Acessibilidade / Reportar erro

Effects of immune exhaustion and senescence of innate immunity in autoimmune disorders

Abstract

Innate immune system activation is crucial in the inflammatory response, but uncontrolled activation can lead to autoimmune diseases. Cellular exhaustion and senescence are two processes that contribute to innate immune tolerance breakdown. Exhausted immune cells are unable to respond adequately to specific antigens or stimuli, while senescent cells have impaired DNA replication and metabolic changes. These processes can impair immune system function and disrupt homeostasis, leading to the emergence of autoimmunity. However, the influence of innate immune exhaustion and senescence on autoimmune disorders is not well understood. This review aims to describe the current findings on the role of innate immune exhaustion and senescence in autoimmunity, focusing on the cellular and molecular changes involved in each process. Specifically, the article explores the markers and pathways associated with immune exhaustion, such as PD-1 and TIM-3, and senescence, including Β-galactosidase (β-GAL), lamin B1, and p16ink4a, and their impact on autoimmune diseases, namely type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and immune-mediated myopathies. Understanding the mechanisms underlying innate immune exhaustion and senescence in autoimmunity may provide insights for the development of novel therapeutic strategies.

Immune exhaustion; Immune senescence; Innate immune system; Autoimmunity and immunology


Introduction

Innate immunity is involved in inflammatory response but its uncontrolled activation has been somehow linked to autoimmunity. As in any other inflammatory pathway, each innate component is tightly modulated by other redundant proteins. Therefore, defects in any of these checkpoints may cause a spectrum of immunodeficiency and autoinflammatory or autoimmune diseases (11. Frizinsky S, Haj-Yahia S, Maayan DM, Lifshitz Y, Maoz-Segal R, Offengenden I, et al. The innate immune perspective of autoimmune and autoinflammatory conditions. Rheumatology (Oxford) 2019; 58: vi1-vi8, doi: 10.1093/rheumatology/kez387.
https://doi.org/10.1093/rheumatology/kez...
).

Cellular exhaustion and senescence may also contribute to innate immune tolerance breakdown. Immune cells become exhausted when unable to adequately respond after a challenge with specific antigens or stimuli (22. Akbar AN, Henson SM. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat Rev Immunol 2011; 11: 289-295, doi: 10.1038/nri2959.
https://doi.org/10.1038/nri2959...
). On the other hand, cell senescence is defined by its inability to replicate as telomeres reach a critical length or DNA is irreparably damaged (33. Aiello A, Farzaneh F, Candore G, Caruso C, Davinelli S, Gambino CM, et al. Immunosenescence and its hallmarks: how to oppose aging strategically? A review of potential options for therapeutic intervention. Front Immunol 2019; 10: 2247, doi: 10.3389/fimmu.2019.02247.
https://doi.org/10.3389/fimmu.2019.02247...
). Although exhaustion and senescence may be considered part of the physiological cell maturation, they may also impair overall immune system function and destabilize homeostasis over time, finally culminating with self-tolerance breakdown and the emergence of autoimmunity (44. Verdon DJ, Mulazzani M, Jenkins MR. Cellular and molecular mechanisms of CD8(+) T cell differentiation, dysfunction and exhaustion. Int J Mol Sci 2020; 21: 7357, doi: 10.3390/ijms21197357.
https://doi.org/10.3390/ijms21197357...
).

The innate immune system is particularly affected by immune exhaustion and senescence, which can be observed by the constant turnover of effector cells and soluble factors. Nevertheless, to the best of our knowledge few articles address the influence of innate immune exhaustion and senescence on autoimmune disorders. Herein, we aimed to describe the current findings of innate immune exhaustion and senescence on autoimmunity, mainly focusing on structural cellular modification and molecular pathways involved in each process.

Immune exhaustion

Cell surface costimulatory signaling modulators are hallmarks of immune exhaustion [Table 1, (55. Huang X, Venet F, Wang YL, Lepape A, Yuan Z, Chen Y, et al. PD-1 expression by macrophages plays a pathologic role in altering microbial clearance and the innate inflammatory response to sepsis. Proc Nat Acad Sci USA 2009; 106: 6303-6303, doi: 10.1073/pnas.0809422106.
https://doi.org/10.1073/pnas.0809422106...
-6. Phong B, Avery L, Menk AV, Delgoffe GM, Kane LP. cutting edge: murine mast cells rapidly modulate metabolic pathways essential for distinct effector functions. J Immunol 2017; 198: 640-644, doi: 10.4049/jimmunol.1601150.
https://doi.org/10.4049/jimmunol.1601150...
77. Jahromi NH, Marchetti L, Moalli F, Duc D, Basso C, Tardent H, et al. Intercellular adhesion molecule-1 (ICAM-1) and ICAM-2 differentially contribute to peripheral activation and CNS entry of autoaggressive Th1 and Th17 cells in experimental autoimmune encephalomyelitis. Front Immunol 2020; 10: 3056, doi: 10.3389/fimmu.2019.03056.
https://doi.org/10.3389/fimmu.2019.03056...
)] and can be easily assessed by using different tools like flow cytometry, immunohistochemistry, and western blot. Programmed cell death protein 1 (PD1) and its ligands 1 (PD-L1) and 2 (PD-L2) constitute an important regulatory pathway that impedes costimulatory signaling during T cell activation (88. Araki K, Youngblood B, Ahmed R. Programmed cell death 1-directed immunotherapy for enhancing T-cell function. Cold Spring Harb Symp Quant Biol 2013; 78: 239-247, doi: 10.1101/sqb.78.019869.
https://doi.org/10.1101/sqb.78.019869...
). Simultaneous co-expression of PD1/PDL-1 with other inhibitory receptors on T cells, such as lymphocyte activation gene 3 protein (LAG3), 2B4/CD244, CD160, T cell immunoglobulin domain and mucin domain-containing protein 3 (TIM3), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4) is highly suggestive of an immune exhaustion phenotype. A PD-1/PD-L1 pathway neutralization-induced bystander effect on NK cells was observed in an experimental IL-2-dependent exhaustion mouse model, resulting from the global competition that exists between NK and CD8+ T cells for IL-2 as a key regulator of these cells' activation (99. Golden-Mason L, Palmer BE, Kassam N, Townshend-Bulson L, Livingston S, McMahon BJ, et al. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol 2009; 83: 9122-9130, doi: 10.1128/JVI.00639-09.
https://doi.org/10.1128/JVI.00639-09...
). PD-1 and TIM-3 combined target may be the most efficacious manner to improve anti-tumor response in vivo as demonstrated in BALB/c and C57BL/6 mice models (1010. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2010; 207: 2187-2194, doi: 10.1084/jem.20100643.
https://doi.org/10.1084/jem.20100643...
). Interestingly, the higher the number of inhibitory receptors expressed by exhausted T cells, the more severe the exhaustion process evolves, a pattern apparently also shared by innate immune cells. Lin et al. (1111. Lin R, Zhang Y, Pradhan K, Li L. TICAM2-related pathway mediates neutrophil exhaustion. Sci Rep 2020; 10: 14397, doi: 10.1038/s41598-020-71379-y.
https://doi.org/10.1038/s41598-020-71379...
) showed that LPS stimulus caused neutrophil exhaustion after TIR domain-containing adapter molecule 2 (TICAM2) and PD1-mediated activation of Src family kinases (SFK). PD1/PD-L1 are also expressed on exhausted murine monocytes/macrophages and dendritic cells (DC) from septic peritonitis induced by a cecal ligation murine model, similarly to those derived from septic shock patients (1212. Zasada M, Lenart M, Rutkowska-Zapała M, Stec M, Durlak W, Grudzień A, et al. Analysis of PD-1 expression in the monocyte subsets from non-septic and septic preterm neonates. PloS One 2017, 8; 12: e0186819, doi: 10.1371/journal.pone.0186819.
https://doi.org/10.1371/journal.pone.018...
). Hence, as PD1/PD-L1 and CTLA-4 are currently important targets for cancer immunotherapy (1313. Seliger B. Basis of PD1/PD-L1 therapies. J Clin Med 2019; 8: 2168, doi: 10.3390/jcm8122168.
https://doi.org/10.3390/jcm8122168...
), other exhaustion markers, such as TIM3, have similarly been considered (1414. da Silva IP, Gallois A, Jimenez-Baranda S, Khan S, Anderson AC, Kuchroo VK, et al. Reversal of NK-Cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol Res 2014; 2: 410-422, doi: 10.1158/2326-6066.CIR-13-0171.
https://doi.org/10.1158/2326-6066.CIR-13...
).

Table 1
Key cell surface costimulatory signaling modulators associated with innate immunity exhaustion.

Immune senescence

Cells become senescent when their DNA replication ability progressively deteriorates, resulting in striking metabolic modifications and expression of immune senescence markers. There is compelling evidence that immune senescence plays a significant role in immune dysfunction and disability in older people (1515. Maue AC, Yager EJ, Swain SL, Woodland DL, Blackman MA, Haynes L. T-cell immunosenescence: lessons learned from mouse models of aging. Trends Immunol 2009; 30: 301-305, doi: 10.1016/j.it.2009.04.007.
https://doi.org/10.1016/j.it.2009.04.007...
). Elderly have worse T cell memory responses than young people, which may likely be the result of a combination of factors including reduced TCR repertoire diversity, poor T cell assistance, and substantial decreased naive T cell count along with aging, as shown in HIV-infected elders (1616. Álvarez S, Braãas F, Sánchez-Conde M, Moreno S, de Quirós JCLB, Muãoz-Fernández MÁ. Frailty, markers of immune activation and oxidative stress in HIV infected elderly. PloS One 2020; 15: e0230339, doi: 10.1371/journal.pone.0230339.
https://doi.org/10.1371/journal.pone.023...
). Previous studies indicate that the ageing process or repeated cell activation cycles significantly impede the ability of immune cells to start primary responses against novel antigens, although immune responses against previously recognized antigens may still be conserved (1717. Lazuardi L, Jenewein B, Wolf AM, Pfister G, Tzankov A, Grubeck-Loebenstein B. Age-related loss of naïve T cells and dysregulation of T-cell/B-cell interactions in human lymph nodes. Immunology 2005; 114: 37-43, doi: 10.1111/j.1365-2567.2004.02006.x.
https://doi.org/10.1111/j.1365-2567.2004...
). This difficulty is usually increased by an impairment of innate immune effector cells, mainly neutrophils and monocytes, and may result in susceptibility to infectious diseases (1818. Fagnoni FF, Vescovini R, Passeri G, Bologna G, Pedrazzoni M, Lavagetto G, et al. Shortage of circulating naive CD8+ T cells provides new insights on immunodeficiency in aging. Blood 2000; 95: 2860-2868, doi: 10.1182/blood.V95.9.2860.009k35_2860_2868.
https://doi.org/10.1182/blood.V95.9.2860...
). The microbicidal function of senescent neutrophils is highly impaired, mostly because of a reduced chemotactic ability, which in turn delays tissue recovery as shown in mouse lungs (1919. Nomellini V, Faunce DE, Gomez CR, Kovacs EJ. An age-associated increase in pulmonary inflammation after burn injury is abrogated by CXCR2 inhibition. J Leukoc Biol 2008; 83: 1493-1501, doi: 10.1189/jlb.1007672.
https://doi.org/10.1189/jlb.1007672...
). Moreover, reduced neutrophil phagocytic activity against opsonized E. Coli and Fcγ receptor CD16 surface expression were previously shown in elderly humans (2020. Butcher SK, Chahal H, Nayak L, Sinclair A, Henriquez NV, Sapey E, et al. Senescence in innate immune responses: reduced neutrophil phagocytic capacity and CD16 expression in elderly humans. J Leukoc Biol 2001; 70: 881-886, doi: 10.1189/jlb.70.6.881.
https://doi.org/10.1189/jlb.70.6.881...
).

Impaired intracellular signaling have also been reported in senescent neutrophils, including reduced calcium intake, decreased kinase and phosphatase activities [namely, phosphoinositide-3 kinase (PI-3K), mitogen-activated protein kinase (MAPK), protein kinase B, and Src homology region 2 domain-containing phosphatase-1 (SHP-1)], and impaired Janus kinase (JAK)-signal transducer and activator of transcription (STAT) interaction (2121. Wessels I, Jansen J, Rink L, Uciechowski P. Immunosenescence of polymorphonuclear neutrophils. ScientificWorldJournal 2010; 10: 145-160, doi: 10.1100/tsw.2010.14.
https://doi.org/10.1100/tsw.2010.14...
). Altered intracellular signaling in senescent neutrophils may also hamper oxidative burst and phagocytic activity. For most other aspects of neutrophil senescence our understanding is still incomplete.

Other features of immune cell senescence have also been described, mainly leading to the irreversible pause of cell growth and development of a proinflammatory senescence-associated secretory phenotype (SASP; Figure 1 and Table 2) (2222. Kumari R, Jat P. Mechanisms of cellular senescence: cell cycle arrest and senescence associated secretory phenotype. Front Cell Dev Biol 2021; 9: 645593, doi: 10.3389/fcell.2021.645593.
https://doi.org/10.3389/fcell.2021.64559...
) such as β-galactosidase (β-GAL) and p16INK4a (2323. de Mera-Rodríguez JA, Álvarez-Hernán G, Gaãán Y, Martín-Partido G, Rodríguez-León J, Francisco-Morcillo J. Is senescence-associated β-galactosidase a reliable in vivo marker of cellular senescence during embryonic development? Front Cell Dev Biol 2021; 9: 623175, doi: 10.3389/fcell.2021.623175.
https://doi.org/10.3389/fcell.2021.62317...
). As a component of the cyclin-dependent kinase (CDK) inhibitors family, p16INK4a blocks retinoblastoma protein, ultimately impeding S-phase entry and cell growth (2424. Giacinti C, Giordano A. RB and cell cycle progression. Oncogene 2006; 25: 5220-5227, doi: 10.1038/sj.onc.1209615.
https://doi.org/10.1038/sj.onc.1209615...
). Liu et al. (2525. Liu JY, Souroullas GP, Diekman BO, Krishnamurthy J, Hall BM, Sorrentino JA, et al. Cells exhibiting strong p16 (INK4a) promoter activation in vivo display features of senescence. Proc Natl Acad Sci USA 2019; 116: 2603-2611, doi: 10.1073/pnas.1818313116.
https://doi.org/10.1073/pnas.1818313116...
) demonstrated a senescent phenotype in murine peritoneal macrophages derived from hybrid C57BL6/129SvEv transgenic model. Clearance of p16INK4a-expressing cells attenuates senescence and improves the healthy lifespan of a progeroid mouse model and aged control mice, as β-GAL was upregulated after p16INK4a activation (2525. Liu JY, Souroullas GP, Diekman BO, Krishnamurthy J, Hall BM, Sorrentino JA, et al. Cells exhibiting strong p16 (INK4a) promoter activation in vivo display features of senescence. Proc Natl Acad Sci USA 2019; 116: 2603-2611, doi: 10.1073/pnas.1818313116.
https://doi.org/10.1073/pnas.1818313116...
).

Figure 1
A, Main variations in senescence intracellular markers with ageing. B, The senescent stage: innate sensing involves multiple stressors, such as telomere attrition, oxidative stress, irradiation, ageing, and oncogene activation. Any stressor may induce three main senescence responses: i) AKT-dependent phosphoinositide 3-kinase (PI-3K) downregulation, which triggers reactive oxygen species (ROS) production, causing DNA damage; ii) direct double-stranded DNA damage releasing fragments enriched with yH2AX and repressive histone markers (H3K9me3, H3K27me3); and iii) lamin B1 downregulation. Upon disruption of the nuclear envelope favored by loss of lamin B1, yH2AX-, H3K9me3-, and H3K27me3-enriched DNA fragments are recognized by cyclic GMP-AMP synthase (cGAS), which activates nuclear factor κB (NF-κB) and interferon (IFN) pathways, culminating with senescence-associated secretory phenotype (SASP). SASP reinforces and amplifies senescence in a paracrine manner, activating immune cells for senescence immune surveillance, which in turn increases mitochondrial dysfunctional ROS release and upregulates β-galactosidase (β-GAL) and p16INK4a. Moreover, ROS overproduction is a major lamin B1 downregulator, feeding back the whole senescent process.
Table 2
Physiologic features of the main components involved in senescence-associated secretory phenotype (SASP).

Another SASP biomarker is lamin B1, a structural cell nuclear component involved in regulating many nuclear functions (2626. Shimi T, Butin-Israeli V, Adam SA, Hamanaka RB, Goldman AE, Lucas CA, et al. The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev 2011; 25: 2579-2593, doi: 10.1101/gad.179515.111.
https://doi.org/10.1101/gad.179515.111...
). Lamin B1 is downregulated in ultraviolet radiation (UV) in vitro-induced human senescent cells; similar evidence was demonstrated upon chronic in vivo UV exposure and skin regeneration (2727. Wang AS, Ong PF, Chojnowski A, Clavel C, Dreesen O. Loss of lamin B1 is a biomarker to quantify cellular senescence in photoaged skin. Sci Rep 2017; 7: 15678, doi: 10.1038/s41598-017-15901-9.
https://doi.org/10.1038/s41598-017-15901...
). In addition, lamin B1 gene and protein expression declined in UV-induced murine senescence (2828. Freund A, Laberge RM, Demaria M, Campisi J. Lamin B1 loss is a senescence-associated biomarker. Mol Biol Cell 2012; 23: 2066-2075, doi: 10.1091/mbc.e11-10-0884.
https://doi.org/10.1091/mbc.e11-10-0884...
).

The cell maturation process is also affected by immune senescence, as human monocytes and dendritic cells (DC) progressively decline with time. Paradoxically, while senescent monocyte absolute count increases with ageing, peripheral macrophage number decreases and they become progressively resistant to Toll-like receptor (TLR) activation (2929. Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol 2010; 184: 2518-2527, doi: 10.4049/jimmunol.0901022.
https://doi.org/10.4049/jimmunol.0901022...
). Bella et al. (3030. Bella SD, Bierti L, Presicce P, Arienti R, Valenti M, Saresella M, et al. Peripheral blood dendritic cells and monocytes are differently regulated in the elderly. Clin Immunol 2007; 122: 220-228, doi: 10.1016/j.clim.2006.09.012.
https://doi.org/10.1016/j.clim.2006.09.0...
) demonstrated that LPS-stimulated interleukin (IL)-12 and IL-10 in vitro production by murine senescent monocytes and DC is impaired. On the other hand, IL-6, IL-8, and IL-1α production increases as cells become senescent. IL-1α blockade in senescent cells markedly reduced IL-6 and IL-8 secretion (3131. Orjalo AV, Bhaumik D, Gengler BK, Scott GK, Campisi J. Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc Natl Acad Sci USA 2009; 106: 17031-17036, doi: 10.1073/pnas.0905299106.
https://doi.org/10.1073/pnas.0905299106...
). Similar dysfunction was demonstrated in senescent neutrophils and monocytes, whose TLR2/6, 3, 5, and 9-stimulated cytokines in vitro production is also totally defective (2121. Wessels I, Jansen J, Rink L, Uciechowski P. Immunosenescence of polymorphonuclear neutrophils. ScientificWorldJournal 2010; 10: 145-160, doi: 10.1100/tsw.2010.14.
https://doi.org/10.1100/tsw.2010.14...
). Moreover, a previous study showed impairment of TLR gene expression in C57BL/6 mice splenic and peritoneal senescent macrophages (3232. Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol 2013; 13: 875-887, doi: 10.1038/nri3547.
https://doi.org/10.1038/nri3547...
).

How exhausted or senescent innate effector cells may affect autoimmunity pathophysiology

Cell exhaustion or senescence represents a state of cellular dysfunction characterized by suppressed cellular functionality. Within the context of autoimmunity, exhausted or senescent cells exhibit impaired functionality, thereby compromising immune system capacity of effectively eliminating pathogens, neoplasms, and autoreactive cells (3333. McHugh D, Gil J. Senescence and aging: Causes, consequences, and therapeutic avenues. J Cell Biol 2017; 217: 65-77, doi: 10.1083/jcb.201708092.
https://doi.org/10.1083/jcb.201708092...
). Experimental evidence has demonstrated the presence of exhausted and senescent cells in both human and murine systems, highlighting their involvement in autoimmunity.

Type 1 diabetes (T1D)

Diana et al. (3434. Diana J, Simoni Y, Furio L, Beaudoin L, Agerberth B, Barrat F, et al. Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med 2013; 19: 65-73, doi: 10.1038/nm.3042.
https://doi.org/10.1038/nm.3042...
) showed that pancreatic beta cell death increases tissue migration and activation of B lymphocytes, neutrophils, macrophages, and plasmacytoid DC in young female non-obese diabetic (NOD) mice. Neutrophils cultivated in vitro in highly concentrated glucose medium developed SASP, however, it is still unclear whether glucose-induced senescence impairs neutrophil extracellular traps (NET) release (3535. Joshi MB, Lad A, Prasad ASB, Balakrishnan A, Ramachandra L, Satyamoorthy K. High glucose modulates IL-6 mediated immune homeostasis through impeding neutrophil extracellular trap formation. FEBS Lett 2013; 587: 2241-2246, doi: 10.1016/j.febslet.2013.05.053.
https://doi.org/10.1016/j.febslet.2013.0...
), oxidative burst (3636. Marhoffer W, Stein M, Schleinkofer L, Federlin K. Evidence of ex vivo and in vitro impaired neutrophil oxidative burst and phagocytic capacity in type 1 diabetes mellitus. Diabetes Res Clin Pract 1983; 19: 183-188, doi: 10.1016/0168-8227(93)90112-I.
https://doi.org/10.1016/0168-8227(93)901...
), and phagocytic activity (3737. Wilson RM, Reeves WG. Neutrophil phagocytosis and killing in insulin-dependent diabetes. Clin Exp Immunol 1986; 63: 478-484.). Although neutrophil exhaustion may impede T1D progression, it would also increase the risk of infectious diseases, as observed in diabetic patients (3838. Kummer U, Zobeley J, Brasen JC, Fahmy R, Kindzelskii AL, Petty AR, et al. Elevated glucose concentrations promote receptor-independent activation of adherent human neutrophils: an experimental and computational approach. Biophys J 2007; 92: 2597-2607, doi: 10.1529/biophysj.106.086769.
https://doi.org/10.1529/biophysj.106.086...
).

Hyperglycemia-induced SASP amplifies diabetes-related endovascular and tissue inflammation and insulin resistance, and inhibits extracellular matrix production, thus creating a vicious circle. T1D SASP induces proinflammatory M1 macrophages maturation via NF-κB activation and increases reactive oxygen species (ROS) production and intracellular acidosis. Furthermore, histopathological analysis of diabetic wounds shows a protracted population of M1 phenotype-polarized senescent macrophages and a low expression of NLRP3, caspase1, and IL-1 (3939. Zhang X, Dai J, Li L, Chen H, Chai Y. NLRP3 Inflammasome expression and signaling in human diabetic wounds and in high glucose induced macrophages. J Diabetes Res 2017; 2017: 5281358, doi: 10.1155/2017/5281358.
https://doi.org/10.1155/2017/5281358...
). Interestingly, transcriptome of M1 phenotype hyperglycemic medium-induced THP-1 cells revealed a clear SASP-like signature, as IL-1α, IL-6, IL-8, PAI-1, TGF-β, TNF-α, MCP-1, ICAM-1, and IGFBP6 gene expression were strikingly upregulated (4040. Prattichizzo F, De Nigris V, Mancuso E, Spiga R, Giuliani A, Matacchione G, et al. Short-term sustained hyperglycaemia fosters an archetypal senescence-associated secretory phenotype in endothelial cells and macrophages. Redox Biol 2018; 15: 170-181, doi: 10.1016/j.redox.2017.12.001.
https://doi.org/10.1016/j.redox.2017.12....
). The histopathology analysis of diabetic mouse incisional wounds revealed a chronic inflammatory infiltrate enriched with senescent C-X-C motif chemokine receptor 2 (CXCR2)-positive macrophages (4141. Wang Z, Shi C. Cellular senescence is a promising target for chronic wounds: a comprehensive review. Burns Trauma 2020; 8: tkaa021, doi: 10.1093/burnst/tkaa021.
https://doi.org/10.1093/burnst/tkaa021...
). CXCR2 is a pro-fibrotic inflammatory chemokine receptor associated with SASP in primary human dermal fibroblasts (4242. Wilkinson HN, Clowes C, Banyard KL, Matteuci P, Mace KA, Hardman MJ. Elevated local senescence in diabetic wound healing is linked to pathological repair via CXCR2. J Invest Dermatol 2019; 139: 1171-1181.e6, doi: 10.1016/j.jid.2019.01.005.
https://doi.org/10.1016/j.jid.2019.01.00...
). Immune senescence is a primary determinant of diabetic wound healing failure and closely linked to diabetic complications, which are a major cause of morbidity and shortened lifespan (4040. Prattichizzo F, De Nigris V, Mancuso E, Spiga R, Giuliani A, Matacchione G, et al. Short-term sustained hyperglycaemia fosters an archetypal senescence-associated secretory phenotype in endothelial cells and macrophages. Redox Biol 2018; 15: 170-181, doi: 10.1016/j.redox.2017.12.001.
https://doi.org/10.1016/j.redox.2017.12....
).

Rheumatoid arthritis (RA)

Monocytes and neutrophils have emerged as key players in synovial inflammation and cartilage damage. Senescent monocytes have gained attention due to their shorter telomere length and mainly develop a non-classical (CD14dimCD16bright) pro-inflammatory phenotype (4343. Yang X, Chang Y, Wei W. Emerging role of targeting macrophages in rheumatoid arthritis: focus on polarization, metabolism and apoptosis. Cell Prolif 2020; 53: e12854, doi: 10.1111/cpr.12854.
https://doi.org/10.1111/cpr.12854...
). Notably, senescent non-classical monocytes express chemokine receptors that facilitate their migration to inflamed tissues and senescence-associated β-galactosidase (4444. Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc 2009; 4: 1798-1806, doi: 10.1038/nprot.2009.191.
https://doi.org/10.1038/nprot.2009.191...
).

Neutrophils, on the other hand, may play a crucial role in inducing a senescent phenotype in neighboring cells, including monocytes. As their involvement in cartilage damage and destruction is attributed to the release of ROS (4545. Alarcon MF, McLaren Z, Wright HL. Neutrophils in the pathogenesis of rheumatoid arthritis and systemic Lupus erythematosus: same foe different M.O. Front Immunol 2021; 12: 649693-649693, doi: 10.3389/fimmu.2021.649693.
https://doi.org/10.3389/fimmu.2021.64969...
) and telomeres are particularly sensitive to oxidative stress, ROS release by RA neutrophils may induce neighboring cell senescence by significant telomere shortening (4646. Saretzki G, Murphy MP, von Zglinicki T. MitoQ counteracts telomere shortening and elongates lifespan of fibroblasts under mild oxidative stress. Aging Cell 2003; 2: 141-143, doi: 10.1046/j.1474-9728.2003.00040.x.
https://doi.org/10.1046/j.1474-9728.2003...
). When co-cultured with neutrophils, human fibroblasts rapidly express senescence markers and shorter telomeres compared with control fibroblasts cultured alone. Moreover, the rate of dysfunctional telomeres, characterized by their association with DNA damage response factors (namely, telomere-associated foci, p16Ink4a, and p21) were increased in neutrophil-induced senescent cells. Noteworthy, premature senescence and telomere damage were prevented when extracellular ROS were scavenged by adding recombinant catalase (4747. Lagnado A, Leslie J, Ruchaud-Sparagano MH, Victorelli S, Hirsova P, Ogrodnik M, et al. Neutrophils induce paracrine telomere dysfunction and senescence in ROS-dependent manner. EMBO J 2021; 40: e106048, doi: 10.15252/embj.2020106048.
https://doi.org/10.15252/embj.2020106048...
).

Systemic lupus erythematosus (SLE)

Similar to RA, neutrophil dysregulation also contributes to SLE pathogenesis. NET release probably exposes DNA and encrypted nuclear proteins to the immune system in SLE, culminating with autoantibody production, such as anti-double stranded DNA and anti-acetylated/methylated histones (4545. Alarcon MF, McLaren Z, Wright HL. Neutrophils in the pathogenesis of rheumatoid arthritis and systemic Lupus erythematosus: same foe different M.O. Front Immunol 2021; 12: 649693-649693, doi: 10.3389/fimmu.2021.649693.
https://doi.org/10.3389/fimmu.2021.64969...
).

SLE neutrophils usually peak faster and produce higher ROS levels than those from healthy individuals (4848. Perazzio SF, Salomão R, Silva NP, Andrade LE. Increased neutrophil oxidative burst metabolism in systemic Lupus erythematosus. Lupus 2012; 21: 1543-1551, doi: 10.1177/0961203312461060.
https://doi.org/10.1177/0961203312461060...
). Nevertheless, neutrophils from active SLE patients paradoxically produce lower ROS levels than those from individuals with inactive SLE, probably due to neutrophil exhaustion (4949. Elloumi N, Mansour RB, Marzouk S, Mseddi M, Fakhfakh R, Gargouri B, et al. Differential reactive oxygen species production of neutrophils and their oxidative damage in patients with active and inactive systemic lupus erythematosus. Immunol Lett 2017; 184: 1-6, doi: 10.1016/j.imlet.2017.01.018.
https://doi.org/10.1016/j.imlet.2017.01....
). Conversely, peripheral PD-L1-expressing neutrophil count of SLE patients with active or severe disease is higher than those with inactive and milder conditions (5050. Luo Q, Huang Z, Ye J, Deng Y, Fang L, Li X, et al. PD-L1-expressing neutrophils as a novel indicator to assess disease activity and severity of systemic Lupus erythematosus. Arthritis Res Ther 2016; 18: 47-47, doi: 10.1186/s13075-016-0942-0.
https://doi.org/10.1186/s13075-016-0942-...
).

Low-density granulocytes (LDG), a specific subset of neutrophils, have emerged as a captivating area of investigation in the field of SLE. LDG rely on the lower density compared to conventional neutrophils, with isolation typically achieved by density gradient centrifugation techniques (5151. Herteman N, Vargas A, Lavoie JP. Characterization of circulating low-density neutrophils intrinsic properties in healthy and asthmatic horses. Sci Rep 2017; 7: 7743, doi: 10.1038/s41598-017-08089-5.
https://doi.org/10.1038/s41598-017-08089...
). LDG can exhibit an enhanced pro-inflammatory profile, characterized by heightened cytokine synthesis including TNF-α, IL-6, IL-8, and IFN (5252. Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, et al. A distinct subset of proinflammatory neutrophils isolated from patients with systemic Lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol 2010; 184: 3284-3297, doi: 10.4049/jimmunol.0902199.
https://doi.org/10.4049/jimmunol.0902199...
) and are commonly elevated in peripheral blood of active lupus patients, especially those presenting vasculitis, cutaneous manifestations, or high anti-double stranded DNA titers (5252. Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, et al. A distinct subset of proinflammatory neutrophils isolated from patients with systemic Lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol 2010; 184: 3284-3297, doi: 10.4049/jimmunol.0902199.
https://doi.org/10.4049/jimmunol.0902199...
). Additionally, LDG display an increased propensity for spontaneous production of NET (5353. van der Hoogen L, van der Linden M, Meyaard L, Ruth DEFS, van Roon JA, Radstake TR. Neutrophil extracellular traps and low-density granulocytes are associated with the interferon signature in systemic lupus erythematosus, but not in antiphospholipid syndrome. Ann Rheum Dis 2020; 79: e135, doi: 10.1136/annrheumdis-2019-215781.
https://doi.org/10.1136/annrheumdis-2019...
), further contributing to the pathogenesis of SLE when taken together (5151. Herteman N, Vargas A, Lavoie JP. Characterization of circulating low-density neutrophils intrinsic properties in healthy and asthmatic horses. Sci Rep 2017; 7: 7743, doi: 10.1038/s41598-017-08089-5.
https://doi.org/10.1038/s41598-017-08089...
). As neutrophil hyperactivation induces degranulation and NET release, thus reducing cell density and resulting in exhaustion, one can hypothesize that LDG may play a role in neutrophil senescence. However, further comprehensive investigations are warranted to fully understand the role of LDG in SLE pathogenesis, particularly regarding their impact on senescence or exhaustion profiles in innate immune cells (5454. Hong CW. Current understanding in neutrophil differentiation and heterogeneity. Immune Netw 2017; 17: 298-306, doi: 10.4110/in.2017.17.5.298.
https://doi.org/10.4110/in.2017.17.5.298...
).

Immune-mediated necrotizing myopathy (IMNM)

IMNM is a specific form of autoimmune myopathy distinguished by pronounced weakness in the proximal muscles, myofiber necrosis, and infiltration of inflammatory cells as neutrophils and macrophages (5555. Pinal-Fernandez I, Casal-Dominguez M, Mammen AL. Immune-mediated necrotizing myopathy. Curr Rheumatol Rep 2018; 20: 21, doi: 10.1007/s11926-018-0732-6.
https://doi.org/10.1007/s11926-018-0732-...
). Knauss et al. (5656. Knauss S, Preusse C, Allenbach Y, Leonard-Louis S, Touat M, Fischer N, et al. PD1 pathway in immune-mediated myopathies. Neurol Neuroimmunol Neuroinflamm 2019; 6: e558, doi: 10.1212/NXI.0000000000000558.
https://doi.org/10.1212/NXI.000000000000...
) showed the high expression of PD-1, LAG-3, and TIM-3, a classic T cell exhausted phenotype, in muscle specimens extracted from 12 IMNM patients. Moreover, the authors also detected high expression of PD-L1 on macrophages and PD-L2 on myofibers. Interestingly, PD-L2 staining in myofibers was partially overlapping PD1 staining on CD3+ T lymphocytes, implicating the formation of the so called “immunologic synapses” and a potential role of PD-L2/PD1 interaction in modulating T-cell activation and macrophages cells.

PD-1 contributes to skeletal muscle regeneration by facilitating the transition of macrophages from a proinflammatory to an anti-inflammatory phenotype (5757. Zhuang S, Russell A, Guo Y, Xu Y, Xiao W. IFN-γ blockade after genetic inhibition of PD-1 aggravates skeletal muscle damage and impairs skeletal muscle regeneration. Cell Mol Biol Lett 2023; 28: 27, doi: 10.1186/s11658-023-00439-8.
https://doi.org/10.1186/s11658-023-00439...
). IFN-γ, on the other hand, stimulates the formation of proinflammatory macrophages, which seem to impede myogenesis in vitro (5757. Zhuang S, Russell A, Guo Y, Xu Y, Xiao W. IFN-γ blockade after genetic inhibition of PD-1 aggravates skeletal muscle damage and impairs skeletal muscle regeneration. Cell Mol Biol Lett 2023; 28: 27, doi: 10.1186/s11658-023-00439-8.
https://doi.org/10.1186/s11658-023-00439...
). However, Zhuang et al. (5757. Zhuang S, Russell A, Guo Y, Xu Y, Xiao W. IFN-γ blockade after genetic inhibition of PD-1 aggravates skeletal muscle damage and impairs skeletal muscle regeneration. Cell Mol Biol Lett 2023; 28: 27, doi: 10.1186/s11658-023-00439-8.
https://doi.org/10.1186/s11658-023-00439...
) surprisingly revealed that blocking IFN-γ signaling of PD-1 knockout models actually exacerbated inflammation in the injured muscle, impeded muscle regeneration, and intensified muscle fibrosis. This detrimental effect was attributed to the inhibition of macrophage infiltration and transition from a proinflammatory to an anti-inflammatory state, associated with an increased influx of neutrophils into the muscle tissue.

Taken together, all these pieces of evidence support the hypothesis that anti-PD-L1 therapy holds the potential to ameliorate inflammation in IMNM. However, this hypothesis is currently being investigated, and preliminary data thus far have yielded discouraging results. Recent advancements in the field have led to a growing utilization of immune checkpoint inhibitors (ICI) across a wide range of malignancies. Nevertheless, this therapeutic strategy has unveiled a novel spectrum of adverse effects, especially in myositis induced by ICI therapy, which has an high mortality rate when co-occurring with other autoimmune manifestations such as myocarditis and myasthenia gravis (5858. Dalakas MC. Neurological complications of immune checkpoint inhibitors: what happens when you ‘take the brakes off' the immune system. Ther Adv Neurol Disord 2018; 11: 1756286418799864, doi: 10.1177/1756286418799864.
https://doi.org/10.1177/1756286418799864...
).

Multiple sclerosis

Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system in young adults (5959. Filippi M, Bar-Or A, Piehl F, Preziosa P, Solari A, Vukusic S, et al. Multiple sclerosis. Nat Rev Dis Primers 2018; 4: 43, doi: 10.1038/s41572-018-0041-4.
https://doi.org/10.1038/s41572-018-0041-...
). The global population of individuals over 65 years old with MS is on the rise as the life expectancy for those living with MS has improved (6060. Vaughn CB, Jakimovski D, Kavak KS, Ramanathan M, Benedict RHB, et al. Epidemiology and treatment of multiple sclerosis in elderly populations. Nat Rev Neurol 2019; 15: 329-342, doi: 10.1038/s41582-019-0183-3.
https://doi.org/10.1038/s41582-019-0183-...
). With this growing awareness, the challenges associated with aging, immunosenescence, and MS are also being recognized. These challenges also include a limited understanding of the long-term effects of disease-modifying therapies.

Two distinguished phases of MS's pathophysiology are recognized: early inflammatory and progressive phases (6161. Perdaens O, van Pesch V. Molecular mechanisms of immunosenescene and inflammaging: relevance to the immunopathogenesis and treatment of multiple sclerosis. Front Neurol 2021; 12: 811518, doi: 10.3389/fneur.2021.811518.
https://doi.org/10.3389/fneur.2021.81151...
). While during the first phase, the blood-brain barrier (BBB) is disrupted, allowing peripheral adaptive immune cell infiltration into the central nervous system (CNS), during the progressive phase, T and B cells influx is reduced as the BBB is closed and the inflammation is sustained by innate CNS-resident microglia and astrocytes. These cells produce cytokines, such as TNF-α and IL-6, and release ROS, culminating with myelin damage. While microglia and astrocytes become primed into a pro-inflammatory phenotype, their phagocytic activity is reduced and they progressively acquire a clear SASP. Improper clearance of myelin debris occurs, and oligodendrocyte progenitor cell recruitment and differentiation become less effective (6262. Sim FJ, Zhao C, Penderis J, Franklin RJ. The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J Neurosci 2002; 22: 2451-2459, doi: 10.1523/JNEUROSCI.22-07-02451.2002.
https://doi.org/10.1523/JNEUROSCI.22-07-...
). These successive events become self-sustained and are amplified by senescent processes, resulting in a significant oxidative burst that leads to mitochondrial DNA damage-induced dysfunction, energy failure, and axonal loss. Moreover, cell cycle arrest and phenotypic changes in senescent cells might affect their functions and their regenerative capacity (6363. Koutsoudaki PN, Papadopoulos D, Passias PG, Koutsoudaki P, Gorgoulis VG. Cellular senescence and failure of myelin repair in multiple sclerosis. Mech Ageing Dev 2020; 192: 111366, doi: 10.1016/j.mad.2020.111366.
https://doi.org/10.1016/j.mad.2020.11136...
).

Immune exhaustion and senescence-blocking factors of innate effector components

Modern lifestyle with a high-fat diet and excessive alcohol consumption, obesity, sedentary lifestyle, and smoking are important causes of low-grade chronic systemic inflammation, which puts the immune homeostasis in a state called inflammaging (6464. Calder PC, Ahluwalia N, Brouns F, Buetler T, Clement K, Cunningham K, et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br J Nutr 2011; 106: 3, S5-S78, doi: 10.1017/S0007114511005460.
https://doi.org/10.1017/S000711451100546...
,6565. Calder PC, Bosco N, Bourdet-Sicard R, Capuron L, Delzenne N, Doré J, et al. Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res Rev 2017; 40: 95-119, doi: 10.1016/j.arr.2017.09.001.
https://doi.org/10.1016/j.arr.2017.09.00...
). Obesity, hyperglycemia (6666. Morey M, O'Gaora P, Pandit A, Hélary C. Hyperglycemia acts in synergy with hypoxia to maintain the pro-inflammatory phenotype of macrophages. PloS One 2019; 14: e0220577, doi: 10.1371/journal.pone.0220577.
https://doi.org/10.1371/journal.pone.022...
), and sedentary lifestyle (6767. Brugnara L, Murillo S, Novials A, Rojo-Martínez G, Soriguer F, Goday A, et al. Low physical activity and its association with diabetes and other cardiovascular risk factors: a nationwide, population-based study. PloS One 2016; 11: e0160959, doi: 10.1371/journal.pone.0160959.
https://doi.org/10.1371/journal.pone.016...
) increase proinflammatory cytokine production, such as IL-6 and TNF-α, and shift the memory:naive T cell ratio towards mature forms in humans (6868. Aguilar EG, Murphy WJ. Obesity induced T cell dysfunction and implications for cancer immunotherapy. Curr Opin Immunol 2018; 51: 181-186, doi: 10.1016/j.coi.2018.03.012.
https://doi.org/10.1016/j.coi.2018.03.01...
). A similar immunophenotype has been described in peripheral T cells of mice and other chordates (6969. Messaoudi I, Warner J, Fischer M, Park B, Hill B, Mattison J, et al. Delay of T cell senescence by caloric restriction in aged long-lived nonhuman primates. Proc Natl Acad Sci USA 2006; 103: 19448-19453, doi: 10.1073/pnas.0606661103.
https://doi.org/10.1073/pnas.0606661103...
). Fat deposits stimulate neutrophil (7070. Elgazar-Carmon V, Rudich A, Hadad N, Levy R. Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res 2008; 49: 1894-1903, doi: 10.1194/jlr.M800132-JLR200.
https://doi.org/10.1194/jlr.M800132-JLR2...
,7171. Svahn SL, Gutiérrez S, Ulleryd MA, Nookaew I, Osla V, Beckman F, et al. Dietary polyunsaturated fatty acids promote neutrophil accumulation in the spleen by altering chemotaxis and delaying cell death. Infection Immun 2019; 87: e00270-e00319, doi: 10.1128/IAI.00270-19.
https://doi.org/10.1128/IAI.00270-19...
), macrophage, and T cell recruitment into the adipose tissue, which may dysregulate immune response and accelerate immune senescence and exhaustion (7272. Kado T, Nawaz A, Takikawa A, Usui I, Tobe K. Linkage of CD8(+) T cell exhaustion with high-fat diet-induced tumourigenesis. Sci Rep 2019; 9: 12284-12284, doi: 10.1038/s41598-019-48678-0.
https://doi.org/10.1038/s41598-019-48678...
).

To investigate the association between obesity and senescent cell accumulation, Ogrodnik et al. (7373. Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, et al. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab 2019; 29: 1061-1077.e8, doi: 10.1016/j.cmet.2018.12.008.
https://doi.org/10.1016/j.cmet.2018.12.0...
) studied the role of senescence in obesity-related neuropsychiatric disorders of the INK-ATTAC mouse model, which allows p16Ink4a-expressing cell elimination. The researchers found that obesity-induced senescent glial cells in the vicinity of the lateral ventricle, a region associated with adult neurogenesis, exhibited excessive fat deposits. Interestingly, neurogenesis was restored by clearing out senescent cells from leptin knockout mice fed with a high-fat diet.

In addition, adipocyte hyperplasia and hypertrophy increase adipocyte hypoxia, fatty acid metabolic dysregulation, chemokine secretion, adipocyte cell death, and inflammatory cell recruitment (7474. Khan S, Chan YT, Revelo XS, Winer DA. The immune landscape of visceral adipose tissue during obesity and aging. Front Endocrinol (Lausanne) 2020; 11: 267, doi: 10.3389/fendo.2020.00267.
https://doi.org/10.3389/fendo.2020.00267...
), ultimately inducing inflammaging and cell SASP, and generating a positive feedback loop that contributes to local and systemic inflammation. Studies demonstrated that both obese mice and human adipose tissues recruit pathogenic autoantibodies-secreting B cells (7575. Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 2011; 17: 610-617, doi: 10.1038/nm.2353.
https://doi.org/10.1038/nm.2353...
) and favor proinflammatory cytokine secretion during aging, generating SASP senescent cells (7676. Ovadya Y, Landsberger T, Leins H, Vadai E, Gal H, Biran A, et al. Impaired immune surveillance accelerates accumulation of senescent cells and aging. Nat Commun 2018; 9: 5435, doi: 10.1038/s41467-018-07825-3.
https://doi.org/10.1038/s41467-018-07825...
). Furthermore, a high-fat diet also upregulated p16INK4a in cortical and hippocampal mouse neurons (7777. Tarantini S, Valcarcel-Ares MN, Yabluchanskiy A, Tucsek Z, Hertelendy P, Kiss T, et al. Nrf2 Deficiency exacerbates obesity-induced oxidative stress, neurovascular dysfunction, blood-brain barrier disruption, neuroinflammation, amyloidogenic gene expression, and cognitive decline in mice, mimicking the aging phenotype. J Gerontol A Biol Sci Med Sci 2018; 73: 853-863, doi: 10.1093/gerona/glx177.
https://doi.org/10.1093/gerona/glx177...
) and hepatocytes (7878. Zhang X, Xu GB, Zhou D, Pan YX. High-fat diet modifies expression of hepatic cellular senescence gene p16(INK4a) through chromatin modifications in adult male rats. Genes Nutr 2018; 13: 6, doi: 10.1186/s12263-018-0595-5.
https://doi.org/10.1186/s12263-018-0595-...
). Interestingly, possible functional impairments in adipose tissue neutrophils induced by aging are still unclear.

Sedentarism also induces inflammaging (7979. Kaastrup K, Grønbæk K. The impact of sedentary lifestyle, high-fat diet, tobacco smoke, and alcohol intake on the hematopoietic stem cell niches. HemaSphere 2021; 5: e615, doi: 10.1097/HS9.0000000000000615.
https://doi.org/10.1097/HS9.000000000000...
), but may be reverted by moderate physical activity (8080. Englund DA, Sakamoto AE, Fritsche CM, Heeren AA, Zhang X, Kotajarvi BR, et al. Exercise reduces circulating biomarkers of cellular senescence in humans. Aging Cell 2021; 20: e13415, doi: 10.1111/acel.13415.
https://doi.org/10.1111/acel.13415...
). Recently, a 10-week program of regular walking increased neutrophil phagocytic and chemotactic activity after bacterial stimuli in elderly adults with rheumatoid arthritis (8181. Bartlett DB, Willis LH, Slentz CA, Hoselton A, Kelly L, Huebner JL, et al. Ten weeks of high-intensity interval walk training is associated with reduced disease activity and improved innate immune function in older adults with rheumatoid arthritis: a pilot study. Arthritis Res Ther 2018; 20: 127, doi: 10.1186/s13075-018-1624-x.
https://doi.org/10.1186/s13075-018-1624-...
). Moreover, neutrophil chemotactic activity of healthy elderly individuals who walked at least 10,000 steps per day was higher than that of age-matched adults who walked only 5000 steps per day (8282. Bartlett DB, Fox O, McNulty CL, Greenwood HL, Murphy L, Sapey E, et al. Habitual physical activity is associated with the maintenance of neutrophil migratory dynamics in healthy older adults. Brain Behav Immun 2016; 56: 12-20, doi: 10.1016/j.bbi.2016.02.024.
https://doi.org/10.1016/j.bbi.2016.02.02...
). Similarly, corrective lifestyle interventions that prevent sedentarism and improve diet quality have the potential to prevent obesity, inflammation, aging, and the exhaustion process (7272. Kado T, Nawaz A, Takikawa A, Usui I, Tobe K. Linkage of CD8(+) T cell exhaustion with high-fat diet-induced tumourigenesis. Sci Rep 2019; 9: 12284-12284, doi: 10.1038/s41598-019-48678-0.
https://doi.org/10.1038/s41598-019-48678...
). Regular moderate-intensity physical activity suppresses IL-6, TNFα (8383. Keller C, Keller P, Giralt M, Hidalgo J, Pedersen BK. Exercise normalises overexpression of TNF-alpha in knockout mice. Biochem Biophys Res Commun 2004; 321: 179-182, doi: 10.1016/j.bbrc.2004.06.129.
https://doi.org/10.1016/j.bbrc.2004.06.1...
), and IL-1β (8484. Nakajima K, Takeoka M, Mori M, Hashimoto S, Sakurai A, Nose H, et al. Exercise effects on methylation of ASC gene. Int J Sports Med 2010; 31: 671-675, doi: 10.1055/s-0029-1246140.
https://doi.org/10.1055/s-0029-1246140...
), increases telomere length, and downregulates p16INK4a (8585. Werner C, Fürster T, Widmann T, Pöss J, Roggia C, Hanhoun M, et al. Physical exercise prevents cellular senescence in circulating leukocytes and in the vessel wall. Circulation 2009; 120: 2438-2447, doi: 10.1161/CIRCULATIONAHA.109.861005.
https://doi.org/10.1161/CIRCULATIONAHA.1...
), thus attenuating the hallmarks of aging.

New approaches for anti-senescence therapy

Two distinct therapies targeting senescence have been identified: senolytic agents, a promising experimental class of drugs that selectively induce senescent cells to undergo apoptosis, like navitoclax, dasatinib plus quercetin, 17-DMAG (17-dimethylaminoethylamino-17-demethoxygeldanamycin), and senostatic agents, like ruxolitinib, rapamycin, and metformin, which inhibit SASP signaling pathways (8686. Song S, Lam EW, Tchkonia T, Kirkland JL, Sun Y. Senescent cells: emerging targets for human aging and age-related diseases. Trends Biochem Sci 2020; 45: 578-592, doi: 10.1016/j.tibs.2020.03.008.
https://doi.org/10.1016/j.tibs.2020.03.0...
). On the other hand, probiotic bacteria in humans seem to present beneficial effects as anti-senescence therapy (8787. Boyajian JL, Ghebretatios M, Schaly S, Islam P, Prakash S. Microbiome and Human aging: probiotic and prebiotic potentials in longevity, skin health and cellular senescence. Nutrients 2021; 13: 4550, doi: 10.3390/nu13124550.
https://doi.org/10.3390/nu13124550...
). A 4-week high-fiber diet with 5% inulin program suppressed IL-1β, TNFα, IL-6, NLRP3, and TLR4 gene expression, induced IL1RN anti-inflammatory microglial gene expression, improved aging-associated neuroinflammation, and altered microbiome by reducing Ruminococcus spp and Rikenellaceae spp in adult and aged BALB/C mice (8888. Matt SM, Allen JM, Lawson MA, Mailing LJ, Woods JA, Johnson RW. Butyrate and dietary soluble fiber improve neuroinflammation associated with aging in mice. Front Immunol 2018; 9: 1832, doi: 10.3389/fimmu.2018.01832.
https://doi.org/10.3389/fimmu.2018.01832...
). However, more studies in the area are still needed, focusing especially on anti-innate immunity senescence treatment.

New approaches for anti-exhaustion therapy

Checkpoint inhibitors emerged as a transformative anti-tumor therapeutic strategy in oncologic patients by facilitating adaptive immunity activation but have also been considered an anti-exhaustion alternative therapy lately. A mouse model of cancer with B16F10 cell transplant demonstrated SASP with PD-1, TIM3, and LAG3 overexpression on CD8+ and CD4+ T cells, which were reversed after PD1 blockade (8989. Fu J, Yu A, Xiao X, Tang J, Zu X, Chen W, et al. CD4(+) T cell exhaustion leads to adoptive transfer therapy failure which can be prevented by immune checkpoint blockade. Am J Cancer Res 2020; 10: 4234-4250.). Anti-TIM3 also reversed SASP from a cecal ligation mouse model of sepsis by upregulating TLR4-induced NF-κB pathway activation in LPS-stimulated peritoneal macrophages (9090. Yang X, Jiang X, Chen G, Xiao Y, Geng S, Kang C, et al. T cell Ig mucin-3 promotes homeostasis of sepsis by negatively regulating the TLR response. J Immunol 2013; 190: 2068-2079, doi: 10.4049/jimmunol.1202661.
https://doi.org/10.4049/jimmunol.1202661...
). Certainly, additional studies are still needed to determine whether these agents could be critical in cell recovery.

Conclusion

Here we have briefly described the current state of the research on immune senescence and exhaustion, uncovering the main pathways affecting innate immunity in the context of autoimmune diseases. We highlighted key SASP-induced components such as PD1, TICAM2, TIM3, Β-galactosidase (β-GAL), p16ink4a, and lamin B. NF-κB and IFN pathways also play a pivotal, though intricate, role in driving cell SASP. In addition, we discussed possible targeted interventions able to block immune senescence (senolytic agents like navitoclax, dasatinib plus quercetin, and 17-DMAG; and senostatic agents like ruxolitinib, rapamycin, and metformin) and exhaustion (especially checkpoint inhibitors). Innate immunity is our first line of defense and is mainly composed by short-lived cells, which may pose challenges in studying exhaustion and senescence due to their gradual nature, but the field holds significant untapped potential. Much remains to be explored in this domain, and it is evident that further studies are imperative to unravel the pathophysiological intricacies associated with these molecules and pathways. These endeavors may in turn contribute to the identification of novel therapeutic targets and improve our understanding of autoimmune diseases.

Acknowledgments

This study was supported by Fundação de Amparo è Pesquisa do Estado de São Paulo (FAPESP, grant #2018/00555-6). A.L.S. Cunha was also supported by FAPESP (grant #2019/23469-0).

References

  • 1
    Frizinsky S, Haj-Yahia S, Maayan DM, Lifshitz Y, Maoz-Segal R, Offengenden I, et al. The innate immune perspective of autoimmune and autoinflammatory conditions. Rheumatology (Oxford) 2019; 58: vi1-vi8, doi: 10.1093/rheumatology/kez387.
    » https://doi.org/10.1093/rheumatology/kez387
  • 2
    Akbar AN, Henson SM. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat Rev Immunol 2011; 11: 289-295, doi: 10.1038/nri2959.
    » https://doi.org/10.1038/nri2959
  • 3
    Aiello A, Farzaneh F, Candore G, Caruso C, Davinelli S, Gambino CM, et al. Immunosenescence and its hallmarks: how to oppose aging strategically? A review of potential options for therapeutic intervention. Front Immunol 2019; 10: 2247, doi: 10.3389/fimmu.2019.02247.
    » https://doi.org/10.3389/fimmu.2019.02247
  • 4
    Verdon DJ, Mulazzani M, Jenkins MR. Cellular and molecular mechanisms of CD8(+) T cell differentiation, dysfunction and exhaustion. Int J Mol Sci 2020; 21: 7357, doi: 10.3390/ijms21197357.
    » https://doi.org/10.3390/ijms21197357
  • 5
    Huang X, Venet F, Wang YL, Lepape A, Yuan Z, Chen Y, et al. PD-1 expression by macrophages plays a pathologic role in altering microbial clearance and the innate inflammatory response to sepsis. Proc Nat Acad Sci USA 2009; 106: 6303-6303, doi: 10.1073/pnas.0809422106.
    » https://doi.org/10.1073/pnas.0809422106
  • 6
    Phong B, Avery L, Menk AV, Delgoffe GM, Kane LP. cutting edge: murine mast cells rapidly modulate metabolic pathways essential for distinct effector functions. J Immunol 2017; 198: 640-644, doi: 10.4049/jimmunol.1601150.
    » https://doi.org/10.4049/jimmunol.1601150
  • 7
    Jahromi NH, Marchetti L, Moalli F, Duc D, Basso C, Tardent H, et al. Intercellular adhesion molecule-1 (ICAM-1) and ICAM-2 differentially contribute to peripheral activation and CNS entry of autoaggressive Th1 and Th17 cells in experimental autoimmune encephalomyelitis. Front Immunol 2020; 10: 3056, doi: 10.3389/fimmu.2019.03056.
    » https://doi.org/10.3389/fimmu.2019.03056
  • 8
    Araki K, Youngblood B, Ahmed R. Programmed cell death 1-directed immunotherapy for enhancing T-cell function. Cold Spring Harb Symp Quant Biol 2013; 78: 239-247, doi: 10.1101/sqb.78.019869.
    » https://doi.org/10.1101/sqb.78.019869
  • 9
    Golden-Mason L, Palmer BE, Kassam N, Townshend-Bulson L, Livingston S, McMahon BJ, et al. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol 2009; 83: 9122-9130, doi: 10.1128/JVI.00639-09.
    » https://doi.org/10.1128/JVI.00639-09
  • 10
    Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med 2010; 207: 2187-2194, doi: 10.1084/jem.20100643.
    » https://doi.org/10.1084/jem.20100643
  • 11
    Lin R, Zhang Y, Pradhan K, Li L. TICAM2-related pathway mediates neutrophil exhaustion. Sci Rep 2020; 10: 14397, doi: 10.1038/s41598-020-71379-y.
    » https://doi.org/10.1038/s41598-020-71379-y
  • 12
    Zasada M, Lenart M, Rutkowska-Zapała M, Stec M, Durlak W, Grudzień A, et al. Analysis of PD-1 expression in the monocyte subsets from non-septic and septic preterm neonates. PloS One 2017, 8; 12: e0186819, doi: 10.1371/journal.pone.0186819.
    » https://doi.org/10.1371/journal.pone.0186819
  • 13
    Seliger B. Basis of PD1/PD-L1 therapies. J Clin Med 2019; 8: 2168, doi: 10.3390/jcm8122168.
    » https://doi.org/10.3390/jcm8122168
  • 14
    da Silva IP, Gallois A, Jimenez-Baranda S, Khan S, Anderson AC, Kuchroo VK, et al. Reversal of NK-Cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol Res 2014; 2: 410-422, doi: 10.1158/2326-6066.CIR-13-0171.
    » https://doi.org/10.1158/2326-6066.CIR-13-0171
  • 15
    Maue AC, Yager EJ, Swain SL, Woodland DL, Blackman MA, Haynes L. T-cell immunosenescence: lessons learned from mouse models of aging. Trends Immunol 2009; 30: 301-305, doi: 10.1016/j.it.2009.04.007.
    » https://doi.org/10.1016/j.it.2009.04.007
  • 16
    Álvarez S, Braãas F, Sánchez-Conde M, Moreno S, de Quirós JCLB, Muãoz-Fernández MÁ. Frailty, markers of immune activation and oxidative stress in HIV infected elderly. PloS One 2020; 15: e0230339, doi: 10.1371/journal.pone.0230339.
    » https://doi.org/10.1371/journal.pone.0230339
  • 17
    Lazuardi L, Jenewein B, Wolf AM, Pfister G, Tzankov A, Grubeck-Loebenstein B. Age-related loss of naïve T cells and dysregulation of T-cell/B-cell interactions in human lymph nodes. Immunology 2005; 114: 37-43, doi: 10.1111/j.1365-2567.2004.02006.x.
    » https://doi.org/10.1111/j.1365-2567.2004.02006.x
  • 18
    Fagnoni FF, Vescovini R, Passeri G, Bologna G, Pedrazzoni M, Lavagetto G, et al. Shortage of circulating naive CD8+ T cells provides new insights on immunodeficiency in aging. Blood 2000; 95: 2860-2868, doi: 10.1182/blood.V95.9.2860.009k35_2860_2868.
    » https://doi.org/10.1182/blood.V95.9.2860.009k35_2860_2868
  • 19
    Nomellini V, Faunce DE, Gomez CR, Kovacs EJ. An age-associated increase in pulmonary inflammation after burn injury is abrogated by CXCR2 inhibition. J Leukoc Biol 2008; 83: 1493-1501, doi: 10.1189/jlb.1007672.
    » https://doi.org/10.1189/jlb.1007672
  • 20
    Butcher SK, Chahal H, Nayak L, Sinclair A, Henriquez NV, Sapey E, et al. Senescence in innate immune responses: reduced neutrophil phagocytic capacity and CD16 expression in elderly humans. J Leukoc Biol 2001; 70: 881-886, doi: 10.1189/jlb.70.6.881.
    » https://doi.org/10.1189/jlb.70.6.881
  • 21
    Wessels I, Jansen J, Rink L, Uciechowski P. Immunosenescence of polymorphonuclear neutrophils. ScientificWorldJournal 2010; 10: 145-160, doi: 10.1100/tsw.2010.14.
    » https://doi.org/10.1100/tsw.2010.14
  • 22
    Kumari R, Jat P. Mechanisms of cellular senescence: cell cycle arrest and senescence associated secretory phenotype. Front Cell Dev Biol 2021; 9: 645593, doi: 10.3389/fcell.2021.645593.
    » https://doi.org/10.3389/fcell.2021.645593
  • 23
    de Mera-Rodríguez JA, Álvarez-Hernán G, Gaãán Y, Martín-Partido G, Rodríguez-León J, Francisco-Morcillo J. Is senescence-associated β-galactosidase a reliable in vivo marker of cellular senescence during embryonic development? Front Cell Dev Biol 2021; 9: 623175, doi: 10.3389/fcell.2021.623175.
    » https://doi.org/10.3389/fcell.2021.623175
  • 24
    Giacinti C, Giordano A. RB and cell cycle progression. Oncogene 2006; 25: 5220-5227, doi: 10.1038/sj.onc.1209615.
    » https://doi.org/10.1038/sj.onc.1209615
  • 25
    Liu JY, Souroullas GP, Diekman BO, Krishnamurthy J, Hall BM, Sorrentino JA, et al. Cells exhibiting strong p16 (INK4a) promoter activation in vivo display features of senescence. Proc Natl Acad Sci USA 2019; 116: 2603-2611, doi: 10.1073/pnas.1818313116.
    » https://doi.org/10.1073/pnas.1818313116
  • 26
    Shimi T, Butin-Israeli V, Adam SA, Hamanaka RB, Goldman AE, Lucas CA, et al. The role of nuclear lamin B1 in cell proliferation and senescence. Genes Dev 2011; 25: 2579-2593, doi: 10.1101/gad.179515.111.
    » https://doi.org/10.1101/gad.179515.111
  • 27
    Wang AS, Ong PF, Chojnowski A, Clavel C, Dreesen O. Loss of lamin B1 is a biomarker to quantify cellular senescence in photoaged skin. Sci Rep 2017; 7: 15678, doi: 10.1038/s41598-017-15901-9.
    » https://doi.org/10.1038/s41598-017-15901-9
  • 28
    Freund A, Laberge RM, Demaria M, Campisi J. Lamin B1 loss is a senescence-associated biomarker. Mol Biol Cell 2012; 23: 2066-2075, doi: 10.1091/mbc.e11-10-0884.
    » https://doi.org/10.1091/mbc.e11-10-0884
  • 29
    Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol 2010; 184: 2518-2527, doi: 10.4049/jimmunol.0901022.
    » https://doi.org/10.4049/jimmunol.0901022
  • 30
    Bella SD, Bierti L, Presicce P, Arienti R, Valenti M, Saresella M, et al. Peripheral blood dendritic cells and monocytes are differently regulated in the elderly. Clin Immunol 2007; 122: 220-228, doi: 10.1016/j.clim.2006.09.012.
    » https://doi.org/10.1016/j.clim.2006.09.012
  • 31
    Orjalo AV, Bhaumik D, Gengler BK, Scott GK, Campisi J. Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc Natl Acad Sci USA 2009; 106: 17031-17036, doi: 10.1073/pnas.0905299106.
    » https://doi.org/10.1073/pnas.0905299106
  • 32
    Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol 2013; 13: 875-887, doi: 10.1038/nri3547.
    » https://doi.org/10.1038/nri3547
  • 33
    McHugh D, Gil J. Senescence and aging: Causes, consequences, and therapeutic avenues. J Cell Biol 2017; 217: 65-77, doi: 10.1083/jcb.201708092.
    » https://doi.org/10.1083/jcb.201708092
  • 34
    Diana J, Simoni Y, Furio L, Beaudoin L, Agerberth B, Barrat F, et al. Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med 2013; 19: 65-73, doi: 10.1038/nm.3042.
    » https://doi.org/10.1038/nm.3042
  • 35
    Joshi MB, Lad A, Prasad ASB, Balakrishnan A, Ramachandra L, Satyamoorthy K. High glucose modulates IL-6 mediated immune homeostasis through impeding neutrophil extracellular trap formation. FEBS Lett 2013; 587: 2241-2246, doi: 10.1016/j.febslet.2013.05.053.
    » https://doi.org/10.1016/j.febslet.2013.05.053
  • 36
    Marhoffer W, Stein M, Schleinkofer L, Federlin K. Evidence of ex vivo and in vitro impaired neutrophil oxidative burst and phagocytic capacity in type 1 diabetes mellitus. Diabetes Res Clin Pract 1983; 19: 183-188, doi: 10.1016/0168-8227(93)90112-I.
    » https://doi.org/10.1016/0168-8227(93)90112-I
  • 37
    Wilson RM, Reeves WG. Neutrophil phagocytosis and killing in insulin-dependent diabetes. Clin Exp Immunol 1986; 63: 478-484.
  • 38
    Kummer U, Zobeley J, Brasen JC, Fahmy R, Kindzelskii AL, Petty AR, et al. Elevated glucose concentrations promote receptor-independent activation of adherent human neutrophils: an experimental and computational approach. Biophys J 2007; 92: 2597-2607, doi: 10.1529/biophysj.106.086769.
    » https://doi.org/10.1529/biophysj.106.086769
  • 39
    Zhang X, Dai J, Li L, Chen H, Chai Y. NLRP3 Inflammasome expression and signaling in human diabetic wounds and in high glucose induced macrophages. J Diabetes Res 2017; 2017: 5281358, doi: 10.1155/2017/5281358.
    » https://doi.org/10.1155/2017/5281358
  • 40
    Prattichizzo F, De Nigris V, Mancuso E, Spiga R, Giuliani A, Matacchione G, et al. Short-term sustained hyperglycaemia fosters an archetypal senescence-associated secretory phenotype in endothelial cells and macrophages. Redox Biol 2018; 15: 170-181, doi: 10.1016/j.redox.2017.12.001.
    » https://doi.org/10.1016/j.redox.2017.12.001
  • 41
    Wang Z, Shi C. Cellular senescence is a promising target for chronic wounds: a comprehensive review. Burns Trauma 2020; 8: tkaa021, doi: 10.1093/burnst/tkaa021.
    » https://doi.org/10.1093/burnst/tkaa021
  • 42
    Wilkinson HN, Clowes C, Banyard KL, Matteuci P, Mace KA, Hardman MJ. Elevated local senescence in diabetic wound healing is linked to pathological repair via CXCR2. J Invest Dermatol 2019; 139: 1171-1181.e6, doi: 10.1016/j.jid.2019.01.005.
    » https://doi.org/10.1016/j.jid.2019.01.005
  • 43
    Yang X, Chang Y, Wei W. Emerging role of targeting macrophages in rheumatoid arthritis: focus on polarization, metabolism and apoptosis. Cell Prolif 2020; 53: e12854, doi: 10.1111/cpr.12854.
    » https://doi.org/10.1111/cpr.12854
  • 44
    Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo Nat Protoc 2009; 4: 1798-1806, doi: 10.1038/nprot.2009.191.
    » https://doi.org/10.1038/nprot.2009.191
  • 45
    Alarcon MF, McLaren Z, Wright HL. Neutrophils in the pathogenesis of rheumatoid arthritis and systemic Lupus erythematosus: same foe different M.O. Front Immunol 2021; 12: 649693-649693, doi: 10.3389/fimmu.2021.649693.
    » https://doi.org/10.3389/fimmu.2021.649693
  • 46
    Saretzki G, Murphy MP, von Zglinicki T. MitoQ counteracts telomere shortening and elongates lifespan of fibroblasts under mild oxidative stress. Aging Cell 2003; 2: 141-143, doi: 10.1046/j.1474-9728.2003.00040.x.
    » https://doi.org/10.1046/j.1474-9728.2003.00040.x
  • 47
    Lagnado A, Leslie J, Ruchaud-Sparagano MH, Victorelli S, Hirsova P, Ogrodnik M, et al. Neutrophils induce paracrine telomere dysfunction and senescence in ROS-dependent manner. EMBO J 2021; 40: e106048, doi: 10.15252/embj.2020106048.
    » https://doi.org/10.15252/embj.2020106048
  • 48
    Perazzio SF, Salomão R, Silva NP, Andrade LE. Increased neutrophil oxidative burst metabolism in systemic Lupus erythematosus Lupus 2012; 21: 1543-1551, doi: 10.1177/0961203312461060.
    » https://doi.org/10.1177/0961203312461060
  • 49
    Elloumi N, Mansour RB, Marzouk S, Mseddi M, Fakhfakh R, Gargouri B, et al. Differential reactive oxygen species production of neutrophils and their oxidative damage in patients with active and inactive systemic lupus erythematosus. Immunol Lett 2017; 184: 1-6, doi: 10.1016/j.imlet.2017.01.018.
    » https://doi.org/10.1016/j.imlet.2017.01.018
  • 50
    Luo Q, Huang Z, Ye J, Deng Y, Fang L, Li X, et al. PD-L1-expressing neutrophils as a novel indicator to assess disease activity and severity of systemic Lupus erythematosus Arthritis Res Ther 2016; 18: 47-47, doi: 10.1186/s13075-016-0942-0.
    » https://doi.org/10.1186/s13075-016-0942-0
  • 51
    Herteman N, Vargas A, Lavoie JP. Characterization of circulating low-density neutrophils intrinsic properties in healthy and asthmatic horses. Sci Rep 2017; 7: 7743, doi: 10.1038/s41598-017-08089-5.
    » https://doi.org/10.1038/s41598-017-08089-5
  • 52
    Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, et al. A distinct subset of proinflammatory neutrophils isolated from patients with systemic Lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol 2010; 184: 3284-3297, doi: 10.4049/jimmunol.0902199.
    » https://doi.org/10.4049/jimmunol.0902199
  • 53
    van der Hoogen L, van der Linden M, Meyaard L, Ruth DEFS, van Roon JA, Radstake TR. Neutrophil extracellular traps and low-density granulocytes are associated with the interferon signature in systemic lupus erythematosus, but not in antiphospholipid syndrome. Ann Rheum Dis 2020; 79: e135, doi: 10.1136/annrheumdis-2019-215781.
    » https://doi.org/10.1136/annrheumdis-2019-215781
  • 54
    Hong CW. Current understanding in neutrophil differentiation and heterogeneity. Immune Netw 2017; 17: 298-306, doi: 10.4110/in.2017.17.5.298.
    » https://doi.org/10.4110/in.2017.17.5.298
  • 55
    Pinal-Fernandez I, Casal-Dominguez M, Mammen AL. Immune-mediated necrotizing myopathy. Curr Rheumatol Rep 2018; 20: 21, doi: 10.1007/s11926-018-0732-6.
    » https://doi.org/10.1007/s11926-018-0732-6
  • 56
    Knauss S, Preusse C, Allenbach Y, Leonard-Louis S, Touat M, Fischer N, et al. PD1 pathway in immune-mediated myopathies. Neurol Neuroimmunol Neuroinflamm 2019; 6: e558, doi: 10.1212/NXI.0000000000000558.
    » https://doi.org/10.1212/NXI.0000000000000558
  • 57
    Zhuang S, Russell A, Guo Y, Xu Y, Xiao W. IFN-γ blockade after genetic inhibition of PD-1 aggravates skeletal muscle damage and impairs skeletal muscle regeneration. Cell Mol Biol Lett 2023; 28: 27, doi: 10.1186/s11658-023-00439-8.
    » https://doi.org/10.1186/s11658-023-00439-8
  • 58
    Dalakas MC. Neurological complications of immune checkpoint inhibitors: what happens when you ‘take the brakes off' the immune system. Ther Adv Neurol Disord 2018; 11: 1756286418799864, doi: 10.1177/1756286418799864.
    » https://doi.org/10.1177/1756286418799864
  • 59
    Filippi M, Bar-Or A, Piehl F, Preziosa P, Solari A, Vukusic S, et al. Multiple sclerosis. Nat Rev Dis Primers 2018; 4: 43, doi: 10.1038/s41572-018-0041-4.
    » https://doi.org/10.1038/s41572-018-0041-4
  • 60
    Vaughn CB, Jakimovski D, Kavak KS, Ramanathan M, Benedict RHB, et al. Epidemiology and treatment of multiple sclerosis in elderly populations. Nat Rev Neurol 2019; 15: 329-342, doi: 10.1038/s41582-019-0183-3.
    » https://doi.org/10.1038/s41582-019-0183-3
  • 61
    Perdaens O, van Pesch V. Molecular mechanisms of immunosenescene and inflammaging: relevance to the immunopathogenesis and treatment of multiple sclerosis. Front Neurol 2021; 12: 811518, doi: 10.3389/fneur.2021.811518.
    » https://doi.org/10.3389/fneur.2021.811518
  • 62
    Sim FJ, Zhao C, Penderis J, Franklin RJ. The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J Neurosci 2002; 22: 2451-2459, doi: 10.1523/JNEUROSCI.22-07-02451.2002.
    » https://doi.org/10.1523/JNEUROSCI.22-07-02451.2002
  • 63
    Koutsoudaki PN, Papadopoulos D, Passias PG, Koutsoudaki P, Gorgoulis VG. Cellular senescence and failure of myelin repair in multiple sclerosis. Mech Ageing Dev 2020; 192: 111366, doi: 10.1016/j.mad.2020.111366.
    » https://doi.org/10.1016/j.mad.2020.111366
  • 64
    Calder PC, Ahluwalia N, Brouns F, Buetler T, Clement K, Cunningham K, et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br J Nutr 2011; 106: 3, S5-S78, doi: 10.1017/S0007114511005460.
    » https://doi.org/10.1017/S0007114511005460
  • 65
    Calder PC, Bosco N, Bourdet-Sicard R, Capuron L, Delzenne N, Doré J, et al. Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res Rev 2017; 40: 95-119, doi: 10.1016/j.arr.2017.09.001.
    » https://doi.org/10.1016/j.arr.2017.09.001
  • 66
    Morey M, O'Gaora P, Pandit A, Hélary C. Hyperglycemia acts in synergy with hypoxia to maintain the pro-inflammatory phenotype of macrophages. PloS One 2019; 14: e0220577, doi: 10.1371/journal.pone.0220577.
    » https://doi.org/10.1371/journal.pone.0220577
  • 67
    Brugnara L, Murillo S, Novials A, Rojo-Martínez G, Soriguer F, Goday A, et al. Low physical activity and its association with diabetes and other cardiovascular risk factors: a nationwide, population-based study. PloS One 2016; 11: e0160959, doi: 10.1371/journal.pone.0160959.
    » https://doi.org/10.1371/journal.pone.0160959
  • 68
    Aguilar EG, Murphy WJ. Obesity induced T cell dysfunction and implications for cancer immunotherapy. Curr Opin Immunol 2018; 51: 181-186, doi: 10.1016/j.coi.2018.03.012.
    » https://doi.org/10.1016/j.coi.2018.03.012
  • 69
    Messaoudi I, Warner J, Fischer M, Park B, Hill B, Mattison J, et al. Delay of T cell senescence by caloric restriction in aged long-lived nonhuman primates. Proc Natl Acad Sci USA 2006; 103: 19448-19453, doi: 10.1073/pnas.0606661103.
    » https://doi.org/10.1073/pnas.0606661103
  • 70
    Elgazar-Carmon V, Rudich A, Hadad N, Levy R. Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J Lipid Res 2008; 49: 1894-1903, doi: 10.1194/jlr.M800132-JLR200.
    » https://doi.org/10.1194/jlr.M800132-JLR200
  • 71
    Svahn SL, Gutiérrez S, Ulleryd MA, Nookaew I, Osla V, Beckman F, et al. Dietary polyunsaturated fatty acids promote neutrophil accumulation in the spleen by altering chemotaxis and delaying cell death. Infection Immun 2019; 87: e00270-e00319, doi: 10.1128/IAI.00270-19.
    » https://doi.org/10.1128/IAI.00270-19
  • 72
    Kado T, Nawaz A, Takikawa A, Usui I, Tobe K. Linkage of CD8(+) T cell exhaustion with high-fat diet-induced tumourigenesis. Sci Rep 2019; 9: 12284-12284, doi: 10.1038/s41598-019-48678-0.
    » https://doi.org/10.1038/s41598-019-48678-0
  • 73
    Ogrodnik M, Zhu Y, Langhi LGP, Tchkonia T, Krüger P, Fielder E, et al. Obesity-induced cellular senescence drives anxiety and impairs neurogenesis. Cell Metab 2019; 29: 1061-1077.e8, doi: 10.1016/j.cmet.2018.12.008.
    » https://doi.org/10.1016/j.cmet.2018.12.008
  • 74
    Khan S, Chan YT, Revelo XS, Winer DA. The immune landscape of visceral adipose tissue during obesity and aging. Front Endocrinol (Lausanne) 2020; 11: 267, doi: 10.3389/fendo.2020.00267.
    » https://doi.org/10.3389/fendo.2020.00267
  • 75
    Winer DA, Winer S, Shen L, Wadia PP, Yantha J, Paltser G, et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat Med 2011; 17: 610-617, doi: 10.1038/nm.2353.
    » https://doi.org/10.1038/nm.2353
  • 76
    Ovadya Y, Landsberger T, Leins H, Vadai E, Gal H, Biran A, et al. Impaired immune surveillance accelerates accumulation of senescent cells and aging. Nat Commun 2018; 9: 5435, doi: 10.1038/s41467-018-07825-3.
    » https://doi.org/10.1038/s41467-018-07825-3
  • 77
    Tarantini S, Valcarcel-Ares MN, Yabluchanskiy A, Tucsek Z, Hertelendy P, Kiss T, et al. Nrf2 Deficiency exacerbates obesity-induced oxidative stress, neurovascular dysfunction, blood-brain barrier disruption, neuroinflammation, amyloidogenic gene expression, and cognitive decline in mice, mimicking the aging phenotype. J Gerontol A Biol Sci Med Sci 2018; 73: 853-863, doi: 10.1093/gerona/glx177.
    » https://doi.org/10.1093/gerona/glx177
  • 78
    Zhang X, Xu GB, Zhou D, Pan YX. High-fat diet modifies expression of hepatic cellular senescence gene p16(INK4a) through chromatin modifications in adult male rats. Genes Nutr 2018; 13: 6, doi: 10.1186/s12263-018-0595-5.
    » https://doi.org/10.1186/s12263-018-0595-5
  • 79
    Kaastrup K, Grønbæk K. The impact of sedentary lifestyle, high-fat diet, tobacco smoke, and alcohol intake on the hematopoietic stem cell niches. HemaSphere 2021; 5: e615, doi: 10.1097/HS9.0000000000000615.
    » https://doi.org/10.1097/HS9.0000000000000615
  • 80
    Englund DA, Sakamoto AE, Fritsche CM, Heeren AA, Zhang X, Kotajarvi BR, et al. Exercise reduces circulating biomarkers of cellular senescence in humans. Aging Cell 2021; 20: e13415, doi: 10.1111/acel.13415.
    » https://doi.org/10.1111/acel.13415
  • 81
    Bartlett DB, Willis LH, Slentz CA, Hoselton A, Kelly L, Huebner JL, et al. Ten weeks of high-intensity interval walk training is associated with reduced disease activity and improved innate immune function in older adults with rheumatoid arthritis: a pilot study. Arthritis Res Ther 2018; 20: 127, doi: 10.1186/s13075-018-1624-x.
    » https://doi.org/10.1186/s13075-018-1624-x
  • 82
    Bartlett DB, Fox O, McNulty CL, Greenwood HL, Murphy L, Sapey E, et al. Habitual physical activity is associated with the maintenance of neutrophil migratory dynamics in healthy older adults. Brain Behav Immun 2016; 56: 12-20, doi: 10.1016/j.bbi.2016.02.024.
    » https://doi.org/10.1016/j.bbi.2016.02.024
  • 83
    Keller C, Keller P, Giralt M, Hidalgo J, Pedersen BK. Exercise normalises overexpression of TNF-alpha in knockout mice. Biochem Biophys Res Commun 2004; 321: 179-182, doi: 10.1016/j.bbrc.2004.06.129.
    » https://doi.org/10.1016/j.bbrc.2004.06.129
  • 84
    Nakajima K, Takeoka M, Mori M, Hashimoto S, Sakurai A, Nose H, et al. Exercise effects on methylation of ASC gene. Int J Sports Med 2010; 31: 671-675, doi: 10.1055/s-0029-1246140.
    » https://doi.org/10.1055/s-0029-1246140
  • 85
    Werner C, Fürster T, Widmann T, Pöss J, Roggia C, Hanhoun M, et al. Physical exercise prevents cellular senescence in circulating leukocytes and in the vessel wall. Circulation 2009; 120: 2438-2447, doi: 10.1161/CIRCULATIONAHA.109.861005.
    » https://doi.org/10.1161/CIRCULATIONAHA.109.861005
  • 86
    Song S, Lam EW, Tchkonia T, Kirkland JL, Sun Y. Senescent cells: emerging targets for human aging and age-related diseases. Trends Biochem Sci 2020; 45: 578-592, doi: 10.1016/j.tibs.2020.03.008.
    » https://doi.org/10.1016/j.tibs.2020.03.008
  • 87
    Boyajian JL, Ghebretatios M, Schaly S, Islam P, Prakash S. Microbiome and Human aging: probiotic and prebiotic potentials in longevity, skin health and cellular senescence. Nutrients 2021; 13: 4550, doi: 10.3390/nu13124550.
    » https://doi.org/10.3390/nu13124550
  • 88
    Matt SM, Allen JM, Lawson MA, Mailing LJ, Woods JA, Johnson RW. Butyrate and dietary soluble fiber improve neuroinflammation associated with aging in mice. Front Immunol 2018; 9: 1832, doi: 10.3389/fimmu.2018.01832.
    » https://doi.org/10.3389/fimmu.2018.01832
  • 89
    Fu J, Yu A, Xiao X, Tang J, Zu X, Chen W, et al. CD4(+) T cell exhaustion leads to adoptive transfer therapy failure which can be prevented by immune checkpoint blockade. Am J Cancer Res 2020; 10: 4234-4250.
  • 90
    Yang X, Jiang X, Chen G, Xiao Y, Geng S, Kang C, et al. T cell Ig mucin-3 promotes homeostasis of sepsis by negatively regulating the TLR response. J Immunol 2013; 190: 2068-2079, doi: 10.4049/jimmunol.1202661.
    » https://doi.org/10.4049/jimmunol.1202661

Publication Dates

  • Publication in this collection
    17 June 2024
  • Date of issue
    2024

History

  • Received
    30 Nov 2023
  • Accepted
    22 Apr 2024
Associação Brasileira de Divulgação Científica Av. Bandeirantes, 3900, 14049-900 Ribeirão Preto SP Brazil, Tel. / Fax: +55 16 3315-9120 - Ribeirão Preto - SP - Brazil
E-mail: bjournal@terra.com.br