Abstract
Purpose of Review
The etiology of autoimmune diseases remains incompletely understood. Here, we highlight recent advances in the role of pro-inflammatory metabolic pathways in autoimmune disease, including treatment with antioxidants and mTOR inhibitors.
Recent Findings
Recent studies show that mTOR pathway activation, glucose utilization, mitochondrial oxidative phosphorylation, and antioxidant defenses play critical roles in the pathogenesis of autoimmune diseases, including rheumatoid arthritis, immune thrombocytopenia, Sjögren’s syndrome, large vessel vasculitis, and systemic lupus erythematosus. mTOR activity leads to Th1 and Th17 cell proliferation, Treg depletion, plasma cell differentiation, macrophage dysfunction, and increased antibody and immune complex production, ultimately resulting in tissue inflammation. mTOR also affects the function of connective tissue cells, including fibroblast-like synoviocytes, endothelial cells, and podocytes. mTOR inhibition via rapamycin and N-acetylcysteine, and blockade of glucose utilization show clinical efficacy in both mouse models and clinical trials, such as systemic lupus erythematosus.
Summary:
The mTOR pathway is a central regulator of growth and survival signals, integrating environmental cues to control cell proliferation and differentiation. Activation of mTOR underlies inflammatory lineage specification, and mTOR blockade-based therapies show promising efficacy in several autoimmune diseases.
Keywords: mTOR, autoimmunity, metabolism, mitochondrial oxidative stress, rapamycin, antioxidant therapy
Introduction
The ability to control the immune system promises the secret of health and longevity. As a complex web of cytokine signaling, inter-cell communication, and ubiquitous existence in almost every tissue of the body, the immune system may just be the critical mediator of host physiology and pathology. Increasingly, the role of the mechanistic Target of Rapamycin (mTOR) pathway has been identified as a critical mediator of immune function.
Discovered after an expedition to the island of Rapa Nui (Easter Island), rapamycin (known clinically as sirolimus) was isolated from soil Streptomyces hygroscopicus. Rapamycin was noted for its many properties, including antifungal and immunosuppressive effects(1, 2). These effects were found to be due to rapamycin forming a complex with FKBP12 and exerting its function on the aptly named mechanistic Target of Rapamycin (mTOR)(3–5). mTOR is a serine/threonine kinase that acts as a central regulator of growth and metabolic signals, integrating environmental inputs and controlling cellular machinery(6).
mTOR exists within two separate complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) (Figure 1A). mTORC1 functions primarily to stimulate cell growth downstream of growth factors, nutrients, and energy. mTORC1 accomplishes growth promotion by upregulating mRNA transcription, protein synthesis, and metabolic changes that include lipid and nucleotide synthesis(6). mTORC2 signaling is less characterized but promotes cell survival and proliferation downstream of certain growth factors. mTORC2-regulated pathways include apoptosis, glucose metabolism, cell migration, cytoskeleton arrangement, and ion transport to ensure survival of the cell (Figure 1A)(6).
Figure 1.
mTOR activity contributes to autoimmune disease. A) mTORC1 and mTORC2 are distinct complexes that integrate environmental cues to shape cellular phenotype. mTORC1 senses a variety of signals including amino acid sufficiency, glucose sufficiency, cytokines, growth hormones, and cellular energy, and controls mRNA transcription, protein, nucleotide, and lipid synthesis, and metabolic changes within the cell. mTORC2 signaling is less characterized, but activation occurs downstream of the PI3K/AKT pathway. mTORC2 has roles primarily in cytoskeletal reorganization, although it also has roles in apoptosis, ion transport, and glucose metabolism, among others. B) The mTOR pathway is implicated in a variety of autoimmune diseases via controlling cellular development. mTOR activity skews immune cells towards a pro-inflammatory phenotype, as well promoting proliferation in other cell lineages, ultimately promoting disease pathogenesis. TKR = Tyrosine kinase receptor. ROS = Reactive oxidative species. RAPA = Rapamycin. PPP = Pentose phosphate pathway. DC = Dendritic cell. M0 = Macrophage. FLS = Fibroblast-like synoviocyte. MKC = Megakaryocyte. EC = Endothelial cell. SLE = Systemic lupus erythematosus. APS = Antiphospholipid syndrome. PsA = Psoriatic arthritis. AS = Ankylosing spondylitis. RA = Rheumatoid arthritis. ITP = Immune thrombocytopenia.
The immunosuppressive effects of rapamycin led to its approval by the FDA in 1999 for anti-rejection therapy after kidney transplantation. Similarly, rapamycin showed promising results in alleviating inflammatory autoimmune diseases, including mouse models of lupus and multiple sclerosis(7–9). Due to its successful application in such therapies, mTOR signaling in the immune system has become of great interest. mTOR regulates many facets of immune cell development, including control over whether T cells become regulatory- or effector-type cells(10–13). In particular, dysregulation of mTOR, along with altered immune cell characteristics, prominently appears in the initiation and progression of autoimmune diseases (Figure 1B and Table 1) (14). This dysregulation has been identified as a central biomarker of disease and target of treatment, and in-depth understanding of the mTOR pathway may unlock powerful treatment options in a variety of diseases(14). Here, current advances in the understanding of the mTOR pathway in select diseases, as well as therapeutic targeting of mTOR, will be reviewed.
Table 1.
Pharmacological blockade of the mTOR pathway in autoimmune diseases. mTOR blockade has shown to be beneficial in autoimmune diseases by inhibiting mTOR activity, thereby downregulating inflammation and stabilizing disease activity.
Disease | Organ | Target cell/Antigen | Effector | mTOR blockade |
---|---|---|---|---|
SLE | Systemic | Podocyte/Kidney Neuron/Brain Hepatocyte/Liver Endothelium/Vessel Epithelium/Lung/Pleura/Pericardium |
DN/Th17 T cells Treg/EMT contraction B cells Immune complexes |
Rapa(52, 57, 65) NAC(66), Metformin(67) |
APS | Systemic | Vascular endothelium | T cells B cell Macrophages DCs Hepatocytes |
Rapa(62, 63, 65) |
GN | Kidney | Podocytes | T cells B cells Macrophages DCs |
Rapa(60) |
RA | Joint Synovium | Chondrocytes | FLS T cells B cells Macrophages DCs |
Everolimus(22), NAC(68, 69) |
OA | Joint | Chondrocytes | Chondrocytes | Rapa(70, 71) |
PsA | Skin, Joint | Keratinocytes, Chondrocytes | T cells B cells Macrophages DCs |
Rapa(72) |
SSc | Systemic | Fibroblasts | T cells B cells Macrophages DCs |
Rapa(73, 74) |
PHTN | Lung | Vascular endothelium, Smooth muscle cells | T cells B cells Macrophages DCs |
Everolimus(75) |
Vasculitis | Blood Vessels | Endothelial cells | Endothelial cells T cells Macrophages |
Rapa(47, 48) |
SjS | Exocrine Glands | Glandular epithelium | T cells B cells DCs | Rapa(44), NAC(76) |
ITP | Blood vessels | Megakaryocytes/Platelets | T cells B cells Macrophages DCs |
Rapa(33, 34) |
AIHA | Bone Marrow | RBCs | T cells B cells |
Rapa(77) |
SLE = Systemic lupus erythematosus. APS = Antiphospholipid syndrome. GN = Glomerulonephritis. RA = Rheumatoid arthritis. OA = Osteoarthritis. PsA = Psoriatic arthritis. SSc = Systemic sclerosis. PHTN = Pulmonary hypertension. SjS = Sjögren’s syndrome. ITP = Immune thrombocytopenia. AIHA = Autoimmune hemolytic anemia. RBC = Red blood cells. EMT = Effector memory T cell. DCs = Dendritic cells. FLS = Fibroblast-like synoviocytes. Rapa = Rapamycin/Sirolimus. NAC = N-acetylcysteine.
mTOR controls immune cell development
mTOR has a profound role in T cell lineage development. Part of the adaptive immune system, T cells are critical to raising host defenses and maintaining immunocompetence. T cells participate in both upregulating and downregulating the immune response by differentiating into effector T cells and regulatory T cells (Tregs), respectively. T effector cells can activate other members of the immune system, while Tregs inhibit cytokine production and cell proliferation. mTOR activation is critical for the development of effector T cells, as mTOR activity leads to STAT activation and transcription of genes, such as STAT3-mediated induction of IL-17 and IFN-γ(15). T cell activation and differentiation into effector subtypes require promotion of glycolysis and flux through the PPP, both of which are regulated by mTOR(16*). Specifically, proinflammatory Th1 and Th17 cell differentiation require activation of mTORC1, while Th2 cell differentiation requires mTORC2(15). mTORC1 induction of Th17 differentiation is mediated by mTORC1 activation of S6K1 and a related kinase, S6K2(17). S6K2 activation targets nuclear translocation of RORγ, a transcription factor required for Th17 differentiation, while S6K1 inhibits Gfi1, a negative regulator of Th17 differentiation(17).
Rapamycin blocks the differentiation of Th17 cells, underscoring the necessity of mTORC1 signaling in the development of proinflammatory cell lineages(17). Likewise, inhibition or depletion of mTORC1 and mTORC2 together lead to an expansion of regulatory T cells (Tregs)(15). This dual expansion of Tregs and contraction of T effector cells highlight an important mechanism behind mTOR’s role in promoting the inflammatory process, as metabolic shifts mediated by mTOR activity direct the lineage development of T cells(16*).
Aside from T cells, mTOR also controls the fate of other immune cells. B cell proliferation and survival occurs via the mTOR pathway, as rapamycin has shown the ability to attenuate B cell activating factor (BAFF) stimulation(18, 19*). In macrophages, rapamycin prevents the development of M2 macrophages (canonically anti-inflammatory), without having such an effect on M1 macrophages. mTOR activity appears crucial for M2 macrophage development(20). mTOR is critical in the development of multiple immune cell lineages. In turn, aberrations in mTOR signaling can change the immune profile of the body, leading to pathological autoimmunity.
Rheumatoid Arthritis
Joint destruction in rheumatoid arthritis (RA) is seen as the end result of synovitis mediated by cytokine and autoantibody production, as well as cartilage damage mediated by intra-articular cells(21). Previous research has shown efficacy of mTOR blockade in reducing joint inflammation in RA(22). This effect is seen despite any significant changes in T cell mTOR activity within RA patients(23). However, RA does exhibit increased mTORC1 activity in fibroblast-like synoviocytes (FLS) and osteoclasts, with inhibition of mTOR showing promise in targeting these intra-articular cells(24). Recent research has highlighted the role of mTOR in TNF-mediated pro-inflammatory functions(25*).
In RA-FLSs, TNF is shown to activate mTOR via AKT. In turn, mTOR antagonizes nF-κB (via IκB-α), while also promoting transcription of interferon-regulated genes through STAT1 activation(25*). mTOR’s inhibition of nF-κB may point to why use of rapalogues increases inflammatory markers in RA(22). However, overall this novel pathway shows mTOR’s environmental sensing ability can be coupled to pro-inflammatory cytokine and chemokine production, and therapeutic targeting of mTOR in this pathway may be an efficacious strategy in RA treatment.
The JAK/STAT pathway may also be a target for RA treatment. Signal transducer and activator of transcription 3 (STAT3) has been identified in mouse models as a critical mediator of rheumatoid arthritis and potential therapeutic target(26, 27). Since mTOR and STAT3 have complex crosstalk between the two(15, 28), targeted inhibition of the JAK/STAT pathway as well as the mTOR pathway are potentially valid approaches to therapy.
mTOR and thrombocytopenia
Immune thrombocytopenia (ITP) is a common autoimmune-mediated platelet destruction syndrome thought to be due to abnormal T cell function(29). ITP is marked by an increase in proinflammatory Th17 cells and a decrease in Tregs, similar to other autoimmune diseases, covered here, suspected to be under regulation of mTOR(30). However, how mTOR regulates T cell function specifically in the context of ITP remains unknown. One study has shown that indirubin, a drug with potentially therapeutic effect in ITP murine models, ameliorates disease via increased expression of PD1 and PTEN, leading to the suppression of mTOR in CD4+ T cells(31),(32). This work highlights a potential role for mTOR targeting. Indeed, inhibition of mTOR has been used clinically to positive effects for ITP. Rapamycin, when used in combination with the corticosteroid prednisone, appears to have promising potential in ITP therapy(33). Combination therapy exhibited low toxicity and was shown to lead to a sustained increase in platelets along with an increase in Treg levels(33). More recent reporting showed that sirolimus alone, instead of in combination with prednisone, can also be effective for ITP(34). Sirolimus alone achieved complete response in many cases, making it an attractive option for sparing steroid use and limiting toxicity of ITP therapy(34).
Interestingly, aside from T cell abnormalities, recent work highlights the role of the mTOR pathway and autophagy within megakaryocytes in ITP(35*). Not only does ITP involve autoimmune destruction of platelets, but also dysfunctional platelet production by megakaryocytes(36). Proliferation of megakaryocytes and proper platelet production is thought to be under the control of autophagy pathways(37, 38). However, while these catabolic signals are crucial for maintaining megakaryopoiesis and highlight the potential benefit of autophagy-stimulating rapamycin treatment, it appears, paradoxically, that mTOR signaling is also required for platelet production and that induction of autophagy may actually be a pathogenic factor of ITP(39–41). While rapamycin appears to be beneficial in ameliorating the immune response and clinical disease, its effect on megakaryocyte function, helpful or harmful, requires further research. Overall, targeting autophagy with rapamycin in ITP may be a promising therapeutic pathway to reduce inflammatory destruction of platelets(42).
Sjögren’s Syndrome
Sjögren’s syndrome (SjS) is defined by autoimmune destruction of exocrine glands, as well inflammatory damage to multiple organ systems(43). Previous research has shown that mTOR is implicated in the pathogenesis of SjS, as rapamycin was an effective therapy in a mouse model (44). This effect was due to inhibition of mTOR-mediated T cell activation(44). mTOR may also play a role in human SjS, raising the potential of mTOR-based therapy in this disease(45*). Using peripheral blood mononuclear cells (PBMC) and salivary gland tissue. Interestingly, circulating B cells appear to have decreased expression of mTOR, while B cells within the salivary glands show increased mTOR expression. Plasma cells and T cells within the salivary gland also showed mTOR activation(45*).
Vasculitis
Takayasu arteritis (TA) and giant cell arteritis (GCA) are two large vessel vasculitides whose etiology remains elusive(46). However, the mTOR pathway increasingly appears to play a significant part in the pathogenesis of disease(47*, 48*). Within endothelial cells of TA patients, both mTORC1 and mTORC2 are shown to be activated(47*, 48*). mTORC1 activity in GCA is contrasted by recent work, as it may not be activated in GCA endothelial cells(47*), or possibly have an intermediate activity between that of TA patients and healthy controls(48*).
Further, mTORC1 potentially drives T cell inflammation in large vessel vasculitis, as rapamycin treatment increases Treg and decreases effector T cell populations from cultured patient PBMCs(48*). Interestingly, patient sera and purified IgG is shown to activate mTORC1 and induce endothelial cell proliferation, potentially by targeting a 60–65 kDa antigen on endothelial cells(47*, 48*). Identification of this antigen, as well as further research into the mTOR pathway in vasculitides, is a promising avenue of therapy.
Systemic Lupus Erythematosus
Systemic Lupus Erythematosus (SLE) is a chronic inflammatory disease with potentially life-threatening consequences(49). Multi-organ involvement, variable clinical course, and incompletely understood pathogenic mechanisms create a challenge for pharmacological management(50). However, abnormal activation of T cells appears central to SLE pathogenesis, and the role of mTOR in mediating these effects is critical. mTOR activation follows T cell receptor (TCR) signal transduction and controls increased calcium fluxing in SLE, while rapamycin normalizes this signaling defect(51–53*),. mTORC1 senses a variety of stress signals including the accumulation of amino acids during autophagy, such as leucine and isoleucine, kynurenine, and glutamine(24). Glutaminolysis may enhance mitochondrial oxidative stress and mTORC1 activation in Th17 cells (54*) (2). Deficiency of mRNA splicing also promotes activation of mTORC1 (55*).Further, mTOR activation in SLE T cells leads to the expansion of IL-4 producing double-negative T cells, expansion of Th17 cells, and contraction of Tregs, which contribute to the proinflammatory profile of SLE(56), (57). Other immune cells also contribute to SLE pathogenesis. B cells are antibody-secreting cells responsible for autoantibody production in SLE. Evidence indicates that autoimmune B cells are dependent on mTOR signaling (58). SLC15A4, a lysosomal histidine transporter, is necessary for B cell production of autoantibodies and type I interferon downstream of Toll-Like Receptor 7 (a sensor of single-stranded RNA) stimulation(58). This pathway is mediated by mTORC1, as loss of SLC15A4, as well as direct inhibition of mTOR, lead to disruption of mTORC1 signaling and ablation of autoantibody production(58).
Paradoxically, mTOR appears to mediate an anti-inflammatory M2 phenotype of macrophages, rather than proinflammatory M1 phenotype and rapamycin treatment actually promotes a shift towards M1 macrophages(20). Interestingly, M2 macrophages are a dominant subpopulation in lupus nephritis biopsies, but whether these macrophages are there to rescue the disease or contribute to pathogenesis is unclear(59). The possibility remains that the ‘canonical’ roles of M1/M2 macrophages are reversed in lupus nephritis.
mTOR prominently features in specific organ involvement of SLE. Kidney damage is a potentially fatal consequence of SLE (49). In a murine model of complex-mediated glomerulonephritis, rapamycin treatment is beneficial in treating disease and prolonging survival(60). Leukocyte infiltration, including B cells, T cells, and macrophages, is a hallmark of glomerulonephritis, and this infiltration appears to be under the control of mTOR and treatable by rapamycin(60). Glomerular architecture is also important in lupus nephritis. Podocytes maintain the slit-diaphragm filtration system of the kidney and are known to be injured in lupus nephritis(61*). In lupus nephritis, these podocytes exhibit increased autophagy(61*). Inhibiting this autophagic process further aggravated podocyte damage, while induction of autophagy ameliorated disease, raising the possibility that podocytes upregulate autophagy in a protective attempt(61*).
Antiphospholipid antibodies (aPL) are a diagnostic criterion of SLE, leading to antiphospholipid syndrome (APS), which can occur in patients with and without SLE. mTORC1 and mTORC2 are activated in APS endothelial cells, associated with intimal hyperplasia, and APS IgG stimulates endothelial cell mTOR activity in vitro(62). Further, sirolimus is clinically beneficial for APS kidney transplant recipients. These patients exhibit reduced renal lesions, stable GFR, and improved graft survival, compared to patients that did not receive sirolimus(62). Further research in mouse models of APS show rapamycin treatment is capable of blocking antiphospholipid antibody production(63).
Targeting mTOR in SLE therapy is promising. In several SLE murine models, inhibition of mTOR decreases production of autoantibodies, decreases proteinuria (a marker of kidney damage), decreases inflammation in multiple organ systems, and even expands lifespan(8, 60, 64),. In human patient populations, a recent phase 1/2 trial studied sirolimus in SLE patients resistant/ intolerant of conventional treatment(65*). After 12 months of treatment, more than half of patients responded to sirolimus therapy, showing decreased disease activity, decreased prednisone usage, sustained antiphospholipid antibody reduction, and restored altered Treg/Th17 ratios(65*). Notably, immunophenotype changes were predictive of responders of non-responders, identifying immunophenotyping as an important biomarker for precision medicine, although the mechanisms underlying differential response require further study.
Concluding Remarks
mTOR signaling, while vastly complex, works to incorporate catabolic and anabolic signals to determine cell fate. By directly and indirectly sensing amino acids, growth hormones, energy, and other factors, mTOR regulates downstream signals of growth, proliferation, and survival. While some deride mTOR for doing everything, it is important to appreciate the role it has in the development and mediation of autoimmunity. mTOR activation and proinflammatory changes lie at the heart of many autoimmune disorders. mTOR is critical to the skewing of Treg/Th17 populations, pathogenesis of other immune cells, and controlling autophagic pathways in multiple cell types in various organs. Targeting this pathway, through both conventional and novel mTOR inhibitors, unlocks a promising therapy in the diseases covered here, among others.
However, some questions remain outstanding for a more complete picture of this therapeutic goal. Primarily, identification of patients that will benefit from treatment and become successful responders is an important goal in the era of precision medicine. While immunophenotyping is predictive of those patients that respond to therapy after initiation, the ability to predict those who will respond to treatment beforehand remains elusive(65*). Similarly, predicting patients that will experience adverse events from conventional mTOR inhibitors, such as sirolimus, is critical for tailoring treatment to populations that will tolerate it. Serious side effects of mTOR inhibition include hyperlipidemia, pneumonia, and sepsis(14). Identification of patients at risk of developing these side effects would be extremely beneficial to therapy goals. In the meantime, combination or alternative therapies that target the mTOR pathway may increase the beneficial effects of sirolimus and reduce side effects. One example of such treatment is N-acetyl cysteine (NAC), a metabolite that shows promising potential in the treatment of SLE via altering the mTOR pathway(66). NAC reduces reactive oxidative species upstream of mTOR activation and may attenuate oxidative damage(66). Combination therapy of NAC and sirolimus could have synergistic effects in therapy, allowing a lower dose and better control of serious side effects. Future work should be directed at successful mitigation of mTOR’s proinflammatory effects, while simultaneously mitigating the off-target effects of mTOR therapy. In pursuit of this, a more complete understanding of the molecular mechanisms behind mTOR signaling can help in the development of new therapeutic targets.
Key Points.
The mTOR pathway integrates environmental cues to shape and alter cellular phenotypes.
mTOR activity, particularly in T cells, potentially drives inflammation in a variety of autoimmune diseases.
Targeted therapy of the mTOR pathway with rapamycin, rapalogues, and novel inhibitors show promising efficacy in the treatment of autoimmune disease.
Financial Support
This work was supported by grants R01 AI 072648 and R01 AI 122176 from the National Institutes of Health and the Central New York Community Foundation.
Footnotes
Conflicts of Interest
None
Reference:
- 1.Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J Antibiot (Tokyo). 1975;28(10):721–6. [DOI] [PubMed] [Google Scholar]
- 2.Martel RR, Klicius J, Galet S. Inhibition of the immune response by rapamycin, a new antifungal antibiotic. Can J Physiol Pharmacol. 1977;55(1):48–51. [DOI] [PubMed] [Google Scholar]
- 3.Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, Lane WS, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. 1994;369(6483):756–8. [DOI] [PubMed] [Google Scholar]
- 4.Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell. 1994;78(1):35–43. [DOI] [PubMed] [Google Scholar]
- 5.Sabers CJ, Martin MM, Brunn GJ, Williams JM, Dumont FJ, Wiederrecht G, et al. Isolation of a protein target of the FKBP12-rapamycin complex in mammalian cells. J Biol Chem. 1995;270(2):815–22. [DOI] [PubMed] [Google Scholar]
- 6.Saxton RA, Sabatini DM. mTOR Signaling in Growth, Metabolism, and Disease. Cell. 2017;169(2):361–71. [DOI] [PubMed] [Google Scholar]
- 7.Carlson RP, Baeder WL, Caccese RG, Warner LM, Sehgal SN. Effects of orally administered rapamycin in animal models of arthritis and other autoimmune diseases. Ann N Y Acad Sci. 1993;685:86–113. [DOI] [PubMed] [Google Scholar]
- 8.Warner LM, Adams LM, Sehgal SN. Rapamycin prolongs survival and arrests pathophysiologic changes in murine systemic lupus erythematosus. Arthritis Rheum. 1994;37(2):289–97. [DOI] [PubMed] [Google Scholar]
- 9.Martin DF, DeBarge LR, Nussenblatt RB, Chan CC, Roberge FG. Synergistic effect of rapamycin and cyclosporin A in the treatment of experimental autoimmune uveoretinitis. J Immunol. 1995;154(2):922–7. [PubMed] [Google Scholar]
- 10.Thomson AW, Turnquist HR, Raimondi G. Immunoregulatory functions of mTOR inhibition. Nat Rev Immunol. 2009;9(5):324–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Powell JD, Delgoffe GM. The mammalian target of rapamycin: linking T cell differentiation, function, and metabolism. Immunity. 2010;33(3):301–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Chi H Regulation and function of mTOR signalling in T cell fate decisions. Nat Rev Immunol. 2012;12(5):325–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Araki K, Ellebedy AH, Ahmed R. TOR in the immune system. Curr Opin Cell Biol. 2011;23(6):707–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Perl A mTOR activation is a biomarker and a central pathway to autoimmune disorders, cancer, obesity, and aging. Ann N Y Acad Sci. 2015;1346(1):33–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol. 2011;12(4):295–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *16.Huang N, Perl A. Metabolism as a Target for Modulation in Autoimmune Diseases. Trends Immunol. 2018;39(7):562–76. [DOI] [PubMed] [Google Scholar]; Outstanding review on pathways that control ROS production and mTOR activation, as well as idenfication of therapeutic targets in autoimmune disease.
- 17.Kurebayashi Y, Nagai S, Ikejiri A, Ohtani M, Ichiyama K, Baba Y, et al. PI3K-Akt-mTORC1-S6K1/2 axis controls Th17 differentiation by regulating Gfi1 expression and nuclear translocation of RORgamma. Cell Rep. 2012;1(4):360–73. [DOI] [PubMed] [Google Scholar]
- 18.Zeng Q, Zhang H, Qin J, Xu Z, Gui L, Liu B, et al. Rapamycin inhibits BAFF-stimulated cell proliferation and survival by suppressing mTOR-mediated PP2A-Erk1/2 signaling pathway in normal and neoplastic B-lymphoid cells. Cell Mol Life Sci. 2015;72(24):4867–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *19.Zeng Q, Qin S, Zhang H, Liu B, Qin J, Wang X, et al. Rapamycin attenuates BAFF-extended proliferation and survival via disruption of mTORC1/2 signaling in normal and neoplastic B-lymphoid cells. J Cell Physiol. 2018;233(1):516–29. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original research on rapamycin’s ability to abrogate B cell proliferation via mTORC1 and mTORC2, identifying mTOR as a therapeutic target in B cell proliferative diseases.
- 20.Mercalli A, Calavita I, Dugnani E, Citro A, Cantarelli E, Nano R, et al. Rapamycin unbalances the polarization of human macrophages to M1. Immunology. 2013;140(2):179–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Boissier MC, Semerano L, Challal S, Saidenberg-Kermanac’h N, Falgarone G. Rheumatoid arthritis: from autoimmunity to synovitis and joint destruction. J Autoimmun. 2012;39(3):222–8. [DOI] [PubMed] [Google Scholar]
- 22.Bruyn GA, Tate G, Caeiro F, Maldonado-Cocco J, Westhovens R, Tannenbaum H, et al. Everolimus in patients with rheumatoid arthritis receiving concomitant methotrexate: a 3-month, double-blind, randomised, placebo-controlled, parallel-group, proof-of-concept study. Ann Rheum Dis. 2008;67(8):1090–5. [DOI] [PubMed] [Google Scholar]
- 23.Yang Z, Fujii H, Mohan SV, Goronzy JJ, Weyand CM. Phosphofructokinase deficiency impairs ATP generation, autophagy, and redox balance in rheumatoid arthritis T cells. J Exp Med. 2013;210(10):2119–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Perl A Activation of mTOR (mechanistic target of rapamycin) in rheumatic diseases. Nat Rev Rheumatol. 2016;12(3):169–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *25.Karonitsch T, Kandasamy RK, Kartnig F, Herdy, Dalwigk K, Niederreiter B, et al. mTOR Senses Environmental Cues to Shape the Fibroblast-like Synoviocyte Response to Inflammation. Cell Rep. 2018;23(7):2157–67. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original study identifying a signaling circuit in which TNF activates mTOR within FLS, identifying mTOR as a therapeutic target.
- 26.Mori T, Miyamoto T, Yoshida H, Asakawa M, Kawasumi M, Kobayashi T, et al. IL-1beta and TNFalpha-initiated IL-6-STAT3 pathway is critical in mediating inflammatory cytokines and RANKL expression in inflammatory arthritis. Int Immunol. 2011;23(11):701–12. [DOI] [PubMed] [Google Scholar]
- 27.Oike T, Sato Y, Kobayashi T, Miyamoto K, Nakamura S, Kaneko Y, et al. Stat3 as a potential therapeutic target for rheumatoid arthritis. Sci Rep. 2017;7(1):10965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhou J, Wulfkuhle J, Zhang H, Gu P, Yang Y, Deng J, et al. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc Natl Acad Sci U S A. 2007;104(41):16158–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Audia S, Mahevas M, Samson M, Godeau B, Bonnotte B. Pathogenesis of immune thrombocytopenia. Autoimmun Rev. 2017;16(6):620–32. [DOI] [PubMed] [Google Scholar]
- 30.Ji L, Zhan Y, Hua F, Li F, Zou S, Wang W, et al. The ratio of Treg/Th17 cells correlates with the disease activity of primary immune thrombocytopenia. PLoS One. 2012;7(12):e50909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhang A, Ning B, Sun N, Wei J, Ju X. Indirubin Increases CD4+CD25+Foxp3+ Regulatory T Cells to Prevent Immune Thrombocytopenia in Mice. PLoS One. 2015;10(11):e0142634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhao Y, Han P, Liu L, Wang X, Xu P, Wang H, et al. Indirubin modulates CD4(+) T-cell homeostasis via PD1/PTEN/AKT signalling pathway in immune thrombocytopenia. J Cell Mol Med. 2019;23(3):1885–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Li J, Wang Z, Dai L, Cao L, Su J, Zhu M, et al. Effects of rapamycin combined with low dose prednisone in patients with chronic immune thrombocytopenia. Clin Dev Immunol. 2013;2013:548085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jasinski S, Weinblatt ME, Glasser CL. Sirolimus as an Effective Agent in the Treatment of Immune Thrombocytopenia (ITP) and Evans Syndrome (ES): A Single Institution’s Experience. J Pediatr Hematol Oncol. 2017;39(6):420–4. [DOI] [PubMed] [Google Scholar]
- *35.Sun RJ, Shan NN. Megakaryocytic dysfunction in immune thrombocytopenia is linked to autophagy. Cancer Cell Int. 2019;19:59. [DOI] [PMC free article] [PubMed] [Google Scholar]; Outstanding review on the role of autophagic pathways and mTOR in the setting of ITP
- 36.McMillan R, Wang L, Tomer A, Nichol J, Pistillo J. Suppression of in vitro megakaryocyte production by antiplatelet autoantibodies from adult patients with chronic ITP. Blood. 2004;103(4):1364–9. [DOI] [PubMed] [Google Scholar]
- 37.Cao Y, Cai J, Zhang S, Yuan N, Li X, Fang Y, et al. Loss of autophagy leads to failure in megakaryopoiesis, megakaryocyte differentiation, and thrombopoiesis in mice. Exp Hematol. 2015;43(6):488–94. [DOI] [PubMed] [Google Scholar]
- 38.Ouseph MM, Huang Y, Banerjee M, Joshi S, MacDonald L, Zhong Y, et al. Autophagy is induced upon platelet activation and is essential for hemostasis and thrombosis. Blood. 2015;126(10):1224–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Raslova H, Baccini V, Loussaief L, Comba B, Larghero J, Debili N, et al. Mammalian target of rapamycin (mTOR) regulates both proliferation of megakaryocyte progenitors and late stages of megakaryocyte differentiation. Blood. 2006;107(6):2303–10. [DOI] [PubMed] [Google Scholar]
- 40.Drayer AL, Olthof SG, Vellenga E. Mammalian target of rapamycin is required for thrombopoietin-induced proliferation of megakaryocyte progenitors. Stem Cells. 2006;24(1):105–14. [DOI] [PubMed] [Google Scholar]
- 41.Liu Z, Mei T. Immune thrombocytopenia induces autophagy and suppresses apoptosis in megakaryocytes. Mol Med Rep. 2018;18(4):4016–22. [DOI] [PubMed] [Google Scholar]
- 42.Shan NN, Dong LL, Zhang XM, Liu X, Li Y. Targeting autophagy as a potential therapeutic approach for immune thrombocytopenia therapy. Crit Rev Oncol Hematol. 2016;100:11–5. [DOI] [PubMed] [Google Scholar]
- 43.Kassan SS, Moutsopoulos HM. Clinical manifestations and early diagnosis of Sjogren syndrome. Arch Intern Med. 2004;164(12):1275–84. [DOI] [PubMed] [Google Scholar]
- 44.Shah M, Edman MC, Janga SR, Shi P, Dhandhukia J, Liu S, et al. A rapamycin-binding protein polymer nanoparticle shows potent therapeutic activity in suppressing autoimmune dacryoadenitis in a mouse model of Sjogren’s syndrome. J Control Release. 2013;171(3):269–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *45.Blokland SLM, Hillen MR, Wichers CGK, Zimmermann M, Kruize AA, Radstake T, et al. Increased mTORC1 activation in salivary gland B cells and T cells from patients with Sjogren’s syndrome: mTOR inhibition as a novel therapeutic strategy to halt immunopathology? RMD Open. 2019;5(1):e000701. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original study demonstrating increased mTOR activity in T and B lymphocytes in salivary glands of patients with Sjögren’s syndrome.
- 46.Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, et al. 2012 revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides. Arthritis Rheum. 2013;65(1):1–11. [DOI] [PubMed] [Google Scholar]
- *47.Hadjadj J, Canaud G, Mirault T, Samson M, Bruneval P, Regent A, et al. mTOR pathway is activated in endothelial cells from patients with Takayasu arteritis and is modulated by serum immunoglobulin G. Rheumatology (Oxford). 2018;57(6):1011–20. [DOI] [PubMed] [Google Scholar]; Original research on the prescence of mTOR activity in Takayasu arteritis.
- *48.Maciejewski-Duval A, Comarmond C, Leroyer A, Zaidan M, Le Joncour A, Desbois AC, et al. mTOR pathway activation in large vessel vasculitis. J Autoimmun. 2018;94:99–109. [DOI] [PubMed] [Google Scholar]; Original research on the prescence of mTOR activity in both Takayasu arteritis as well as giant cell arteritis.
- 49.Tsokos GC. Systemic lupus erythematosus. N Engl J Med. 2011;365(22):2110–21. [DOI] [PubMed] [Google Scholar]
- 50.Francis L, Perl A. Pharmacotherapy of systemic lupus erythematosus. Expert Opin Pharmacother. 2009;10(9):1481–94. [DOI] [PubMed] [Google Scholar]
- 51.Fernandez DR, Telarico T, Bonilla E, Li Q, Banerjee S, Middleton FA, et al. Activation of mammalian target of rapamycin controls the loss of TCRzeta in lupus T cells through HRES-1/Rab4-regulated lysosomal degradation. J Immunol. 2009;182(4):2063–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Fernandez D, Bonilla E, Mirza N, Niland B, Perl A. Rapamycin reduces disease activity and normalizes T cell activation-induced calcium fluxing in patients with systemic lupus erythematosus. Arthritis Rheum. 2006;54(9):2983–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- *53.Kato H, Perl A. Blockade of Treg Cell Differentiation and Function by the Interleukin-21-Mechanistic Target of Rapamycin Axis Via Suppression of Autophagy in Patients With Systemic Lupus Erythematosus. Arthritis Rheumatol. 2018;70(3):427–38. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original research showing that SLE Tregs exhibit increased mTORC1 and mTORC2 activities and diminished autophagy and suppressor function which can be effectively restored by treatment with rapamycin.
- *54.Kono M, Yoshida N, Maeda K, Suarez-Fueyo A, Kyttaris VC, Tsokos GC. Glutaminase 1 Inhibition Reduces Glycolysis and Ameliorates Lupus-Like Disease in MRL/lpr Mice and Experimental Autoimmune Encephalomyelitis. Arthritis Rheumatol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original research demonstrating that inhibition of glutaminolysis is a potential new treatment strategy for patients with SLE and Th17-related autoimmune diseases.
- *55.Katsuyama T, Li H, Comte D, Tsokos GC, Moulton VR. Splicing factor SRSF1 controls T cell hyperactivity and systemic autoimmunity. J Clin Invest. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original research identifying the existence of the SRSF1-PTEN-mTOR pathway as a therapeutic target in SLE patients.
- 56.Kato H, Perl A. Mechanistic target of rapamycin complex 1 expands Th17 and IL-4+ CD4− CD8− double-negative T cells and contracts regulatory T cells in systemic lupus erythematosus. J Immunol. 2014;192(9):4134–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lai ZW, Borsuk R, Shadakshari A, Yu J, Dawood M, Garcia R, et al. Mechanistic target of rapamycin activation triggers IL-4 production and necrotic death of double-negative T cells in patients with systemic lupus erythematosus. J Immunol. 2013;191(5):2236–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kobayashi T, Shimabukuro-Demoto S, Yoshida-Sugitani R, Furuyama-Tanaka K, Karyu H, Sugiura Y, et al. The histidine transporter SLC15A4 coordinates mTOR-dependent inflammatory responses and pathogenic antibody production. Immunity. 2014;41(3):375–88. [DOI] [PubMed] [Google Scholar]
- 59.Olmes G, Buttner-Herold M, Ferrazzi F, Distel L, Amann K, Daniel C. CD163+ M2c-like macrophages predominate in renal biopsies from patients with lupus nephritis. Arthritis Res Ther. 2016;18:90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lui SL, Tsang R, Chan KW, Zhang F, Tam S, Yung S, et al. Rapamycin attenuates the severity of established nephritis in lupus-prone NZB/W F1 mice. Nephrol Dial Transplant. 2008;23(9):2768–76. [DOI] [PubMed] [Google Scholar]
- *61.Qi YY, Zhou XJ, Cheng FJ, Hou P, Ren YL, Wang SX, et al. Increased autophagy is cytoprotective against podocyte injury induced by antibody and interferon-alpha in lupus nephritis. Ann Rheum Dis. 2018;77(12):1799–809. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original research identifying autophagic pathways as cytoprotective for podocytes in lupus nephritis
- 62.Canaud G, Bienaime F, Tabarin F, Bataillon G, Seilhean D, Noel LH, et al. Inhibition of the mTORC pathway in the antiphospholipid syndrome. N Engl J Med. 2014;371(4):303–12. [DOI] [PubMed] [Google Scholar]
- 63.Oaks Z, Winans T, Caza T, Fernandez D, Liu Y, Landas SK, et al. Mitochondrial Dysfunction in the Liver and Antiphospholipid Antibody Production Precede Disease Onset and Respond to Rapamycin in Lupus-Prone Mice. Arthritis Rheumatol. 2016;68(11):2728–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ramos-Barron A, Pinera-Haces C, Gomez-Alamillo C, Santiuste-Torcida I, Ruiz JC, Buelta-Carrillo L, et al. Prevention of murine lupus disease in (NZBxNZW)F1 mice by sirolimus treatment. Lupus. 2007;16(10):775–81. [DOI] [PubMed] [Google Scholar]
- *65.Lai ZW, Kelly R, Winans T, Marchena I, Shadakshari A, Yu J, et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: a single-arm, open-label, phase 1/2 trial. Lancet. 2018;391(10126):1186–96. [DOI] [PMC free article] [PubMed] [Google Scholar]; Original prospective study demonstrating promising clinical efficacy and mechanism of action of sirolimus in SLE.
- 66.Lai ZW, Hanczko R, Bonilla E, Caza TN, Clair B, Bartos A, et al. N-acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2012;64(9):2937–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yin Y, Choi SC, Xu Z, Perry DJ, Seay H, Croker BP, et al. Normalization of CD4+ T cell metabolism reverses lupus. Sci Transl Med. 2015;7(274):274ra18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Biniecka M, Fox E, Gao W, Ng CT, Veale DJ, Fearon U, et al. Hypoxia induces mitochondrial mutagenesis and dysfunction in inflammatory arthritis. Arthritis Rheum. 2011;63(8):2172–82. [DOI] [PubMed] [Google Scholar]
- 69.Paul M, Hemshekhar M, Thushara RM, Sundaram MS, NaveenKumar SK, Naveen S, et al. Methotrexate Promotes Platelet Apoptosis via JNK-Mediated Mitochondrial Damage: Alleviation by N-Acetylcysteine and N-Acetylcysteine Amide. PLoS One. 2015;10(6):e0127558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Vasheghani F, Zhang Y, Li YH, Blati M, Fahmi H, Lussier B, et al. PPARgamma deficiency results in severe, accelerated osteoarthritis associated with aberrant mTOR signalling in the articular cartilage. Ann Rheum Dis. 2015;74(3):569–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhang Y, Vasheghani F, Li YH, Blati M, Simeone K, Fahmi H, et al. Cartilage-specific deletion of mTOR upregulates autophagy and protects mice from osteoarthritis. Ann Rheum Dis. 2015;74(7):1432–40. [DOI] [PubMed] [Google Scholar]
- 72.Javier AF, Bata-Csorgo Z, Ellis CN, Kang S, Voorhees JJ, Cooper KD. Rapamycin (sirolimus) inhibits proliferating cell nuclear antigen expression and blocks cell cycle in the G1 phase in human keratinocyte stem cells. J Clin Invest. 1997;99(9):2094–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Yoshizaki A, Yanaba K, Yoshizaki A, Iwata Y, Komura K, Ogawa F, et al. Treatment with rapamycin prevents fibrosis in tight-skin and bleomycin-induced mouse models of systemic sclerosis. Arthritis Rheum. 2010;62(8):2476–87. [DOI] [PubMed] [Google Scholar]
- 74.Su TI, Khanna D, Furst DE, Danovitch G, Burger C, Maranian P, et al. Rapamycin versus methotrexate in early diffuse systemic sclerosis: results from a randomized, single-blind pilot study. Arthritis Rheum. 2009;60(12):3821–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Seyfarth HJ, Hammerschmidt S, Halank M, Neuhaus P, Wirtz HR. Everolimus in patients with severe pulmonary hypertension: a safety and efficacy pilot trial. Pulm Circ. 2013;3(3):632–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Walters MT, Rubin CE, Keightley SJ, Ward CD, Cawley MI. A double-blind, cross-over, study of oral N-acetylcysteine in Sjogren’s syndrome. Scand J Rheumatol Suppl. 1986;61:253–8. [PubMed] [Google Scholar]
- 77.Bride KL, Vincent T, Smith-Whitley K, Lambert MP, Bleesing JJ, Seif AE, et al. Sirolimus is effective in relapsed/refractory autoimmune cytopenias: results of a prospective multi-institutional trial. Blood. 2016;127(1):17–28. [DOI] [PMC free article] [PubMed] [Google Scholar]