This is observed in the lymphoid organs in course of the continuous monitoring of dendritic cells by T-lymphocytes for exogenous antigens [43]. In the conditions of lymphopenia, IL-7 and IL-15 are produced. IL-7 is produced by the cells of the reticuloendothelial system, stromal cells of the thymus, and bone marrow [44]. IL-15 is produced by the cells of monocyte/macrophage lineage [45], epithelial [46], and dendritic cells [47]. Both IL-7 and IL-15 along with IL-2 belong to the common cytokine receptor γ-chain family and bind with receptor complexes that contain γ-chain/IL-2R-γ—CD127, CD215, and CD25, respectively. In the majority of cases, the effect of IL-7 and IL-15 was mediated by the activation of three signal pathways: JAK-STAT, PI3-K-Akt, and RAS-RAF-MAPK [48],[49]. The activation of the mentioned signal pathways under the influence of IL-7 and IL-15, as well as tonic TCR signal, provide the required level of activation for the survival and proliferation of T-lymphocytes during slow HP. As it was mentioned earlier, such proliferation occurred in the conditions of moderate lymphopenia and had a polyclonal character maintaining the diversity of the TCR repertoire. It is interesting to note that the blocking of TCR-pMHC interaction or the introduction of neutralizing antibodies against IL-7 leads to the inhibition of slow HP [13],[50],[51]. It should be mentioned that IL-7 is important for slow HP of CD4+ and CD8+ lymphocytes, while IL-15 is more important for CD8+ cells [52],[53]. Polyclonality of slow HP is determined at the first stage of selection in the thymus (during positive selection) when lymphocytes are selected that have TCRs capable of recognizing self-antigens in the complex of the MHC. Thus, each T-lymphocyte in the diversity of T-cells can receive a weak “tonic” signal from the TCR and get involved in slow HP under the effect of the elevated levels of IL-7 and IL-15 in the conditions of lymphopenia. This provides not only a quantitative restoration of the pool of T-cells but also the maintenance of the diversity of the TCR repertoire (Figure 2).
Fast HP (in some publications—spontaneous proliferation) is observed in the conditions of “severe” lymphopenia, when a great number of lymphocytic niches become unoccupied. The division of cells occurs more often than 1 time per day. For fast HP of CD8+ lymphocytes, it is enough to contact the epitope in the MHC-I or MHC-II complexes on dendritic cells or B-cells. At the same time, for the induction of fast HP of CD4+ lymphocytes, the TCR has to contact with the epitope in the MHC-II complex on CD11+ dendritic cells [55]. Fast HP requires a strong TCR signal. It is less dependent on cytokines IL-7 and IL-15 and leads to the formation of cells with effector or memory-like phenotype [13],[18]. As was noted before, the strength of the TCR signal, i.e. the avidity of the interaction of TCR-pMHC, is the most important factor that determines the intensity of HP. Recent studies showed that weak and strong TCR signals activated different pathways of intracellular signaling. Thus, a weak TCR signals generated elevated phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and reduced phosphatidylinositol (3,4,5)-trisphosphate (PIP3) levels, whereas strong TCR signals reduced PI(4,5)P2 and elevated PIP3 levels [56]. Low PIP3 levels generated by weak TCR signals were sufficient to activate phosphoinositide-dependent kinase-1 to phosphorylate AKT on Thr-308 but insufficient to activate mTOR complex 2 (mTORC2), whereas elevated PIP3 levels generated by a strong TCR signal were required to activate mTORC2 to phosphorylate Ser-473 on AKT [56]. Different proteoforms of Akt generated by weak and strong TCR signals have different substrate specificity and activate different descending signal pathways. Thus, various strength of the TCR signal determines the activation of different intracellular signal pathways, which determines cell fate [56]. This provides a materialistic background for the division of HP depending on the strength of the TCR signal into slow and fast (Figure 2).
Earlier, the authors shortly described the mechanisms of the formation of the naïve TCR repertoire in the thymus, which occurred due to the action of the factors Aire and Fezf2 [57]. Further formation of the general TCR repertoire is observed in the periphery [58]. In both cases, the determining role is played by the repertoires represented by antigens in the MHC complexes (pMHC). Thus, the diversity of the TCR repertoire of T-lymphocytes depends on the diversity of the representing antigens: the landscape of recognition (TCR) is formed by the landscape of presentation (pMHC). As it was mentioned before, one of the required conditions for fast HP of T-cells is a contact between the TCR and peptide-MHC complex. The avidity of this interaction determines the strength of the TCR signal and the capability of T-cells to compete for the factors that provide proliferation. Thus, urgent restoration of the T-cells pool as a result of fast HP cannot take place with the complete preservation of TCR diversity due to not all possible antigens are present simultaneously in the body, which cannot provide a fast antigen-specific proliferation of the entire diversity of T-lymphocytes. As a result, only 30% of T-lymphocytes get involved in this process. Primarily, these are the cells with relatively high affinity to self-antigens and antigens of commensal flora [10],[15],[17],[36]. Experiments in vivo showed that antigens of commensal microflora also played an important role in the provision of fast HP; it significantly reduced in germ-free lymphopenic mice [59]. However, this may be due not only to the absence of microbial antigens, but also to the absence of costimulation provided by the commensal flora in germ-free mice [60]. Still, fast HP is observed in these animals, which highlights the involvement of self-antigens and food antigens in its induction [59],[61]. This indirectly confirms the role of fast HP as a factor that provides oligoclonal expansion of self-reactive T-cells.
As it was mentioned above, HP is a process that is induced by lymphopenia. Such a “traditional” view on this process requires some specifications. Earlier, it was suggested that in lymphopenic conditions, lymphocyte niches got vacant and the competition for the factors of survival reduced, which led to HP of T-cells [50],[62]–[65]. However, the fact that fast HP can be observed without lymphopenia in animals with a normal level of T-lymphocytes can indicate that the number of peripheral T-lymphocytes is not a factor that directly regulates fast HP [61]. Thus, when polyclonal T-lymphocytes are introduced to TCR transgenic mice that have a pool of T-lymphocytes presented by monoclonal TCR repertoire (i.e. all TCR have common specificity) and primarily naïve phenotype, fast HP of the transferred T-lymphocytes is observed [66]. Besides, even in lymphopenia, the presence of the preserved pool of memory-like T-cells prevents fast HP of the transferred naïve T-lymphocytes [67]. Hence, the most important factor of fast HP is not lymphopenia itself but a decrease in the number of memory T-cells. At the same time, the presence of clonal competition confirms the high antigenic specificity of fast HP [66]. Thus, monoclonal T-lymphocytes enter to HP process after the introduction to the recipients with another clonotype and are not divided after the introduction to the recipients with identical clonotype. Besides, the introduction of monoclonal T-lymphocytes to transgenic recipients with the same clonotype that underwent sublethal irradiation did not lead to their proliferation. At the same time, polyclonal T-lymphocytes got involved in fast proliferation in these conditions [66]. Hence, for fast HP of a certain clone of naïve T-lymphocytes, a decrease in the number of memory T-cells of the identical specificity is required. Taking into account that different groups of draining lymphatic nodes have antigens specific to the respective tissues, it would be logical to suggest the presence of tissue-specific or organ-specific homeostatic proliferation that may be mediated by the appearance of specific vacant niches during the elimination of some clone of effector or memory T-cells. In vivo studies demonstrated HP associated with the intestine that was primarily observed in mesenteric lymphatic nodes and led to the appearance of effector and memory-like T-cells [68]. Based on this fact, it could be suggested that in the physiological conditions, fast HP targets the maintenance of the diversity of TCR of memory T-cells [61] and it barely depends on the presence or absence of general lymphopenia. At the same time, slow HP targets the maintenance of the diversity of TCR of naïve T-lymphocytes and memory T-cells and primarily depends on lymphopenia and an increase in the concentration of IL-7 and IL-15.
It should be mentioned that there is another possible role of HP—it can be a component of antitumor immunity. The results of the studies showed that the induction of HP in the lymphatic nodes with the presentation of a tumor antigen caused the favorable autoimmune anti-tumor response. On the periphery, HP with the recognition of self-antigens leads to the development of the autoimmune response to these antigens including tumor ones. Such a response results from polyclonal homeostatic expansion in lymph nodes and is characterized by CD8+ cellular cytotoxicity and an increase in the concentration of IFNγ, as well as the formation of memory T-cells [69]. However, the presence of potentially self-reactive T-lymphocytes also contributes to the development of the pathological character of HP. It should be mentioned in this context that there are data that indicate that the shift in the emphasis of homeostatic proliferation from CD8+ cells to CD4+ lymphocytes can be one of the causes of the development of autoimmune diseases [15].
One of the most important studies that confirm the association between lymphopenia and HP with the development of autoimmune diseases in humans is the study conducted by Jones et al. in 2013 that included 80 patients with multiple sclerosis. All patients received lymphocyte-depleting humanized monoclonal antibody alemtuzumab (monoclonal antibody that binds to CD52), which caused severe lymphopenia in the majority of patients. Within a year and a half, 46% of patients developed autoimmune diseases, and 20% developed asymptomatic carriage of autoantibodies. Patients with a manifestation of autoimmune disease were characterized by a decrease in the migration from the thymic and a narrowing of the TCR repertoire. Thus, the study on humans showed that HP caused by alemtuzumab significantly increased the risk of the development of autoimmune diseases and was accompanied by the generation of activated, highly proliferating effector and memory-like T-cells that had high potential for the production of inflammatory cytokines, which indicated the prevalence of fast HP in these patients [70].
HP is a physiological process that proceeds differently in people. Some factors can increase its negative influence on the organism: genetic predisposition, severity, rate, and duration of lymphopenia, current inflammatory background in target-tissues, as well as the effectiveness of thymopoiesis. Lymphopenia is observed in many patients with autoimmune diseases [34]. For example, in patients with rheumatoid arthritis, the rate of naïve T-cells pool restoration is insufficient, which is shown by a significant decrease in the number of TREC+ lymphocytes in this patients [71]. Such disturbance is observed from the beginning of the disease and does not depend on the stage, which indicates the primary defect in the generation of naïve T-lymphocytes. However, a decrease in the number of naïve TREC+ cells does not lead to the changes in the number of memory T-cells, which indicates an enhancement of homeostatic proliferation in these patients. Apparently, a disturbance of thymopoiesis and the rate and severity of lymphopenia provides a significant impact on the shaping of the pathological nature of HP.
Besides the negative impact on the TCR landscape, HP may lead to telomere attrition and increased numbers of senescent and exhausted T-cells because T-cells undergoing slow HP do not express the high levels of telomerase necessary to prevent replication-driven telomere length erosion [72],[73]. Also, a previous study showed that recurrent HP results in global gene expression changes, including the up-regulation of both cytotoxic and inhibitory molecules that explain how HP could lead to the simultaneous auto-inflammatory and immunodeficiency syndromes frequently observed in lymphopenic patients [74]. Moreover, recurrent HP may cause epigenetic dysregulation as extensive demethylation of genome discrete sites and leads to up-regulation in the expression of corresponding genes [75]. Thus, HP influences the fate of T cells and the body as a whole, has a complex character, and can have both physiological and pathological features.
Tregs are an important factor in the maintenance of the immune equilibrium. It is well-known that these cells provide peripheral auto-tolerance and can suppress different immune-mediated reactions in response to a wide spectrum of physiological and pathological stimuli, including microorganisms, tumor cells, allogeneic grafts, and fetal cells [76],[77]. There are few studies dedicated to HP of Tregs. Still, some of them show that these cells can get involved in fast HP even in the absence of severe lymphopenia [78],[79]. At the same time, there were no studies on the evaluation of the functional activity of such HP-derived Tregs, which provides the rationale for further studies. It should be mentioned that mechanisms of homeostatic maintenance of Treg cells differ from the mechanisms of effector cells that were described in detail in a recent review [77].
Treg cells are the main cells that provide peripheral tolerance. For this reason, the issue of their functional activity arises in the lymphopenia during HP. According to some data, Tregs can suppress HP of effector lymphocytes with the CTLA-4-mediated mechanism [80]–[82]. However, our data (unpublished) and some studies show that Tregs cannot effectively suppress the proliferation of effector CD4+ and CD8+ lymphocytes that receive a strong signal from TCRs in the presence of IL-7 or IL-15 (conditions typical for fast HP) [83],[84]. Thus, during fast HP, when T-cells receive a strong TCR signal and the levels of IL-7 and IL-15 are elevated, the conditions for the acceleration of proliferation of potentially self-reactive T-lymphocytes are created. This provides an additional mechanism of the negative influence of HP on the immune equilibrium and becomes a risk factor for the development of autoimmune diseases.
It should be mentioned that incapability of Treg cells to suppress fast HP of effector cells can be associated with both the direct negative influence of HP conditions on Tregs (deprivation of IL-2 due to a decrease in the number of its producers) and acquisition of the resistance to the suppressor influence of Tregs by the effector cells [83],[84]. However, this issue requires additional studies.
The influence of homeostatic proliferation on the immune system is many-sided. Even though it exerts physiological function, in some cases, this process can acquire pathological features [2],[4],[13],[15],[18]. Earlier, it was demonstrated that various factors influenced the character of HP. The most important of them is decreased thymopoiesis and the rate and severity of lymphopenia [34],[71]. In cases with moderate lymphopenia, humoral factors are sufficient for the maintenance of the TCR diversity and the restoration of the T-cell pool takes place without its qualitative alterations [41],[42]. However, when a certain degree of lymphopenia is reached, the mechanisms of urgent restoration of the peripheral population of lymphocytes get activated that require a higher rate of proliferation and depend on a strong TCR signal [10],[13],[17],[18]. Thus, the degree of lymphopenia influences the intensity of homeostatic processes and may lead to qualitative changes that can negatively influence the general state of the immune system. In this case, negative effects are limited to a clonal expansion of lymphocytes with relatively high affinity TCR [10], the transition of these lymphocytes to memory-like T-cells [3],[18], and a decrease in the activity of Tregs due to IL-2 deprivation [77]. In general, this reflects qualitative-quantitative transition, when the changes in the rate of the physiological process cause functional disturbances in the immune system that result in the development of pathology.
[1] |
Sprent J, Tough DF (1994) Lymphocyte life-span and memory. Science 265: 1395-1400. doi: 10.1126/science.8073282
![]() |
[2] |
Jamieson BD, Douek DC, Killian S, et al. (1999) Generation of functional thymocytes in the human adult. Immunity 10: 569-575. doi: 10.1016/S1074-7613(00)80056-4
![]() |
[3] |
Moxham VF, Karegli J, Phillips RE, et al. (2008) Homeostatic proliferation of lymphocytes results in augmented memory-like function and accelerated allograft rejection. J Immunol 180: 3910-3918. doi: 10.4049/jimmunol.180.6.3910
![]() |
[4] |
den Braber I, Mugwagwa T, Vrisekoop N, et al. (2012) Maintenance of peripheral naive T cells is sustained by thymus output in mice but not humans. Immunity 36: 288-297. doi: 10.1016/j.immuni.2012.02.006
![]() |
[5] |
Kozlov VA (2014) Homeostatic proliferation as a basis for the inevitable formation of total immunodeficiency. Med Immunol (Russia) 16: 403-408. doi: 10.15789/1563-0625-2014-5-403-408
![]() |
[6] |
Dummer W, Ernst B, LeRoy E, et al. (2001) Autologous regulation of naive T cell homeostasis within the T cell compartment. J Immunol 166: 2460-2468. doi: 10.4049/jimmunol.166.4.2460
![]() |
[7] |
Chalasani G, Dai Z, Konieczny BT, et al. (2002) Recall and propagation of allospecific memory T cells independent of secondary lymphoid organs. P Natl Acad Sci USA 99: 6175-6180. doi: 10.1073/pnas.092596999
![]() |
[8] |
Miller CN, Hartigan-O'Connor DJ, Lee MS, et al. (2013) IL-7 production in murine lymphatic endothelial cells and induction in the setting of peripheral lymphopenia. Int Immunol 25: 471-483. doi: 10.1093/intimm/dxt012
![]() |
[9] |
Anthony SM, Rivas SC, Colpitts SL, et al. (2016) Inflammatory signals regulate IL-15 in response to lymphodepletion. J Immunol 196: 4544-4552. doi: 10.4049/jimmunol.1600219
![]() |
[10] |
Ge Q, Rao VP, Cho BK, et al. (2001) Dependence of lymphopenia-induced T cell proliferation on the abundance of peptide/MHC epitopes and strength of their interaction with T cell receptors. Proc Natl Acad Sci USA 98: 1728-1733. doi: 10.1073/pnas.98.4.1728
![]() |
[11] |
Yamaki S, Ine S, Kawabe T, et al. (2014) OX40 and IL-7 play synergistic roles in the homeostatic proliferation of effector memory CD4+ T cells. Eur J Immunol 44: 3015-3025. doi: 10.1002/eji.201444701
![]() |
[12] |
Li O, Zheng P, Liu Y (2004) CD24 expression on T cells is required for optimal T cell proliferation in lymphopenic host. J Exp Med 200: 1083-1089. doi: 10.1084/jem.20040779
![]() |
[13] |
Min B, Yamane H, Hu-Li J, et al. (2005) Spontaneous and homeostatic proliferation of CD4 T cells are regulated by different mechanisms. J Immunol 174: 6039-6044. doi: 10.4049/jimmunol.174.10.6039
![]() |
[14] |
Ge Q, Rao VP, Cho BK, et al. (2001) Dependence of lymphopenia-induced T cell proliferation on the abundance of peptide/MHC epitopes and strength of their interaction with T cell receptors. Proc Natl Acad Sci USA 98: 1728-1733. doi: 10.1073/pnas.98.4.1728
![]() |
[15] |
Theofilopoulos AN, Dummer W, Kono DH (2001) T cell homeostasis and systemic autoimmunity. J Clin Invest 108: 335-340. doi: 10.1172/JCI200112173
![]() |
[16] |
Kassiotis G, Zamoyska R, Stockinger B (2003) Involvement of avidity for major histocompatibility complex in homeostasis of naive and memory T cells. J Exp Med 197: 1007-1016. doi: 10.1084/jem.20021812
![]() |
[17] |
Kieper WC, Burghardt JT, Surh CD (2004) A role for TCR affinity in regulating naive T cell homeostasis. J Immunol 172: 40-44. doi: 10.4049/jimmunol.172.1.40
![]() |
[18] |
Goldrath AW, Bogatzki LY, Bevan MJ (2000) Naive T cells transiently acquire a memory-like phenotype during homeostasis-driven proliferation. J Exp Med 192: 557-564. doi: 10.1084/jem.192.4.557
![]() |
[19] |
Klein L, Kyewski B, Allen PM, et al. (2014) Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see). Nat Rev Immunol 14: 377-391. doi: 10.1038/nri3667
![]() |
[20] |
Dolton G, Zervoudi E, Rius C, et al. (2018) Optimized peptide-MHC multimer protocols for detection and isolation of autoimmune T-Cells. Front Immunol 9: 1378. doi: 10.3389/fimmu.2018.01378
![]() |
[21] |
Enouz S, Carrié L, Merkler D, et al. (2012) Autoreactive T cells bypass negative selection and respond to self-antigen stimulation during infection. J Exp Med 209: 1769-1779. doi: 10.1084/jem.20120905
![]() |
[22] |
Yan J, Mamula MJ (2002) Autoreactive T cells revealed in the normal repertoire: escape from negative selection and peripheral tolerance. J Immunol 168: 3188-3194. doi: 10.4049/jimmunol.168.7.3188
![]() |
[23] |
Boehncke WH, Brembilla NC (2019) Autoreactive T-lymphocytes in inflammatory skin diseases. Front Immunol 10: 1198. doi: 10.3389/fimmu.2019.01198
![]() |
[24] |
Koehli S, Naeher D, Galati-Fournier V, et al. (2014) Optimal T-cell receptor affinity for inducing autoimmunity. Proc Natl Acad Sci USA 111: 17248-17253. doi: 10.1073/pnas.1402724111
![]() |
[25] |
Mueller DL (2010) Mechanisms maintaining peripheral tolerance. Nat Immunol 11: 21-27. doi: 10.1038/ni.1817
![]() |
[26] |
Vrisekoop N, Monteiro JP, Mandl JN, et al. (2014) Revisiting thymic positive selection and the mature T cell repertoire for antigen. Immunity 41: 181-190. doi: 10.1016/j.immuni.2014.07.007
![]() |
[27] |
Qi Q, Liu Y, Cheng Y, et al. (2014) Diversity and clonal selection in the human T-cell repertoire. Proc Natl Acad Sci USA 111: 13139-13144. doi: 10.1073/pnas.1409155111
![]() |
[28] |
Zheng SG, Wang J, Wang P, et al. (2007) IL-2 is essential for TGF-beta to convert naive CD4+CD25− cells to CD25+Foxp3+ regulatory T cells and for expansion of these cells. J Immunol 178: 2018-2027. doi: 10.4049/jimmunol.178.4.2018
![]() |
[29] |
Pacholczyk R, Ignatowicz H, Kraj P, et al. (2006) Origin and T cell receptor diversity of Foxp3+CD4+CD25+ T cells. Immunity 25: 249-259. doi: 10.1016/j.immuni.2006.05.016
![]() |
[30] |
Wyss L, Stadinski BD, King CG, et al. (2016) Affinity for self-antigen selects Treg cells with distinct functional properties. Nat Immunol 17: 1093-1101. doi: 10.1038/ni.3522
![]() |
[31] |
Bolotin DA, Poslavsky S, Davydov AN, et al. (2017) Antigen receptor repertoire profiling from RNA-seq data. Nat Biotechnol 35: 908-911. doi: 10.1038/nbt.3979
![]() |
[32] |
Jörg S, Grohme DA, Erzler M, et al. (2016) Environmental factors in autoimmune diseases and their role in multiple sclerosis. Cell Mol Life Sci 73: 4611-4622. doi: 10.1007/s00018-016-2311-1
![]() |
[33] |
Ting YT, Petersen J, Ramarathinam SH, et al. (2018) The interplay between citrullination and HLA-DRB1 polymorphism in shaping peptide binding hierarchies in rheumatoid arthritis. J Biol Chem 293: 3236-3251. doi: 10.1074/jbc.RA117.001013
![]() |
[34] |
Schulze-Koops H (2004) Lymphopenia and autoimmune diseases. Arthritis Res Ther 6: 178-180. doi: 10.1186/ar1208
![]() |
[35] |
Symmons DP, Farr M, Salmon M, et al. (1989) Lymphopenia in rheumatoid arthritis. J R Soc Med 82: 462-463. doi: 10.1177/014107688908200806
![]() |
[36] |
Jones JL, Thompson SA, Loh P, et al. (2013) Human autoimmunity after lymphocyte depletion is caused by homeostatic T-cell proliferation. Proc Natl Acad Sci USA 110: 20200-20205. doi: 10.1073/pnas.1313654110
![]() |
[37] |
Koetz K, Bryl E, Spickschen K, et al. (2000) T cell homeostasis in patients with rheumatoid arthritis. Proc Natl Acad Sci USA 97: 9203-9208. doi: 10.1073/pnas.97.16.9203
![]() |
[38] |
Virot E, Duclos A, Adelaide L, et al. (2017) Autoimmune diseases and HIV infection: A cross-sectional study. Medicine (Baltimore) 96: e5769. doi: 10.1097/MD.0000000000005769
![]() |
[39] | Jerne NK (1974) Towards a network theory of the immune system. Ann Immunol (Paris) 125: 373-389. |
[40] |
Ernst B, Lee DS, Chang JM, et al. (1999) The peptide ligands mediating positive selection in the thymus control T cell survival and homeostatic proliferation in the periphery. Immunity 11: 173-181. doi: 10.1016/S1074-7613(00)80092-8
![]() |
[41] |
Milam AAV, Bartleson JM, Buck MD, et al. (2020) Tonic TCR signaling inversely regulates the basal metabolism of CD4+ T cells. ImmunoHorizons 4: 485-497. doi: 10.4049/immunohorizons.2000055
![]() |
[42] |
Hochweller K, Wabnitz GH, Samstag Y, et al. (2010) Dendritic cells control T cell tonic signaling required for responsiveness to foreign antigen. Proc Natl Acad Sci USA 107: 5931-5936. doi: 10.1073/pnas.0911877107
![]() |
[43] |
Stefanová I, Dorfman JR, Germain RN (2002) Self-recognition promotes the foreign antigen sensitivity of naive T lymphocytes. Nature 420: 429-434. doi: 10.1038/nature01146
![]() |
[44] |
Miller CN, Hartigan-O'Connor DJ, Lee MS, et al. (2013) IL-7 production in murine lymphatic endothelial cells and induction in the setting of peripheral lymphopenia. Int Immunol 25: 471-483. doi: 10.1093/intimm/dxt012
![]() |
[45] |
Saeed S, Revell PA (2001) Production and distribution of interleukin 15 and its receptors (IL-15Ralpha and IL-R2beta) in the implant interface tissues obtained during revision of failed total joint replacement. Int J Exp Pathol 82: 201-209. doi: 10.1111/j.1365-2613.2001.iep185.x
![]() |
[46] |
Regamey N, Obregon C, Ferrari-Lacraz S, et al. (2007) Airway epithelial IL-15 transforms monocytes into dendritic cells. Am J Respir Cell Mol Biol 37: 75-84. doi: 10.1165/rcmb.2006-0235OC
![]() |
[47] |
Park CS, Yoon SO, Armitage RJ, et al. (2004) Follicular dendritic cells produce IL-15 that enhances germinal center B cell proliferation in membrane-bound form. J Immunol 173: 6676-6683. doi: 10.4049/jimmunol.173.11.6676
![]() |
[48] |
Niu N, Qin X (2013) New insights into IL-7 signaling pathways during early and late T cell development. Cell Mol Immunol 10: 187-189. doi: 10.1038/cmi.2013.11
![]() |
[49] |
Mishra A, Sullivan L, Caligiuri MA (2014) Molecular pathways: interleukin-15 signaling in health and in cancer. Clin Cancer Res 20: 2044-2050. doi: 10.1158/1078-0432.CCR-12-3603
![]() |
[50] |
Tan JT, Ernst B, Kieper WC, et al. (2002) Interleukin (IL)-15 and IL-7 jointly regulate homeostatic proliferation of memory phenotype CD8+ cells but are not required for memory phenotype CD4+ cells. J Exp Med 195: 1523-1532. doi: 10.1084/jem.20020066
![]() |
[51] |
Do JS, Visperas A, Oh K, et al. (2012) Memory CD4 T cells induce selective expression of IL-27 in CD8+ dendritic cells and regulate homeostatic naive T cell proliferation. J Immunol 188: 230-237. doi: 10.4049/jimmunol.1101908
![]() |
[52] |
Lenz DC, Kurz SK, Lemmens E, et al. (2004) IL-7 regulates basal homeostatic proliferation of antiviral CD4+ T cell memory. Proc Natl Acad Sci USA 101: 9357-9362. doi: 10.1073/pnas.0400640101
![]() |
[53] |
Berard M, Brandt K, Bulfone-Paus S, et al. (2003) IL-15 promotes the survival of naive and memory phenotype CD8+ T cells. J Immunol 170: 5018-5026. doi: 10.4049/jimmunol.170.10.5018
![]() |
[54] |
Prlic M, Blazar BR, Khoruts A, et al. (2001) Homeostatic expansion occurs independently of costimulatory signals. J Immunol 167: 5664-5668. doi: 10.4049/jimmunol.167.10.5664
![]() |
[55] |
Do JS, Min B (2009) Differential requirements of MHC and of DCs for endogenous proliferation of different T-cell subsets in vivo. Proc Natl Acad Sci USA 106: 20394-20398. doi: 10.1073/pnas.0909954106
![]() |
[56] |
Hawse WF, Cattley RT (2019) T cells transduce T-cell receptor signal strength by generating different phosphatidylinositols. J Biol Chem 294: 4793-4805. doi: 10.1074/jbc.RA118.006524
![]() |
[57] |
Cosway EJ, Lucas B, James KD, et al. (2017) Redefining thymus medulla specialization for central tolerance. J Exp Med 214: 3183-3195. doi: 10.1084/jem.20171000
![]() |
[58] |
Logunova NN, Kriukova VV, Shelyakin PV, et al. (2020) MHC-II alleles shape the CDR3 repertoires of conventional and regulatory naïve CD4+ T cells. Proc Natl Acad Sci USA 117: 13659-13669. doi: 10.1073/pnas.2003170117
![]() |
[59] |
Kieper WC, Troy A, Burghardt JT, et al. (2005) Recent immune status determines the source of antigens that drive homeostatic T cell expansion. J Immunol 174: 3158-3163. doi: 10.4049/jimmunol.174.6.3158
![]() |
[60] |
Do JS, Foucras G, Kamada N, et al. (2012) Both exogenous commensal and endogenous self antigens stimulate T cell proliferation under lymphopenic conditions. Cell Immunol 272: 117-123. doi: 10.1016/j.cellimm.2011.11.002
![]() |
[61] |
Min B (2018) Spontaneous T cell proliferation: A physiologic process to create and maintain homeostatic balance and diversity of the immune system. Front Immunol 9: 547. doi: 10.3389/fimmu.2018.00547
![]() |
[62] |
Schluns KS, Kieper WC, Jameson SC, et al. (2000) Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo. Nat Immunol 1: 426-432. doi: 10.1038/80868
![]() |
[63] |
Ku CC, Murakami M, Sakamoto A, et al. (2000) Control of homeostasis of CD8+ memory T cells by opposing cytokines. Science 288: 675-678. doi: 10.1126/science.288.5466.675
![]() |
[64] |
Tan JT, Dudl E, LeRoy E, et al. (2001) IL-7 is critical for homeostatic proliferation and survival of naive T cells. Proc Natl Acad Sci USA 98: 8732-8737. doi: 10.1073/pnas.161126098
![]() |
[65] |
Goldrath AW, Sivakumar PV, Glaccum M, et al. (2002) Cytokine requirements for acute and Basal homeostatic proliferation of naive and memory CD8+ T cells. J Exp Med 195: 1515-1522. doi: 10.1084/jem.20020033
![]() |
[66] |
Troy AE, Shen H (2003) Cutting edge: homeostatic proliferation of peripheral T lymphocytes is regulated by clonal competition. J Immunol 170: 672-676. doi: 10.4049/jimmunol.170.2.672
![]() |
[67] |
Min B, Foucras G, Meier-Schellersheim M, et al. (2004) Spontaneous proliferation, a response of naive CD4 T cells determined by the diversity of the memory cell repertoire. Proc Natl Acad Sci USA 101: 3874-3879. doi: 10.1073/pnas.0400606101
![]() |
[68] |
Kawabe T, Sun SL, Fujita T, et al. (2013) Homeostatic proliferation of naive CD4+ T cells in mesenteric lymph nodes generates gut-tropic Th17 cells. J Immunol 190: 5788-5798. doi: 10.4049/jimmunol.1203111
![]() |
[69] |
Dummer W, Niethammer AG, Baccala R, et al. (2002) T cell homeostatic proliferation elicits effective antitumor autoimmunity. J Clin Invest 110: 185-192. doi: 10.1172/JCI0215175
![]() |
[70] |
Jones JL, Thompson SA, Loh P, et al. (2013) Human autoimmunity after lymphocyte depletion is caused by homeostatic T-cell proliferation. Proc Natl Acad Sci USA 110: 20200-20205. doi: 10.1073/pnas.1313654110
![]() |
[71] |
Koetz K, Bryl E, Spickschen K, et al. (2000) T cell homeostasis in patients with rheumatoid arthritis. Proc Natl Acad Sci USA 97: 9203-9208. doi: 10.1073/pnas.97.16.9203
![]() |
[72] |
Arruda LCM, Lima-Júnior JR, Clave E, et al. (2018) Homeostatic proliferation leads to telomere attrition and increased PD-1 expression after autologous hematopoietic SCT for systemic sclerosis. Bone Marrow Transpl 53: 1319-1327. doi: 10.1038/s41409-018-0162-0
![]() |
[73] |
O'Bryan JM, Woda M, Co M, et al. (2013) Telomere length dynamics in human memory T cells specific for viruses causing acute or latent infections. Immun Ageing 10: 37. doi: 10.1186/1742-4933-10-37
![]() |
[74] |
Fortner KA, Bond JP, Austin JW, et al. (2017) The molecular signature of murine T cell homeostatic proliferation reveals both inflammatory and immune inhibition patterns. J Autoimmun 82: 47-61. doi: 10.1016/j.jaut.2017.05.003
![]() |
[75] |
Scharer CD, Fortner KA, Dragon JA, et al. (2020) Selective DNA demethylation accompanies T cell homeostatic proliferation and gene regulation in lupus-prone lpr mice. ImmunoHorizons 4: 679-687. doi: 10.4049/immunohorizons.2000078
![]() |
[76] |
Sakaguchi S, Yamaguchi T, Nomura T, et al. (2008) Regulatory T cells and immune tolerance. Cell 133: 775-787. doi: 10.1016/j.cell.2008.05.009
![]() |
[77] |
Shevyrev D, Tereshchenko V (2020) Treg heterogeneity, function, and homeostasis. Front Immunol 10: 3100. doi: 10.3389/fimmu.2019.03100
![]() |
[78] |
Tang AL, Teijaro JR, Njau MN, et al. (2008) CTLA4 expression is an indicator and regulator of steady-state CD4+ FoxP3+ T cell homeostasis. J Immunol 181: 1806-1813. doi: 10.4049/jimmunol.181.3.1806
![]() |
[79] |
Chen Y, Shen S, Gorentla BK, et al. (2012) Murine regulatory T cells contain hyperproliferative and death-prone subsets with differential ICOS expression. J Immunol 188: 1698-1707. doi: 10.4049/jimmunol.1102448
![]() |
[80] |
Sojka DK, Hughson A, Fowell DJ (2009) CTLA-4 is required by CD4+CD25+ Treg to control CD4+ T-cell lymphopenia-induced proliferation. Eur J Immunol 39: 1544-1551. doi: 10.1002/eji.200838603
![]() |
[81] |
Bolton HA, Zhu E, Terry AM, et al. (2015) Selective Treg reconstitution during lymphopenia normalizes DC costimulation and prevents graft-versus-host disease. J Clin Invest 125: 3627-3641. doi: 10.1172/JCI76031
![]() |
[82] |
Winstead CJ, Fraser JM, Khoruts A (2008) Regulatory CD4+CD25+Foxp3+ T cells selectively inhibit the spontaneous form of lymphopenia-induced proliferation of naive T cells. J Immunol 180: 7305-7317. doi: 10.4049/jimmunol.180.11.7305
![]() |
[83] |
Ben Ahmed M, Belhadj Hmida N, Moes N, et al. (2009) IL-15 renders conventional lymphocytes resistant to suppressive functions of regulatory T cells through activation of the phosphatidylinositol 3-kinase pathway. J Immunol 182: 6763-6770. doi: 10.4049/jimmunol.0801792
![]() |
[84] |
Heninger AK, Theil A, Wilhelm C, et al. (2012) IL-7 abrogates suppressive activity of human CD4+CD25+FOXP3+ regulatory T cells and allows expansion of alloreactive and autoreactive T cells. J Immunol 189: 5649-5658. doi: 10.4049/jimmunol.1201286
![]() |
1. | Daniil Shevyrev, Valeriy Tereshchenko, Elena Blinova, Nadezda Knauer, Ekaterina Pashkina, Alexey Sizikov, Vladimir Kozlov, Regulatory T Cells Fail to Suppress Fast Homeostatic Proliferation In Vitro, 2021, 11, 2075-1729, 245, 10.3390/life11030245 | |
2. | Daniil Shevyrev, Valeriy Tereshchenko, Vladimir Kozlov, Immune Equilibrium Depends on the Interaction Between Recognition and Presentation Landscapes, 2021, 12, 1664-3224, 10.3389/fimmu.2021.706136 | |
3. | Daniil Shevyrev, Valeriy Tereshchenko, Vladimir Kozlov, Sergey Sennikov, Phylogeny, Structure, Functions, and Role of AIRE in the Formation of T-Cell Subsets, 2022, 11, 2073-4409, 194, 10.3390/cells11020194 | |
4. | D. V. Shevyrev, V. P. Tereshchenko, S. V. Sennikov, The Enigmatic Nature of the TCR-pMHC Interaction: Implications for CAR-T and TCR-T Engineering, 2022, 23, 1422-0067, 14728, 10.3390/ijms232314728 | |
5. | Marianna Y. Kawashima-Vasconcelos, Maynara Santana-Gonçalves, Djúlio C. Zanin-Silva, Kelen C. R. Malmegrim, Maria Carolina Oliveira, Reconstitution of the immune system and clinical correlates after stem cell transplantation for systemic sclerosis, 2022, 13, 1664-3224, 10.3389/fimmu.2022.941011 | |
6. | Mie Kobayashi-Ishihara, Katarína Frazão Smutná, Florencia E. Alonso, Jordi Argilaguet, Anna Esteve-Codina, Kerstin Geiger, Meritxell Genescà, Judith Grau-Expósito, Clara Duran-Castells, Selina Rogenmoser, René Böttcher, Jennifer Jungfleisch, Baldomero Oliva, Javier P. Martinez, Manqing Li, Michael David, Makoto Yamagishi, Marta Ruiz-Riol, Christian Brander, Yasuko Tsunetsugu-Yokota, Maria J. Buzon, Juana Díez, Andreas Meyerhans, Schlafen 12 restricts HIV-1 latency reversal by a codon-usage dependent post-transcriptional block in CD4+ T cells, 2023, 6, 2399-3642, 10.1038/s42003-023-04841-y |