Skip to main content

The diseased kidney: aging and senescent immunology

Abstract

Immunosenescence is the deterioration of the innate and adaptive immune systems associated with aging and is primarily characterized by a reduction in T cell production and accumulation of atypical subsets. Age-related immunological dysfunction leads to impaired immune protection and persistent low-grade chronic inflammation, resulting in a decreased vaccination response and increased vulnerability to infection, cancer, cardiovascular disease, and autoimmune disease in the elderly. As the elderly constitute a growing proportion of the population with renal disease, immunosenescence is a normal aging process that is prevalent among older people. In addition, immunosenescence seems to be more pronounced in patients with kidney diseases than in healthy controls, as shown by severe chronic inflammation, accumulation of immune cells with the senescent phenotype (CD28 T cells, CD14+CD16+ monocytes), and proinflammatory cytokine production. Immunosenescence inhibits immunological clearance and renal tissue regeneration, thereby increasing the risk of permanent renal damage, infection, and cardiovascular events in patients with kidney disease, lowering the prognosis, and even influencing the efficacy of renal replacement treatment. Biological drugs (senomorphics and senolytics) target the aging immune system and exert renoprotective effects. This review aims to emphasize the features of immunosenescence and its influence on kidney diseases and immunotherapy, highlighting the future directions of kidney disease treatment using senescence-focused techniques.

Introduction

Advances in medical technology have significantly enhanced life expectancy. The worldwide population aged 60 years or older is expected to quadruple by 2050, representing 22% of the global population [1]. In response to the worldwide issues of age-related diseases caused by an aging population, the study of aging has gained growing interest. Intrinsic and extrinsic factors shape the immune system and profoundly change with aging. In other words, the immune response is age dependent [2]. In the 1960s, the gerontologist Roy Walford first introduced the term “immunosenescence”; his landmark book, “The Immunologic Theory of Aging,” laid the foundation for many advanced studies about immunity and aging [3]. The change of cellular phenotype and the immune system composition, termed “immunosenescence” presents two opposing characteristics: increased susceptibility to infectious disease and persistent systemic inflammation. Consequently, the risk of many degenerative diseases [4], especially neurodegeneration [5], cancer [6], cardiovascular [7], and autoimmune diseases [8], is higher in people with immunosenescence. Therefore, preserving immune function may reduce disease incidence and prolong life. Numerous studies on the underlying mechanisms of age-related immune decline have laid the groundwork for approaches to cancer and chronic diseases. Immune checkpoint blockade therapies, such as anti-programmed death 1 (PD-1) and anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), have provided new hope for the treatment of hematological malignancies [9]. Cryopreservation of immune cells and immune cell modification techniques play positive roles in treating senile tumors [10]. T cells are genetically modified to express the chimeric antigen receptor for a specific cell surface antigen to identify and kill tumor cells. The emerging use of senolytics and senomorphics has been investigated to delay aging and treat chronic diseases in humans [11].

Kidney disease has been recognized as a major public health burden over the past decade. The prevalence of chronic kidney disease (CKD) exceeds 10% and is > 50% in high-risk subpopulations [12]. Aging kidneys undergo structural and functional alterations due to the complex interplay between hereditary and environmental changes and cellular dysfunction. Renal structural abnormalities include reduced nephron size and number, tubular interstitial fibrosis, renal capsule fibrosis, glomerulosclerosis, thickening of the glomerular basement membrane, increased glomerulosclerosis, and tubular atrophy. Additionally, renal functional changes manifest as glomerular filtration rate (GFR) decline, accumulation of toxins, and urine and blood component changes [13]. Progressive loss of kidney function ultimately leads to end-stage renal disease (ESRD), characterized by a series of systemic disorders including electrolyte disorders, metabolic dysregulation, and acidosis, also known as uremia, which affects almost all organs in the body.

Cellular senescence is a long-lasting cell cycle arrest induced by replicative senescence (chronological aging) and premature senescence (organ injury) [14], which is suggested to be a hallmark of aging and contributes to chronic diseases. Senescent cells secrete a specific senescence-associated secretory phenotype (SASP), which mainly includes a variety of proinflammatory cytokines (e.g., interleukin (IL)-1α, IL-1β, IL-6, and IL-8), growth factors (e.g., hepatocyte growth factor, transforming growth factor-β, and granulocyte-macrophage colony-stimulating factor), chemokines [e.g., chemokine (C-X-C motif) ligand (CXCL)-1/3 and CXCL-10], and matrix remodeling enzymes (e.g., metalloproteinases) [15]. Renal senescence occurs when senescent cells accumulate in the kidneys and chronic low-grade inflammation is induced. Chronic inflammation impedes intrinsic tissue regeneration and exacerbates renal damage, thus increasing the vulnerability of older adults to kidney disease and reducing their renal regenerative potential.

Age-related changes in the immune system associated with aging play an essential role in the pathogenesis of kidney diseases. Consequently, it is essential to understand the interactions between immunosenescence and renal disorders. Numerous investigations of the underlying processes of age-related alterations in the immune system have paved the way for novel treatment approaches for kidney disorders. This review discusses the recent molecular research on immunosenescence, focusing on innate and adaptive immune cell changes. We will also examine immunosenescence in kidney diseases, evaluate current immunotherapies that target age-related immune system abnormalities, and assess their potential applications in kidney diseases.

Immunosenescence of innate immune response cells

Natural killer (NK) cells

NK cells are key cells in the innate immune system for recognizing and clearing viral infections and abnormal cells (including tumor cells). Recent research has shown that preserving NK cell features in older adults may contribute to longevity and successful aging. Based on the expression levels of the surface molecules CD56 and CD16, NK cells can be divided into two subgroups. CD56bright cells are an immature subset that shows high proliferative activity and the ability to produce a range of cytokines, such as interferon (IFN)-γ, tumor necrosis factor (TNF)-β, and IL-10, and chemokines, such as RANTES and macrophage inflammatory protein-1α. In contrast, CD56dim cells are a mature subset that show high cytotoxic activity and a lower ability to produce cytokines [16]. The peripheral CD56bright NK cells in the elderly is decreased, which may be caused by the degeneration of hematopoietic stem cells (HSCs) in bone marrow (where the NK cells are derived) of aging individuals [17].

Meanwhile, the number of CD56dim NK cell subsets is more in man than that of women [18]. Although the overall cytotoxicity of NK cells did not decrease significantly in healthy older adults, the activity of each NK cell was reduced compared to that in young adults. Extensive phenotypic and functional analysis of NK cells from healthy subjects revealed that NK cells in cord blood displayed specific features, including poor expression of killer immunoglobulin-like receptors and leukocyte immunoglobulin-like receptor-1/immunoglobulin-like transcript receptor-2 (LIR-1/ILT-2), and high expression of both NKG2A and IFN-γ. In contrast, NK cells from older subjects showed a specific increase in LIR-1/ILT-2 levels. The ability to produce IFN-γ was modestly impaired in NK cells from older participants. Importantly, they also observed a perfect recovery of NK cell function in very old subjects following IL2-activation [19]. The expression of natural cytotoxic receptors, such as NKp30 and NKp46, on the surface of NK cells is decreased in the elderly, while both receptors are expressed at high levels in the young [20].

Dendritic cells

Dendritic cells (DCs) are the most potent antigen-presenting cells (APC) with TLRs on their surfaces [21]. Upon antigenic stimulation, DCs capture, process, and present pathogen antigens through major histocompatibility complex II (MHC II) and release cytokines, thus priming naïve T cells and initiating adaptive immunity. DCs have been long focused in immunotherapy as a bridge between innate and adaptive immunities.

The myeloid (mDC), plasmacytoid (pDC), and follicular (fDC) dendritic cell subsets are distinct subsets with particular roles. Several investigations have demonstrated age-related impairments in DCs antigen presentation and T-cell activation in the elderly. The production of type I and III IFNs, which enhance the coordination of innate and acquired immune responses, is reduced in elderly pDCs [22]. The activity of PI3 kinase is markedly diminished in the mDCs of aged donors, resulting in altered phagocytic activity and migration in response to inflammatory stimuli. Moreover, mDCs demonstrate abnormal inflammatory cytokine production (IL-6 and TNF-α) due to the overexpression of nuclear factor-kappa B (NF-κB) in elderly adults [23]. The immune response to autoantigens is also enhanced, leading to decreased immune tolerance and chronic inflammation in the elderly population.

Antigen presentation to naïve B cells and the formation of germinal centers by fDCs with highly expressed FcγRII receptors play a critical role in stimulating robust humoral immunity in response to infection or vaccination [24]. Defects in germinal center formation with impaired immunological memory have been observed in aged mice. Age-related decline in FcγRII receptor expression is implicated in diminished antigen retention and presentation ability, leading to a decline in B cell activation and antibody production [25].

Monocytes and macrophages

Monocytes express a broad range of pattern recognition receptors and play a crucial role in the innate immune response. Monocytes are activated upon pathogen stimulation via TLRs, and subsequently secrete proinflammatory cytokines and present antigens, exerting various immune effector functions. Human monocytes are identified by the expression of CD14 and CD16 on their cell surface. Classical, intermediate, and non-classical monocytes are defined as CD14+CD16, CD14+CD16+, and CD14CD16+, respectively [26]. CD16+ monocytes present a more senescent phenotype with shorter telomeres and an increased inflammatory potential [27]. No significant differences were observed in the number of circulating monocytes in older adults, whereas the proportion of CD16+ monocytes was higher than that in young adults. Macrophages are tissue-resident cells that are known to phagocytose and initiate an inflammatory signaling cascade. Although inflammation is important for insult defense, it is detrimental to the immune system. Monocytes and macrophages are believed to be central components facilitating low-grade chronic inflammation during immunosenescence [28]. Macrophages are recognized as two different phenotypes, classical (M1) and alternative (M2), based on their different inflammatory responses and cytokine release, including TNFα, IL-1β, and IL-12 [29]. M2 monocytes are labeled as anti-inflammatory monocytes and show an obvious age-related reduction in healthy elderly individuals [30]. A larger monocyte population exists in the elderly with impaired TLR expression and biological responses [31]. Thus, when stimulated with TLR stimuli, they reduce proinflammatory cytokine production (such as IL-1β and IFN-γ) compared to young cells [32]. This may help to elucidate innate immune defects in older people. Aged macrophages produce more cyclooxygenase 2 and subsequently prostaglandin 2, which is correlated with increased expression of inflammatory cytokines, such as TNFα and IL-6.

Immunosenescence of adaptive immune response cells

T and B cells are essential for adaptive immunological responses to antigens. Lifetime pathogen exposure is associated with an age-related decline in these immune cells, which is characterized by a reduced output of naïve T/B cells, abnormal phenotype of mature cells, and accumulation of memory cells. These changes in adaptive immune cells result in insufficient immune response to new antigens in older individuals (Fig. 1).

Fig. 1
figure 1

Characteristics of immunosenescence in innate and adaptive immunity. Many immune cell subpopulations are altered during immunosenescence. The mature CD56dim natural killer (NK) cell subsets with high cytotoxic activity are decreased, diminishing the ability to recognize and clear viral infection and tumor cells. Antigen processing and presentation capabilities of macrophages and APCs are also diminished. Declined naïve T cells output of the thymus and decreased antigen stimulation presented by APCs lead to declining T cell response. Fewer naïve B cells are stimulated and transform into plasma cells, thus humoral immunity is impaired. However, the number of memory T and B cells increases

T cells

The thymus provides a variety of microenvironments that support the development and export of naïve T-cells to the periphery [33]. Since puberty, the thymus gland undergoes gradual involution as sex hormones increase, decreasing the T-cell repertoire to new antigens, which plays a crucial role in T cell senescence. In addition, reducing IL-7 levels and oxidative stress damage contribute to the onset of thymic atrophy [34,35,36,37]. In naïve CD4+ T cells, decreased expression of miR-181A promotes the activity of dual-specific phosphatase 6 (DUSP6), thereby contributing to the desensitization of T cell receptors (TCRs) and contraction of TCR diversity [38]. T cells are repeatedly triggered by the same antigens throughout the lifelong chronic antigen load, creating a population of late-differentiated oligoclonal effector memory T cells and reducing the naive T cell reservoir [39]. The adaptive immune response to new antigens is decreased in aged adults due to the shrinkage of naive T cell reservoirs and expansion of immunological memory for previously encountered infections. Moreover, senescent T lymphocytes have subsets with abnormal phenotypes and abnormal expression of cell-surface molecules. After a primary infection, human cytomegalovirus (HCMV) resides in the body and periodically activates the immune system. The accumulation of terminally differentiated HCMV-specific CD8+ T cells and inversion of the CD4+/CD8+ T cell ratio are associated with HCMV seropositivity in the elderly (> 80 years) [40, 41]. Senescent T cells maintain low expression levels of the necessary co-stimulatory molecule CD27/CD28 but upregulate inhibitory receptors on the cell surface, including PD-1 and CTLA-4. CD28 T cells are resistant to apoptosis and less susceptible to modulation by regulatory T cells. Functionally, CD28 T cells exhibit a high cytotoxic trait and express cytotoxic effector molecules (e.g., granase B and perforin) and NK cell receptors, culpable CD28 T cells in the pathogenesis of various age-related diseases [42, 43]. Increased senescent T cells express negative signaling receptors, such as PD-1, which suppresses the T cell response. Interestingly, PD-1 blockade in the elderly partially restores the functional competence of T-cells [44]. Senescent T cells also express CD57 and killer cell lectin-like receptor subfamily G member 1, with high cytotoxic potential [45].

B cells

Age also affects the quantity and variety of B cells as the number of naïve B cells decreases, while that of effector B cells increases. In the peripheral blood, immunoglobulins generated by naïve B cells (IgD, IgM) are replaced by those produced by memory B cells (IgG and IgA) [46]. Additionally, the immunological response to foreign antigens and the ability to reorganize immunoglobulin genes are diminished. Reduced E2A gene expression in activated B cells of the elderly diminishes the induction of E47 (a class I basic helix-loop-helix protein produced by the E2A gene), thereby diminishing antibody avidity and antibody-mediated protection [47, 48]. The memory/effector T cells show reduced expression of CD40L and reduced B cell synthesis [49].

Inflammaging

The state of prolonged, sterile, low-grade inflammation, namely inflammaging, is almost universally present in the elderly [50]. While acute inflammation is regarded as a protective process against harmful pathogens, persistent unresolved inflammation could be disadvantageous. Used to be considered as a part of “immunosenescence,” inflammaging should be regarded as a state different from but closely interacting with immunosenescence. Inflammaging contributes to changes in cellular phenotype and immune system composition, reducing endurance to different antigenic stressors in aging individuals. Both inflammaging and immunosenescence are common mechanisms of age-related decline and diseases, including type 2 diabetes and cardiovascular diseases [51]. In addition, persistent low-grade infection contributes to lineage skewing of HSCs toward myeloid progenitors, and thus the potential of HSCs to replenish lymphoid lineage cells is compromised [52]. Lifelong body adaptation to antigenic load and stress exposure can result in the remodeling of both innate and adaptive immunity, which is globally regarded as immunosenescence.

This state of chronic inflammation is associated with the continuous activation of macrophages via pattern recognition receptors [Toll-like receptors (TLRs). nucleotide-binding oligomerization domain (NOD)-like receptors] interact with damage-associated molecular patterns (DAMPs)/pathogen-associated molecular pattern (PAMPs)/metabolism-associated molecular patterns (MAMPs) that initiate the proinflammatory cascade [53]. The specific changes in senescent microbiota and gut barrier injury in the elderly are great sources of disease-associated pathobionts, which are associated with an increased local and systemic inflammatory status, sustaining inflammaging and dialogue with other organs, particularly the kidneys [54]. Recent reports have shown overexpression of TLRs in multiple kidney diseases, including autoimmune renal diseases, vasculitis, acute kidney injury (AKI), and kidney transplant rejection. TLRs signals activate the myeloid differentiation primary response protein 88 (MyD88)/TIR-domain-containing adapter-inducing interferon-β, and then activate NF-κB target genes, a transcription factor that regulates inflammatory and oxidative stress pathways [55, 56]. Excessive activation of NF-κB leads to an asymptomatic inflammatory tone (IL-1, IL-6, IL-15, IL-18, TNF, and C-reactive protein), renal fibrosis, and CKD progression [57,58,59,60]. Danger signals activate NOD-like receptors, which can form multi-protein complexes or inflammasomes, promote the cleavage of pro-caspase 1 into active caspase-1, and subsequently activate proinflammatory cytokines, including IL-18 and IL-1β [61]. In experimental models, inflammasomes and inflammasome-related genes, particularly NOD-, LRR-, and pyrin domain-containing 3, contribute to the pathogenesis of a wide range of chronic kidney disease, acute kidney injury, and diabetic kidney disease via canonical and non-canonical mechanisms that regulate inflammation, pyroptosis, apoptosis, and fibrosis [62]. During the inflammatory process, cells of innate immunity, such as phagocytic cells, produce high amounts of reactive oxygen species (ROS) to eliminate foreign agents. High ROS levels may contribute to the oxidative damage associated with many diseases and aging. Moreover, oxidant components can act as intracellular secondary messengers during the inflammatory response [63]. Overproduction of oxidative stress can induce an inflammatory response, thus establishing a vicious cycle that fuels immunosenescence. As a result of age-related oxidative injury, senescent cells lose some of their capacity to regulate their own redox and inflammatory balance, which is the basis of immunosenescence, namely “oxi-inflamm-aging’ [64].

Immunosenescence and kidney diseases

Immunosenescence in acute kidney injury

AKI is a life-threatening clinical condition with a high incidence and mortality rate (16–50%) [65]. AKI is associated with an abrupt decrease in kidney function caused by pathogenic conditions such as ischemia and toxic stimuli. AKI is considered a self-limiting syndrome, with most survivors regaining renal function after damage. However, 5% of AKI survivors live with persistent renal impairment and require continuous renal replacement therapy. In older patients, this proportion may be as high as 16%. Patients with severe AKI can develop interstitial fibrosis, CKD, and ESRD later in life [66, 67]. Previous research has reported that renal injury could induce premature aging in the kidneys, suggesting that these changes increase the propensity to develop subsequent progressive CKD.

In the early phase of renal damage, tubular epithelial cells (TECs) senescence is extremely prevalent. A few days after kidney injury, TECs begin to senesce, which is regulated by Toll-like and interleukin 1 receptors (TLR/IL-1R) of the innate immune system [68]. Senescent TECs may cause senescence in surrounding cells in a paracrine manner; hence, the number of senescent cells increases over time [69]. The accumulation of senescent cells can directly deteriorate the microenvironment and contribute to tissue damage, premature renal aging, and increased susceptibility to chronic renal disease. Increased complement C5a binding to C5R on TECs in patients with coronavirus disease 2019 is responsible for TECs senescence and subsequent progression to CKD [70] (Fig. 2).

Fig. 2
figure 2

Senescent TECs accumulation after AKI drives the progression of CKD. Acute kidney injury induces cellular damage and DNA degradation in TECs. Maladaptive repair after AKI leads to TECs senescence. Senescent TECs accumulates in kidney with SASP, which mainly includes proinflammatory cytokines, growth factors, chemokines, and matrix remodeling enzymes. These proinflammatory and profibrotic molecules aggravate immune cells infiltration and tubular cell injury, leading to persistent tubulointerstitial inflammation, proliferation of fibroblasts, and excessive deposition of extracellular matrix, which leads to the exacerbation of renal injury and drives AKI to CKD. Senescent immune cells such as CD28 T cells, CD14+CD16+ monocytes also aggravate chronic inflammation and ROS production in kidney, which promotes the progression of CKD

Tertiary lymphoid tissue develops in the kidneys of various mouse models following the kidney damage. The development of tertiary lymphoid tissue was a consequence of immunosenescence. It is also detectable in young patients with autoimmune disorders such as IgA nephropathy and anti-neutrophil cytoplasmic antibody vasculitis [71]. Tertiary lymphoid tissue spatially occupies a broad area of the renal parenchyma, induces intrarenal inflammation, and impedes intrinsic tissue repair. A substantial number of proinflammatory cytokines are generated in these tertiary lymphoid tissues, which may underlie chronic inflammation (inflammaging) in the kidney.

Immunosenescence in chronic kidney disease and end-stage renal disease

CKD describes structural and functional damage to the kidneys that persists over time and is accompanied by a persistent decrease in GFR. The CKD phases range from asymptomatic stage 1 to ESRD stage 5. From 1990 to 2017, the global prevalence of CKD increased by 29.3% (range: 26.4–32.6%), according to the Global Burden of Disease [72]. Statistically, more patients with CKD eventually develop ESRD and require renal replacement therapy, such as dialysis (43.1%) and kidney transplantation (34.4%) [73]. Treatment for advanced CKD is restricted and the prognosis is poor. Thus, early diagnosis and innovative therapeutics targeting these potential mechanisms are required. Morphological and functional changes in aging kidneys have been covered previously, including decreasing nephron loss, renal fibrosis, GFR reduction, tubule function decrease, and microvascular alteration. Senescent cell markers, senescence-associated galactosidase, p16, and Ki-67, are expressed at higher levels in the kidneys of aging mice and humans [74]. Owing to chronological aging, the older population is more susceptible to CKD. Further, patients with CKD and ESRD may also have premature immunosenescence, making them physiologically older than the general population. An analysis of T cells from patients with ESRD revealed premature immunological aging; the immunological age of patients with ESRD was 20–30 years older than that of healthy controls of the same chronological age [75].

Patients with CKD exhibit cellular alterations characterized by chronic inflammation, advanced cellular senescence, and immune system dysfunction. The long-term retention of uremic molecules and cytokines in patients with CKD leads to chronic inflammation. Premature aging of the immune system may be induced and accelerated in the persistent uremic milieu of patients with CKD [75, 76]. Additionally, oxidative stress, acidosis, infection, dysbiosis, and metabolic dysregulation may contribute to immunosenescence in patients with CKD and ESRD. Subsequently, exacerbated immunosenescence promotes disease development and increases the susceptibility to infection. Studies in pediatric CKD have shown that pediatric T-cell phenotypes were similar to those of the aging population in the chronic inflammation environment of CKD, including T cell exhaustion and senescence, naïve T cell reduction, and CD28 expression loss [77]. The CD14+CD16+ monocytes in patients with CKD were statistically higher than those in healthy controls, expressing higher levels of proinflammatory cytokines and vascular adhesion molecules than CD14+CD16 monocytes. The high frequency of these cells facilitates the state of chronic inflammation, increases the risk of CKD progression to ESRD, and is associated with worse cardiovascular outcomes. Ironically, these changes seem to persist as even after successful kidney transplantation, the ratio of CD28 T cells to CD14+CD16+ monocytes did not normalize inspite of reduction in the serum levels of proinflammatory cytokines [78, 79].

Immunosenescence in kidney replacement therapy

Over the past three decades, the number of patients over 75 years of age who developed ESRD has tripled from 7.6% to over 20% in the United States. As a result of population aging and the increased prevalence of age-related diseases, such as hypertension and diabetes mellitus, the elderly is the fastest-growing segment of patients diagnosed with ESRD, and millions of them die due to a lack of kidney replacement therapy (KRT). Dialysis, including peritoneal dialysis and hemodialysis, is a well-accepted KRT for irreversible renal failure. Kidney transplantation is the best health-preserving and economical KRT for ESRD; the number of global renal transplantation recipients with ESRD has increased by 34.4% from 1990 to 2017 [73]. As the demand for kidney transplants increases significantly, the average age of those on transplant waiting lists is also increasing. Therefore, to alleviate organ shortage, donor eligibility should be expanded to include elderly donors, resulting in more kidney transplants from older persons.

Despite improving the mortality risk of patients on dialysis over the past 50 years, mortality remains unacceptably high due to cardiovascular events and infection [80, 81]. Research found that the hallmark of immunosenescence, such as premature aging of CD4+ T cells, particularly the depletion of naïve T cells, was considered a strong predictor of mortality among patients on hemodialysis [82]. In addition, a 2018 study showed that atherosclerotic cardiovascular events were correlated with the circulating CD4+CD28 T-cell population in patients on hemodialysis. Excess CD4+ T cells expressing this immunosenescence marker may significantly mediate atherosclerotic cardiovascular events in patients on dialysis [83]. In another study, the telomere attrition of mononuclear cells isolated from patients on hemodialysis was higher than that of age-matched controls, indicating that hemodialysis therapy might contribute to monocytic senescence [84]. In contrast, organs from elderly donors are more susceptible to ischemia/reperfusion injury, which enhances the release of DAMPs, resulting in a greater inflammatory response. Considering that older kidneys are more likely to be transplanted into older recipients, acute graft rejection would be more severe if it occurred; the intrinsic functional deficits and higher immunogenicity of the elderly kidney may be important factors.

Advanced age of organ transplant recipients or donors markedly affects renal transplantation outcomes. Retrospective studies have shown a higher acceptance rate of kidney transplantation in older recipients. In renal transplantation recipients older than 60 years, CD4+ T cell depletion is a common hallmark of immunosenescence and has been suggested to be linked to lower rates and weaker acute graft rejection [85, 86].

In addition, kidney transplantation may physiologically accelerate immunosenescence through inflammatory stimuli of alloantigen exposure. Innate Eomes+ CD8+ T cells are a unique subset that is closely associated with IFN-γ production during the innate response. In a pilot cohort of renal recipients, the number of innate Eomes+ CD8+ T cells was increased and exhibited an exacerbated senescent phenotype (CD27, CD28) [87]. Researchers have observed renal transplant recipients 1 year after transplantation. They found that their follow-up anti-thymocyte globulin (ATG) treatment had a toxic effect on thymic output and bone marrow-derived lymphoid progenitor cells. Terminally differentiated CD57+/CD28 T cells are a senescent phenotype correlated with a higher risk of post-transplantation infection and acute rejection, which was found to expand in ATG-treated patients. In renal transplantation, follow-up immunosuppressive treatment [such as ATG and mycophenolate mofetil (MMF)] was associated with accelerated immunological aging after normalization of the uremic milieu with KRT [88]. The suppressive activity of regulatory T cells (Tregs) can be maintained in patients with ESRD by diverse differentiation pathways of inducible costimulatory (ICOS)+ and ICOS recent thymic emigrant-Tregs/responder T-cells [89]. However, this equilibrium is abrupt during long-term renal transplantations.

Therapeutic strategy targeting the senescent immune system

Because of the pivotal role of senescent cells and the senescent immune system in age-related diseases, strategies to remove senescent cells or to block SASP have become imperative, and increasing interest has been aroused in rejuvenating the immune system. Senotherapeutics, including senomorphics and senolytics, can eliminate or delay adverse effects of cellular senescence and aging. Senomorphics are small molecules that dampen chronic inflammation in aging kidneys by blocking pathways related to SASP expression, such as p38 mitogen-activated protein kinase (MAPK) [90], NF-κB [91, 92], and silencing information regulator 2 related enzyme 1/SMAD3 [93]. Senomorphics may also inhibit senescence in neighboring cells by impeding the paracrine signaling of TECs. Senolytics are pharmacological agents that selectively kill senescent cells and impede their accumulation in tissues, consequently delaying senescence and improving age-related diseases. Senolytics diminish the population of senescent cells by interfering with senescent-cell anti-apoptotic pathways, including inhibition of the Bcl-2 family, phosphatidylinositol-3-kinase, thymidine kinases, and fork head box O4-p53 interaction [94]. The natural compound quercetin ameliorates kidney injury by inhibiting M1/M2 macrophage polarization, which is associated with downregulated activities of NF-κB p65 and IFN regulatory factor 5, thus leading to the inactivation of upstream signaling TLR4/Myd88 [95]. Dasatinib plus quercetin significantly decreased the senescent cell burden and renal inflammation in individuals with diabetic kidney disease [96]. The most effective strategy may be immune modulation as it activates the natural clearance of senescent cells, promotes renal tissue repair, and inhibits inflammation in senescent kidneys. T cell senescence plays a major role in age-related decline in immune function. p38 MAPK blockade can reverse senescence in CD8+ T cells by increasing their cellular proliferation, telomerase activity, and mitochondrial biogenesis via the mammalian target of rapamycin -independent pathway [97].

Conclusion

With the steady increase in the number of elderly individuals globally, age-related diseases emerge as a major challenge to health care workers. Apart from functional and structural changes in the kidneys introduced by aging, immune system decline also significantly increases the risk of age-related kidney diseases. Immunosenescence is a loose definition of age-related changes in the innate and adaptive immune responses, which is characterized by shrinkage of naïve immune cell reservoirs, accumulation of late-stage differentiated cells with a senescent phenotype, and immunoglobulin class switching. These changes in the immune system result in two seemingly incompatible aspects: diminished immune response and increased inflammatory response, also known as inflammaging.

TECs senescence and tertiary lymphoid tissue formation occur following AKI. Senescent kidney cells promote a chronic inflammatory microenvironment, which can subsequently cause local tissue damage, hinder tissue repair, and promote immune system senescence. Intrarenal inflammation underlies the development of renal fibrosis and CKD later in life. Immunosenescence is exaggerated in patients with CKD and ESRD. Hallmarks of immunosenescence, including decreased naïve T cells, reduced CD28 expression, and increased proinflammatory macrophages, are convincing predictors of mortality in patients with CKD and ESRD. Renal replacement therapy for old patients with ESRD results in a lower acute rejection rate after the kidney transplantation. However, immunosenescence may increase the risk of chronic, but severe, graft failure. In addition, immunosenescence has been reported to speed up during kidney transplantation and immunosuppressive treatment (e.g., ATG and MMF).

In 1962, Walford first proposed the concept of immunity and aging. Numerous researchers have attempted to determine the relationship between immune aging and diseases that are common in the elderly, including for kidney diseases. However, most available studies (whether performed in humans or animals such as mice) are cross-sectional; ambiguous associations between the kidney disease and the senescent immune phenotype have been demonstrated in these cross-sectional studies. The immune system is shaped and undergoes changes throughout life. Longitudinal studies are required to elucidate the causal relationship between immunosenescence and kidney disease.

Availability of data and materials

Not applicable.

Abbreviations

PD-1:

Programmed death 1

CTLA-4:

Cytotoxic T-lymphocyte-associated protein 4

CKD:

Chronic kidney disease

GFR:

Glomerular filtration rate

ESRD:

End-stage renal disease

SASP:

Senescence-associated secretory phenotype

IL:

Interleukin

CXCL:

Chemokine (C-X-C motif) ligand

NK:

Natural killer

IFN:

Interferon

TNF:

Tumor necrosis factor

HSCs:

Hematopoietic stem cells

LIR-1/ILT-2:

Leukocyte immunoglobulin-like receptor-1/immunoglobulin-like transcript receptor-2

DCs:

Dendritic cells

APC:

Antigen-presenting cell

TLRs:

Toll-like receptors

MHC:

Major histocompatibility complex

mDC:

Myeloid dendritic cell

pDC:

Plasmacytoid dendritic cell

fDC:

Follicular dendritic cell

DUSP6:

Dual specific phosphatase 6

TCR:

T cell receptor

HCMV:

Human cytomegalovirus

TLRs:

Toll-like and interleukin 1 receptors

NOD:

Nucleotide-binding oligomerization domain

DAMPs:

Damage-associated molecular patterns

PAMPs:

Pathogen-associated molecular pattern

MAMPs:

Metabolism-associated molecular patterns

AKI:

Acute kidney injury

MyD88:

Myeloid differentiation primary response protein 88

NF-κB:

Nuclear factor-kappa B

ROS:

Reactive oxygen species

TECs:

Tubular epithelial cells; KRT, kidney replacement therapy

ATG:

Antithymocyte globulin

MMF:

Mycophenolate mofetil

Tregs:

Regulatory T cells

ICOS:

Inducible co-stimulatory

MAPK:

Mitogen-activated protein kinase

ICB:

Immune checkpoint blockade

PD-1:

Programmed death 1

CTLA-4:

Cytotoxic T-lymphocyte-associated protein 4

CAR:

Chimeric Antigen Receptor

GFR:

Glomerular filtration rate

TECs:

Tubular epithelial cells

SASP:

Senescence-associated secretory phenotype

DUSP6:

Dual specific phosphatase 6

TCR:

T cell receptor

NK:

Natural killer

TNF:

Tumor necrosis factor

HSCs:

Hematopoietic stem cells

NCR:

Natural cytotoxic receptors

APC:

Antigen-presenting cell

MHC:

Major histocompatibility complex

DCs:

Dendritic cells

TLRs:

Toll-like receptors

DAMPs:

Damage-associated molecular patterns

PAMPs:

Pathogen-associated molecular pattern

MAMPs:

Metabolism-associated molecular patterns

TNF:

Tumor necrosis factor

CRP:

C-Reactive protein

AKI:

Acute kidney injury

CKD:

Chronic kidney disease

ESRD:

End-stage renal disease

TECs:

Tubular epithelial cells

TLR/IL-1R:

Toll-like and interleukin 1 receptors

COVID-19:

Coronavirus disease 2019

ANCA:

Anti-neutrophil cytoplasmic antibodies

KRT:

Kidney replacement therapy

IRI:

Ischemia/reperfusion injury

ATG:

Antithymocyte globulin

MMF:

Mycophenolate mofetil

Tregs:

Regulatory T cells

ICOS:

Inducible co-stimulatory

RTE:

Recent thymic emigrant

Tresps:

Responder T-cells

SCAPs:

Senescent-cell anti-apoptotic pathways.

References

  1. Organization WH. Ageing and health 2021 [Available from: http://www.who.int/news-room/fact-sheets/detail/ageing-and-health.

  2. Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, et al. An aged immune system drives senescence and ageing of solid organs. Nature. 2021;594(7861):100–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Effros RB. Roy Walford and the immunologic theory of aging. Immun Ageing. 2005;2(1):7.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Stahl EC, Brown BN. Cell Therapy Strategies to Combat Immunosenescence. Organogenesis. 2015;11(4):159–72.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Costantini E, D'Angelo C, Reale M. The Role of Immunosenescence in Neurodegenerative Diseases. Mediators Inflamm. 2018;2018:6039171.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Lian J, Yue Y, Yu W, Zhang Y. Immunosenescence: a key player in cancer development. J Hematol Oncol. 2020;13(1):151.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Song Y, Shen H, Schenten D, Shan P, Lee PJ, Goldstein DR. Aging enhances the basal production of IL-6 and CCL2 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2012;32(1):103–9.

    Article  CAS  PubMed  Google Scholar 

  8. Choi IY, Lee C, Longo VD. Nutrition and fasting mimicking diets in the prevention and treatment of autoimmune diseases and immunosenescence. Mol Cell Endocrinol. 2017;455:4–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Salik B, Smyth MJ, Nakamura K. Targeting immune checkpoints in hematological malignancies. J Hematol Oncol. 2020;13(1):111.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Galeano Nino JL, Kwan RY, Weninger W, Biro M. Antigen-specific T cells fully conserve antitumour function following cryopreservation. Immunol Cell Biol. 2016;94(4):411–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Martel J, Ojcius DM, Wu CY, Peng HH, Voisin L, Perfettini JL, et al. Emerging use of senolytics and senomorphics against aging and chronic diseases. Med Res Rev. 2020;40(6):2114–31.

    Article  CAS  PubMed  Google Scholar 

  12. Eckardt KU, Coresh J, Devuyst O, Johnson RJ, Kottgen A, Levey AS, et al. Evolving importance of kidney disease: from subspecialty to global health burden. Lancet. 2013;382(9887):158–69.

    Article  PubMed  Google Scholar 

  13. O'Sullivan ED, Hughes J, Ferenbach DA. Renal Aging: Causes and Consequences. J Am Soc Nephrol. 2017;28(2):407–20.

    Article  CAS  PubMed  Google Scholar 

  14. Sato Y, Yanagita M. Immunology of the ageing kidney. Nat Rev Nephrol. 2019;15(10):625–40.

    Article  PubMed  Google Scholar 

  15. Birch J, Gil J. Senescence and the SASP: many therapeutic avenues. Genes Dev. 2020;34(23–24):1565–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ponnappan S, Ponnappan U. Aging and immune function: molecular mechanisms to interventions. Antioxid Redox Signal. 2011;14(8):1551–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chidrawar SM, Khan N, Chan YL, Nayak L, Moss PA. Ageing is associated with a decline in peripheral blood CD56bright NK cells. Immun Ageing. 2006;3:10.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Reed RG, Al-Attar A, Presnell SR, Lutz CT, Segerstrom SC. A longitudinal study of the stability, variability, and interdependencies among late-differentiated T and NK cell subsets in older adults. Exp Gerontol. 2019;121:46–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Le Garff-Tavernier M, Beziat V, Decocq J, Siguret V, Gandjbakhch F, Pautas E, et al. Human NK cells display major phenotypic and functional changes over the life span. Aging Cell. 2010;9(4):527–35.

    Article  PubMed  Google Scholar 

  20. Almeida-Oliveira A, Smith-Carvalho M, Porto LC, Cardoso-Oliveira J, Ribeiro Ados S, Falcao RR, et al. Age-related changes in natural killer cell receptors from childhood through old age. Hum Immunol. 2011;72(4):319–29.

    Article  CAS  PubMed  Google Scholar 

  21. Gardner A, de Mingo PA, Ruffell B. Dendritic Cells and Their Role in Immunotherapy. Front Immunol. 2020;11:924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sridharan A, Esposo M, Kaushal K, Tay J, Osann K, Agrawal S, et al. Age-associated impaired plasmacytoid dendritic cell functions lead to decreased CD4 and CD8 T cell immunity. Age (Dordr). 2011;33(3):363–76.

    Article  CAS  PubMed  Google Scholar 

  23. Agrawal A, Gupta S. Impact of aging on dendritic cell functions in humans. Ageing Res Rev. 2011;10(3):336–45.

    Article  CAS  PubMed  Google Scholar 

  24. Carbone A, Gloghini A. Follicular dendritic cell pattern in early lymphomas involving follicles. Adv Anat Pathol. 2014;21(4):260–9.

    Article  CAS  PubMed  Google Scholar 

  25. Aydar Y, Balogh P, Tew JG, Szakal AK. Altered regulation of Fc gamma RII on aged follicular dendritic cells correlates with immunoreceptor tyrosine-based inhibition motif signaling in B cells and reduced germinal center formation. J Immunol. 2003;171(11):5975–87.

    Article  CAS  PubMed  Google Scholar 

  26. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116(16):e74–80.

    Article  CAS  PubMed  Google Scholar 

  27. Ong SM, Hadadi E, Dang TM, Yeap WH, Tan CT, Ng TP, et al. The pro-inflammatory phenotype of the human non-classical monocyte subset is attributed to senescence. Cell Death Dis. 2018;9(3):266.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Prattichizzo F, Bonafe M, Olivieri F, Franceschi C. Senescence associated macrophages and "macroph-aging": are they pieces of the same puzzle? Aging (Albany NY). 2016;8(12):3159–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Solana R, Tarazona R, Gayoso I, Lesur O, Dupuis G, Fulop T. Innate immunosenescence: effect of aging on cells and receptors of the innate immune system in humans. Semin Immunol. 2012;24(5):331–41.

    Article  CAS  PubMed  Google Scholar 

  30. Costantini A, Viola N, Berretta A, Galeazzi R, Matacchione G, Sabbatinelli J, et al. Age-related M1/M2 phenotype changes in circulating monocytes from healthy/unhealthy individuals. Aging (Albany NY). 2018;10(6):1268–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Metcalf TU, Wilkinson PA, Cameron MJ, Ghneim K, Chiang C, Wertheimer AM, et al. Human Monocyte Subsets Are Transcriptionally and Functionally Altered in Aging in Response to Pattern Recognition Receptor Agonists. J Immunol. 2017;199(4):1405–17.

    Article  CAS  PubMed  Google Scholar 

  32. Hearps AC, Martin GE, Angelovich TA, Cheng WJ, Maisa A, Landay AL, et al. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell. 2012;11(5):867–75.

    Article  CAS  PubMed  Google Scholar 

  33. Goronzy JJ, Weyand CM. Successful and Maladaptive T Cell Aging. Immunity. 2017;46(3):364–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Masters AR, Haynes L, Su DM, Palmer DB. Immune senescence: significance of the stromal microenvironment. Clin Exp Immunol. 2017;187(1):6–15.

    Article  CAS  PubMed  Google Scholar 

  35. Shanley DP, Aw D, Manley NR, Palmer DB. An evolutionary perspective on the mechanisms of immunosenescence. Trends Immunol. 2009;30(7):374–81.

    Article  CAS  PubMed  Google Scholar 

  36. Palmer S, Albergante L, Blackburn CC, Newman TJ. Thymic involution and rising disease incidence with age. Proc Natl Acad Sci U S A. 2018;115(8):1883–8.

  37. Thomas R, Wang W, Su DM. Contributions of Age-Related Thymic Involution to Immunosenescence and Inflammaging. Immun Ageing. 2020;17:2.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Li G, Yu M, Lee WW, Tsang M, Krishnan E, Weyand CM, et al. Decline in miR-181a expression with age impairs T cell receptor sensitivity by increasing DUSP6 activity. Nat Med. 2012;18(10):1518–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lanna A, Henson SM, Escors D, Akbar AN. The kinase p38 activated by the metabolic regulator AMPK and scaffold TAB1 drives the senescence of human T cells. Nat Immunol. 2014;15(10):965–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wikby A, Johansson B, Olsson J, Lofgren S, Nilsson BO, Ferguson F. Expansions of peripheral blood CD8 T-lymphocyte subpopulations and an association with cytomegalovirus seropositivity in the elderly: the Swedish NONA immune study. Exp Gerontol. 2002;37(2–3):445–53.

    Article  CAS  PubMed  Google Scholar 

  41. Uppal SS, Verma S, Dhot PS. Normal values of CD4 and CD8 lymphocyte subsets in healthy indian adults and the effects of sex, age, ethnicity, and smoking. Cytometry B Clin Cytom. 2003;52(1):32–6.

    Article  CAS  PubMed  Google Scholar 

  42. Nakajima T, Schulte S, Warrington KJ, Kopecky SL, Frye RL, Goronzy JJ, et al. T-cell-mediated lysis of endothelial cells in acute coronary syndromes. Circulation. 2002;105(5):570–5.

    Article  CAS  PubMed  Google Scholar 

  43. Broux B, Markovic-Plese S, Stinissen P, Hellings N. Pathogenic features of CD4+CD28- T cells in immune disorders. Trends Mol Med. 2012;18(8):446–53.

    Article  CAS  PubMed  Google Scholar 

  44. Lolli C, Altavilla A, Conteduca V, Farolfi A, Casadei C, Schepisi G, et al. A comprehensive review of the role of immune checkpoint inhibitors in elderly patients affected by renal cell carcinoma. Crit Rev Oncol Hematol. 2020;153:103036.

    Article  PubMed  Google Scholar 

  45. Kared H, Martelli S, Ng TP, Pender SL, Larbi A. CD57 in human natural killer cells and T-lymphocytes. Cancer Immunol Immunother. 2016;65(4):441–52.

    Article  CAS  PubMed  Google Scholar 

  46. Bulati M, Buffa S, Candore G, Caruso C, Dunn-Walters DK, Pellicano M, et al. B cells and immunosenescence: a focus on IgG+IgD-CD27- (DN) B cells in aged humans. Ageing Res Rev. 2011;10(2):274–84.

    Article  CAS  PubMed  Google Scholar 

  47. Frasca D, Van der Put E, Riley RL, Blomberg BB. Reduced Ig class switch in aged mice correlates with decreased E47 and activation-induced cytidine deaminase. J Immunol. 2004;172(4):2155–62.

    Article  CAS  PubMed  Google Scholar 

  48. Frasca D, Diaz A, Romero M, Blomberg BB. The generation of memory B cells is maintained, but the antibody response is not, in the elderly after repeated influenza immunizations. Vaccine. 2016;34(25):2834–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Colonna-Romano G, Bulati M, Aquino A, Scialabba G, Candore G, Lio D, et al. B cells in the aged: CD27, CD5, and CD40 expression. Mech Ageing Dev. 2003;124(4):389–93.

    Article  CAS  PubMed  Google Scholar 

  50. Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol. 2018;14(10):576–90.

    Article  CAS  PubMed  Google Scholar 

  51. Monti D, Ostan R, Borelli V, Castellani G, Franceschi C. Inflammaging and human longevity in the omics era. Mech Ageing Dev. 2017;165(Pt B):129–38.

    Article  PubMed  Google Scholar 

  52. Esplin BL, Shimazu T, Welner RS, Garrett KP, Nie L, Zhang Q, et al. Chronic exposure to a TLR ligand injures hematopoietic stem cells. J Immunol. 2011;186(9):5367–75.

    Article  CAS  PubMed  Google Scholar 

  53. Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol. 2013;13(12):875–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Stavropoulou E, Kantartzi K, Tsigalou C, Aftzoglou K, Voidarou C, Konstantinidis T, et al. Microbiome, Immunosenescence, and Chronic Kidney Disease. Front Med (Lausanne). 2021;8:661203.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Adler AS, Sinha S, Kawahara TL, Zhang JY, Segal E, Chang HY. Motif module map reveals enforcement of aging by continual NF-kappaB activity. Genes Dev. 2007;21(24):3244–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Salminen A, Huuskonen J, Ojala J, Kauppinen A, Kaarniranta K, Suuronen T. Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res Rev. 2008;7(2):83–105.

    Article  CAS  PubMed  Google Scholar 

  57. Krabbe KS, Pedersen M, Bruunsgaard H. Inflammatory mediators in the elderly. Exp Gerontol. 2004;39(5):687–99.

    Article  CAS  PubMed  Google Scholar 

  58. Singh T, Newman AB. Inflammatory markers in population studies of aging. Ageing Res Rev. 2011;10(3):319–29.

    Article  CAS  PubMed  Google Scholar 

  59. Csiszar A, Ungvari Z, Koller A, Edwards JG, Kaley G. Aging-induced proinflammatory shift in cytokine expression profile in coronary arteries. FASEB J. 2003;17(9):1183–5.

    Article  CAS  PubMed  Google Scholar 

  60. Puzianowska-Kuznicka M, Owczarz M, Wieczorowska-Tobis K, Nadrowski P, Chudek J, Slusarczyk P, et al. Interleukin-6 and C-reactive protein, successful aging, and mortality: the PolSenior study. Immun Ageing. 2016;13:21.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Sepe V, Libetta C, Gregorini M, Rampino T. The innate immune system in human kidney inflammaging. J Nephrol. 2022;35(2):381–95.

    Article  CAS  PubMed  Google Scholar 

  62. Komada T, Muruve DA. The role of inflammasomes in kidney disease. Nat Rev Nephrol. 2019;15(8):501–20.

    Article  PubMed  Google Scholar 

  63. Forman HJ, Maiorino M, Ursini F. Signaling functions of reactive oxygen species. Biochemistry. 2010;49(5):835–42.

    Article  CAS  PubMed  Google Scholar 

  64. Bauer ME, Fuente ML. The role of oxidative and inflammatory stress and persistent viral infections in immunosenescence. Mech Ageing Dev. 2016;158:27–37.

    Article  CAS  PubMed  Google Scholar 

  65. Susantitaphong P, Cruz DN, Cerda J, Abulfaraj M, Alqahtani F, Koulouridis I, et al. World incidence of AKI: a meta-analysis. Clin J Am Soc Nephrol. 2013;8(9):1482–93.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Sawhney S, Marks A, Fluck N, Levin A, McLernon D, Prescott G, et al. Post-discharge kidney function is associated with subsequent ten-year renal progression risk among survivors of acute kidney injury. Kidney Int. 2017;92(2):440–52.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Coca SG, Yusuf B, Shlipak MG, Garg AX, Parikh CR. Long-term risk of mortality and other adverse outcomes after acute kidney injury: a systematic review and meta-analysis. Am J Kidney Dis. 2009;53(6):961–73.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Jin H, Zhang Y, Ding Q, Wang SS, Rastogi P, Dai DF, et al. Epithelial innate immunity mediates tubular cell senescence after kidney injury. JCI. Insight. 2019;4(2).

  69. Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol. 2013;15(8):978–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Legrand M, Bell S, Forni L, Joannidis M, Koyner JL, Liu K, et al. Pathophysiology of COVID-19-associated acute kidney injury. Nat Rev Nephrol. 2021;17(11):751–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Steinmetz OM, Velden J, Kneissler U, Marx M, Klein A, Helmchen U, et al. Analysis and classification of B-cell infiltrates in lupus and ANCA-associated nephritis. Kidney Int. 2008;74(4):448–57.

    Article  CAS  PubMed  Google Scholar 

  72. Carney EF. The impact of chronic kidney disease on global health. Nat Rev Nephrol. 2020;16(5):251.

    Article  PubMed  Google Scholar 

  73. Collaboration GBDCKD. Global, regional, and national burden of chronic kidney disease, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2020;395(10225):709–33.

    Article  Google Scholar 

  74. Clements ME, Chaber CJ, Ledbetter SR, Zuk A. Increased cellular senescence and vascular rarefaction exacerbate the progression of kidney fibrosis in aged mice following transient ischemic injury. PLoS One. 2013;8(8):e70464.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Betjes MG, Langerak AW, van der Spek A, de Wit EA, Litjens NH. Premature aging of circulating T cells in patients with end-stage renal disease. Kidney Int. 2011;80(2):208–17.

    Article  PubMed  Google Scholar 

  76. Meijers RW, Litjens NH, de Wit EA, Langerak AW, van der Spek A, Baan CC, et al. Uremia causes premature ageing of the T cell compartment in end-stage renal disease patients. Immun Ageing. 2012;9(1):19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. George RP, Mehta AK, Perez SD, Winterberg P, Cheeseman J, Johnson B, et al. Premature T Cell Senescence in Pediatric CKD. J Am Soc Nephrol. 2017;28(1):359–67.

    Article  CAS  PubMed  Google Scholar 

  78. Simmons EM, Langone A, Sezer MT, Vella JP, Recupero P, Morrow JD, et al. Effect of renal transplantation on biomarkers of inflammation and oxidative stress in end-stage renal disease patients. Transplantation. 2005;79(8):914–9.

    Article  CAS  PubMed  Google Scholar 

  79. Betjes MG, Huisman M, Weimar W, Litjens NH. Expansion of cytolytic CD4+CD28- T cells in end-stage renal disease. Kidney Int. 2008;74(6):760–7.

    Article  CAS  PubMed  Google Scholar 

  80. Modi ZJ, Lu Y, Ji N, Kapke A, Selewski DT, Dietrich X, et al. Risk of Cardiovascular Disease and Mortality in Young Adults With End-stage Renal Disease: An Analysis of the US Renal Data System. JAMA Cardiol. 2019;4(4):353–62.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Skov Dalgaard L, Norgaard M, Jespersen B, Jensen-Fangel S, Ostergaard LJ, Schonheyder HC, et al. Risk and Prognosis of Bloodstream Infections among Patients on Chronic Hemodialysis: A Population-Based Cohort Study. PLoS One. 2015;10(4):e0124547.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Xiang F, Chen R, Cao X, Shen B, Chen X, Ding X, et al. Premature aging of circulating T cells predicts all-cause mortality in hemodialysis patients. BMC Nephrol. 2020;21(1):271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Okba AM, El Raouf A, Raafat M, Nazmy Farres M, El Nour A, Melek N, et al. Expanded peripheral CD4(+)CD28(null) T cells and its association with atherosclerotic changes in patients with end stage renal disease on hemodialysis. Hum Immunol. 2019;80(9):748–54.

    Article  CAS  PubMed  Google Scholar 

  84. Carracedo J, Ramirez R, Soriano S, Alvarez de Lara MA, Rodriguez M, Martin-Malo A, et al. Monocytes from dialysis patients exhibit characteristics of senescent cells: does it really mean inflammation? Contrib Nephrol. 2005;149:208–18.

    Article  PubMed  Google Scholar 

  85. Tullius SG, Tran H, Guleria I, Malek SK, Tilney NL, Milford E. The combination of donor and recipient age is critical in determining host immunoresponsiveness and renal transplant outcome. Ann Surg. 2010;252(4):662–74.

    Article  PubMed  Google Scholar 

  86. Trzonkowski P, Debska-Slizien A, Jankowska M, Wardowska A, Carvalho-Gaspar M, Hak L, et al. Immunosenescence increases the rate of acceptance of kidney allotransplants in elderly recipients through exhaustion of CD4+ T-cells. Mech Ageing Dev. 2010;131(2):96–104.

    Article  CAS  PubMed  Google Scholar 

  87. Daniel L, Tassery M, Lateur C, Thierry A, Herbelin A, Gombert JM, et al. Allotransplantation Is Associated With Exacerbation of CD8 T-Cell Senescence: The Particular Place of the Innate CD8 T-Cell Component. Front Immunol. 2021;12:674016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Luttropp K, Nordfors L, McGuinness D, Wennberg L, Curley H, Quasim T, et al. Increased Telomere Attrition After Renal Transplantation-Impact of Antimetabolite Therapy. Transplant Direct. 2016;2(12):e116.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Schaier M, Leick A, Uhlmann L, Kalble F, Morath C, Eckstein V, et al. End-stage renal disease, dialysis, kidney transplantation and their impact on CD4(+) T-cell differentiation. Immunology. 2018;155(2):211–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Alimbetov D, Davis T, Brook AJ, Cox LS, Faragher RG, Nurgozhin T, et al. Suppression of the senescence-associated secretory phenotype (SASP) in human fibroblasts using small molecule inhibitors of p38 MAP kinase and MK2. Biogerontology. 2016;17(2):305–15.

    Article  CAS  PubMed  Google Scholar 

  91. Yaribeygi H, Butler AE, Atkin SL, Katsiki N, Sahebkar A. Sodium-glucose cotransporter 2 inhibitors and inflammation in chronic kidney disease: Possible molecular pathways. J Cell Physiol. 2018;234(1):223–30.

    Article  PubMed  Google Scholar 

  92. Okabe C, Borges RL, de Almeida DC, Fanelli C, Barlette GP, Machado FG, et al. NF-kappaB activation mediates crystal translocation and interstitial inflammation in adenine overload nephropathy. Am J Physiol Renal Physiol. 2013;305(2):F155–63.

    Article  CAS  PubMed  Google Scholar 

  93. Li J, Qu X, Ricardo SD, Bertram JF, Nikolic-Paterson DJ. Resveratrol inhibits renal fibrosis in the obstructed kidney: potential role in deacetylation of Smad3. Am J Pathol. 2010;177(3):1065–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Schroth J, Thiemermann C, Henson SM. Senescence and the Aging Immune System as Major Drivers of Chronic Kidney Disease. Front Cell Dev Biol. 2020;8:564461.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Lu H, Wu L, Liu L, Ruan Q, Zhang X, Hong W, et al. Quercetin ameliorates kidney injury and fibrosis by modulating M1/M2 macrophage polarization. Biochem Pharmacol. 2018;154:203–12.

    Article  PubMed  Google Scholar 

  96. Hickson LJ, Langhi Prata LGP, Bobart SA, Evans TK, Giorgadze N, Hashmi SK, et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine. 2019;47:446–56.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ, et al. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8(+) T cells. J Clin Invest. 2014;124(9):4004–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

Not applicable.

Funding

The work was supported by Foundation of Key R&D plan of Sichuan Province (2019YFS0538); National Natural Science Foundation of China (82200810). The project of 2020 High-level Overseas Chinese Talent Returning Funding; Foundation of Applied Basic Research Project of Sichuan Provincial Science and Technology (2020YJ0179); Foundation for Young Talent Fund of Sichuan Provincial People's Hospital (2022QN02); Discipline construction fund of Sichuan Provincial People's Hospital.

Author information

Authors and Affiliations

Authors

Contributions

Mingxuan Chi, Zijun Tian and Kuai Ma were involved in writing the article. Yunlong Li, Moussa Ide Nasser and Chi Liu critically revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Moussa Ide Nasser or Chi Liu.

Ethics declarations

Ethics approval and consent to participate

This article does not contain any studies with human participants/animals performed by any of the authors.

Consent for publication

All authors approve and consent for publication.

Competing interests

All the authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chi, M., Tian, Z., Ma, K. et al. The diseased kidney: aging and senescent immunology. Immun Ageing 19, 58 (2022). https://doi.org/10.1186/s12979-022-00313-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12979-022-00313-9

Keywords