Next Article in Journal
The Probiotic Properties and Safety of Limosilactobacillus mucosae NK41 and Bifidobacterium longum NK46
Previous Article in Journal
A Point Mutation in Cassette Relieves the Repression Regulation of CcpA Resulting in an Increase in the Degradation of 2,3-Butanediol in Lactococcus lactis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Immune System Aging on Infectious Diseases

by
Eugenia Quiros-Roldan
1,
Alessandra Sottini
2,
Pier Giorgio Natali
3 and
Luisa Imberti
4,*
1
Department of Infectious and Tropical Diseases, ASST- Spedali Civili and DSCS- University of Brescia, 25123 Brescia, Italy
2
Clinical Chemistry Laboratory, Services Department, ASST Spedali Civili of Brescia, 25123 Brescia, Italy
3
Mediterranean Task Force for Cancer Control (MTCC), Via Pizzo Bernina, 14, 00141 Rome, Italy
4
Section of Microbiology, University of Brescia, P. le Spedali Civili, 1, 25123 Brescia, Italy
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(4), 775; https://doi.org/10.3390/microorganisms12040775
Submission received: 1 March 2024 / Revised: 22 March 2024 / Accepted: 9 April 2024 / Published: 11 April 2024
(This article belongs to the Section Molecular Microbiology and Immunology)

Abstract

:
Immune system aging is becoming a field of increasing public health interest because of prolonged life expectancy, which is not paralleled by an increase in health expectancy. As age progresses, innate and adaptive immune systems undergo changes, which are defined, respectively, as inflammaging and immune senescence. A wealth of available data demonstrates that these two conditions are closely linked, leading to a greater vulnerability of elderly subjects to viral, bacterial, and opportunistic infections as well as lower post-vaccination protection. To face this novel scenario, an in-depth assessment of the immune players involved in this changing epidemiology is demanded regarding the individual and concerted involvement of immune cells and mediators within endogenous and exogenous factors and co-morbidities. This review provides an overall updated description of the changes affecting the aging immune system, which may be of help in understanding the underlying mechanisms associated with the main age-associated infectious diseases.

1. Introduction

Aging is not only a gradual and irreversible pathophysiological process, but also a complex phenomenon from a social, psychological, and biological point of view. Since it can impact the functions of many organs and systems, it represents a major risk factor for the onset and/or progression of a wide spectrum of aging-related disorders [1]. As life expectancy is predicted to increase, understanding the mechanisms leading to immune senescence is of major public health relevance, fostering the development of more focused preventive actions, as well as the development of new preventive and therapeutic interventions.
Aging has gone through evolving definitions over the years, such as “a persistent decline in the age-specific fitness components of an organism due to internal physiological deterioration” [2] or “a progressive loss of function accompanied by decreasing fertility and increasing mortality with advancing age” [3]. More recently, a consensus has been reached on framing aging as a “normal way of functioning in biology from a certain age onwards” [4]. For instance, aging in different organisms (especially mammals) has been initially proposed to encompass nine common features, including genomic instability, telomere shortening attrition, epigenetic alteration, deregulation of nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, alteration of intercellular communication, and loss of proteostasis [5]. Over the years, the finding that diminished proteostasis is different from compromised autophagy has enabled the inclusion of the latter in the hallmarks of aging [6]. In addition, three other distinctive derangements, such as alteration of metabolic pathways, adaptation to stress, and, more importantly, chronic inflammation, have been added to the list [7]. These hallmarks, which are strongly correlated with each other, satisfy the following three premises: 1. they are aging-related; 2. the possibility of experimentally accelerating aging by acting on these markers; and 3. the opportunity to decelerate, stop or reverse aging with a combination of preventive and therapeutic interventions.
In humans, senescent cells of various lineages accumulate at various body sites at different rates, from 2- to 20-fold, when comparing young (<35 years) to old (>65 years) healthy individuals [8]. Although all cell types can undergo senescence during aging, this process mainly affects fibroblasts, endothelial cells, chondrocytes, tenocytes, skin and immune cells [9,10,11]. The immune system is one of the most ubiquitous systems of the organism, which can protect the human body from internal or external agents and interacts with neural, circulatory and other systems. Therefore, its alteration may result in increased incidence of many age-related diseases.

2. Aging of Immune System Components in Physiological Conditions

Age-related immune system changes are grouped under the term “immunosenescence”, used over the years to define “the state of dysregulated immune function that contributes to the increased susceptibility of the elderly to infections and, possibly, autoimmune diseases and cancer” [12]; “the changes in the immune system associated with age” [13]; and “a gradual and subjective decline of the immune system and host defense mechanisms” [14]. More generally, immunosenescence refers to a series of complex changes leading to altered innate and adaptive immune system functions which, overall, may result in a state of immunodeficiency [15]. Immunosenescence has been considered a harmful event for many years, due to its involvement in the progressive reduction in the ability to trigger effective antibody and cellular responses against infections and vaccinations. It has also been considered a negative process because it leads to a type of inflammation called “inflammaging”, which defines a reduction in the capability to cope with a variety of stressors and a progressive increase in the pro-inflammatory status [16]. Indeed, inflammaging is characterized by an unbalanced increase in systemic pro-inflammatory cytokines, such as interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF)-α, and reduced levels of anti-inflammatory cytokines, such as IL-10, transforming growth factor-β [17]. Therefore, inflammaging is a chronic, asymptomatic, sterile, low-grade inflammation that occurs in old age in the absence of signs of infection. This chronic inflamed state was initially described as a harmful event with profound adverse health effects that contributed to biological aging and the development of age-related pathologies. In particular, senescent immune cells, by producing cytokines, chemokines, growth factors, proteases, and angiogenic factors, acquire the so-called “senescence-associated secretory phenotype” (SASP) [18]. The persistent and non-resolved production of pro-inflammatory mediators, concomitantly with the adoption of lifestyle factors, including smoking, obesity, alcohol, lack of exercise, and exposure to ultraviolet radiation, are known to increase the risk for age-related multi-organ/system diseases (Figure 1) [15,19,20,21,22] and premature morbidity and mortality.
In recent years, the perception regarding immunosenescence and inflammaging events has changed considerably. Indeed, while the accumulation of pro-inflammatory factors and inflammaging were suggested as standing at the origin of most diseases of the elderly, age-related inflammation showed a closer correlation with longevity than any other parameters [22,23]. In addition, age-related thymic involution (see below), which leads to a restriction of the T-cell receptor (TCR) repertoire, may also result in lower energy consumption, favoring other body survival-supportive functions and activities [23]. Therefore, it has been proposed that, without the existence of the immunosenescence/inflammation binomial, which represents two aspects of the same phenomenon, human longevity would be greatly reduced. From an evolutionary point of view, immunosenescence is an optimization of the resources of the aging body, even if it ultimately may lead to pathologies and death [23]. An alternative possibility is that immunosenescence is the phenomenon in which adaptive immunity decreases over time, while inflammaging is a phenomenon in which innate immunity is activated [24].
According to this theory, inflammaging and immunosenescence progress in parallel, sustaining a mutually maintained vicious loop: the increased release of inflammatory mediators induced by inflammaging contributes to inhibiting the adaptive immune system and promoting immunosenescence; vice versa, the decrease in the adaptive immune system response strengthens the stimulation of the innate immune system (in an attempt to protect the organism) and maintains inflammaging. One of the most recent hypotheses on the meaning of immune system age-related changes is that these modifications are linked to the immune system response to permanent stress. This novel vision considers these changes as a permanent process of adaptation to stress that could have either beneficial or harmful consequences, depending on both genetic and environmental exposure. The prevalence of one of the conditions may result either in healthy longevity or pathological aging burdened by age-related diseases [25].
Overall, aging of the immune system in physiological conditions involves complex alterations in both innate and adaptive immunity, leading to decreased immune responsiveness, increased susceptibility to harmful agents and infections, and impaired vaccine efficacy. Understanding these changes is essential for developing strategies to promote healthy aging and improve immune function in older adults.

2.1. Age-Associated Changes in the Immune Compartments

Several age-related modifications affect the innate and adaptive immune system [26,27]. In aging bone marrow, hematopoietic stem cells show reduced self-renewal potential and increased skewing toward myelopoiesis [28], probably because of compromised Pax5 expression and Rag2 recombinase function and bone marrow niche alterations [29]. The number of circulating neutrophils does not significantly change [30], although the function of these cells may be compromised in aging, since activated neutrophils are more prone to apoptosis and deficient in phagocytosis and chemotaxis [29]. In addition, aging, by hampering neutrophil extracellular traps (NETs) efficacy, reduces NETosis, the neutrophil-mediated defense mechanism in which DNA and enzymes are extruded, forming a network trapping and killing different pathogens [31].
Aged tissue-derived macrophages may display compromised phagocytosis and an altered response to lipopolysaccharide [32]. Macrophages, upon stimulation, may undergo pyroptosis, also known as cellular inflammatory necrosis, which is different from other kinds of cell death. Pyroptosis is thought to play a key role in the clearance of infectious agents by exposing pathogenic antigens to the adaptive immune system and secreting cytokines and eicosanoids to promote inflammatory and repair responses [33]. More recently, the role of pyroptosis in the age-related diseases has attracted increased research attention. However, the elucidation of pyroptosis pathophysiology is still a work in progress, and it is not known whether modifications of a specific pyroptotic pathway may be beneficial, or may upregulate other pathways implicated in age-associated diseases that may paradoxically exacerbate disease progression [34].
Although information regarding the numbers and performance of dendritic cells (DCs) is relatively scarce, the number of circulating conventional and plasmacytoid DCs appears to be reduced in frail, healthy elderly [35]. While natural killer (NK) cells significantly increase, with the majority of these cells being CD56dim, their proliferative ability declines with age [36].
One of the main and early investigated adaptive immune system impairments occurs as a result of thymic involution, such as loss of organ mass, disruption of architecture, enhancement of perivascular space, and increased adiposity, which fail to meet the demand for new T-cell output [37,38]. Data resulting from the measurement of thymic emigrants by flow cytometer [39,40,41] or TCR excision circles (TRECs) by real-time PCR [37,42,43,44] have demonstrated that the release of new T cells decreases progressively during aging. The holes created in the lymphocyte compartment by low thymic output lead to a gradual increase in effector memory cells, thus old people have low numbers of naïve T cells and a high number of memory T lymphocytes, mainly cytotoxic CD8+ cells in an advanced stage of differentiation [13]. In addition, a restriction on TCR repertoire diversity [45], both in memory CD4+ and CD8+ T cells, occurs [46]. It is of interest that the accumulation of highly differentiated T cells and the significant reduction in naïve T cells are not observed in long-lived individuals [47], so centenarians show unexpectedly larger TCR repertoires [48].
The production of B lymphocytes, as well as of myeloid cells, is impaired in aged bone marrow [29]. Although the output of new B cells, quantified by measuring K deleting recombinant excision circles (KRECs), remains conserved [49], with the peripheral number unchanged, aging is associated with a decline in the frequency of naïve B cells and in the percentage of switch memory B cells [50,51]. Moreover, the accumulation of a subset of atypical B cells, termed age-associated B cells (ABCs), characterizes the aging B-cell compartments. These cells have distinct phenotypes, gene expression profiles, special survival requirements, variations in B-cell receptor repertoires, and unique functions [52]. As a result, antibody affinity and diversity also decline, leading to impaired antibody responses [53]. The exact cause and significance of all these changes are not clear, but alterations to immune cell signaling may be one of prominent cause of malfunctioning immunity, as extensively discussed in Fulop et al. [54].
Figure 2 summarizes the principal age-related modifications of the innate and adaptive immune system cells.
Aging affects several immune system tissues and organs. Increased architectural (as well as vascular) fibrosis induces a progressive reduction in lymph nodes number and size, leading to reduced local cell traffic and impaired intercellular interactions [55,56,57]. In addition, changes in the local production of adequate amounts of chemokines and cytokines were observed, which ensured an efficient immune response [57,58]. Aging also causes deterioration in the spleen. This organ preserves the physiologic populations of white blood cells and platelets that, mobilized by pathogenic invaders in the blood, sustain a protective immune response [59]. With advancing age, the sinusoidal stromal cell linings, at the border between the follicular and marginal zones, become disorganized, leading to altered immune system cell localization, resulting in an improper antigen-presenting capacity [60,61]. Finally, evidence has been gathered of an overall decline in mucosal immunity, especially in the gastrointestinal tracts of the elderly [62].

2.2. Sex-Differences in Innate and Adaptive Immunity Increase with Age

As sex differences characterize the immune system during the entire life course, further ones occur during aging, with older men displaying higher monocyte activity [63], expression of myeloid cell-related genes [64], and a faster decline in the number and function of B and T cells [63].
Aging female T cells produce more IL-10 [65], capable of neutralizing age-related inflammaging. The high humoral response observed in women can favor the appearance of autoreactive clones [66], as supported by epidemiological studies showing that about 80% of autoimmune diseases occur in females at earlier age [67]. Further, immunosenescence develops later and slower in women [68], and this has been associated with their longer life expectancy [69,70]. Unfortunately, this longer life expectancy is associated with worse health conditions [17].
Although detailed information is lacking regarding sex differences in immune performance, a number of advances have been made. A study, enrolling 500 males and 500 females aged between 20 and 70, tested eight lymphocyte subpopulations, 3 million single nucleotide polymorphisms, and 560 genes of the immune system before and after stimulation with a series of infectious agents and a superantigen. The data demonstrated that age preferentially influences CD8+ cells, while sex has a specific influence on CD4+ cells and macrophages, as well as multiple genes related to the immune system. In contrast, genetic factors are correlated with only a few changes in immune system genes, but these changes are extremely puissant [71]. Another study, performed with Transposase Accessible Chromatin with Sequencing (ATAC-seq), demonstrated that the switching on and off of some genes follows different individual patterns, with a random array of activation. In men and women, the turning on and silencing of immune system genes occur through different “switches”, which partially explains why the two sexes may differ in percentages and types of immune players [72]. Although both sexes equally respond to drugs targeting immunosenescence [68], women are reported to be more sensitive to immunostimulating strategies [73]. Understanding how sex differences in innate and adaptive immunity evolve with age is important for developing personalized approaches to health and vaccination strategies.

2.3. Immune System Complexity Increases with Age

Since only 30% of the variations observed in the immune system appear to be due to hereditary factors [74], a model has been proposed in which the immune response evolves over time in main waves shaped by environmental stimuli, leading to divergences in the immune system that become more and more marked as the years pass [75]. The first dramatic change occurs in the newborn, requiring protection from invasive pathogens and tolerance to “beneficial” pathogens. Cord blood cells show extreme individual variability, which flattens during the first weeks of life, including an initial short-lasting neutrophil expansion, and the increase in CD4+ and CD8+ lymphocytes and B cells occurring in all children [76]. B cells, NK cells and DCs reach adult-like maturation status in the first trimester, following increased exposure to a variety of environmental agents. The progressive senescence of the immune system during adulthood occurs earlier than was previously appreciated, most likely because comparative analyses were performed between young subjects and very old people, thus missing the intermediate age [74,77]. The limit of this comparison was bypassed by comparing homozygous twins with an average age of 48 years with those aged between 12 and 30 years. The study demonstrates that the inter-individual variability of most immune system subpopulations starts before the age of 50 [78], well before the age of 60, conventionally established as the initiation of immunosenescence.
One should remember that, to appreciate significant changes related to IS, long-term follow-up analyses should be performed, since a rather stable immune system cell composition is commonly observed in the short period of observation [79]. Indeed, a trend of naïve CD8+ cell expansion in the young and a decrease in the elderly was observed in a 10-year follow-up. However, this does not necessarily translate to the individual level: for example, naïve CD8+ T cells decline over time at the group level, but not individually, because in some cases, the variability over time proceeds in an opposite direction. This individual variability has been observed for other subpopulations, such as B cells, CD8+CD28− T cells, CD8+ T cells expressing the CD57 marker, and NK cells [77]. Current knowledge on human immune variability comes mainly from well-selected population-based cohort studies. Therefore, to promote a better understanding of the impact of immune variability on aging, these approaches must be extended to populations of different backgrounds, life stages, lifestyle, and environments.

2.4. Biomarkers of Aged Immune System

As described above, the immune system ages and the changes that occur can be highlighted by studying specific markers of aging. Converging literature recognizes IL-6 and TNF-α as relevant biomarkers for the aging innate immune system [80]. While IL-6 serum concentrations in healthy individuals are undetectable or minimal, elevated levels have been reported, especially in advanced age, to be associated with disability and mortality; TNF-α age-related changes behave similarly [81]. The production of other key cytokines important for immune modulation, including IL-7, IL-11, IL-15, and granulocyte-macrophage colony-stimulating factor, is also affected by aging [29,80]. Although total white blood cell counts and lymphocyte counts have been the most studied biomarkers of adaptive immunity, they can further differentiate after their maturation in response to pathogens, so it is unclear when senescence is induced in these cells [82]. Similarly, C-reactive protein plasma level, which is a frequently used screening test in daily clinical practice, seems to be a rather unspecific biomarker of the aging immune system.
Telomere length measure, recognized as one of the most reliable biomarkers of aging [83], provides a rough estimate of the rate of the immunosenescence process and can hardly be regarded as clinically relevant [84]. More informative is the quantification of TRECs and KRECs [43], which mirrors the capability to homeostatically replenish both lymphocyte pools. A decline in the diversity of the TCR repertoire owing to thymic involution has been implicated as causing defective immune responses in the elderly [46].
Upfront to a lack of immune cell global markers of senescence, individual immune system cell lineages have been identified. T cells of aged individuals express a CD27−CD28−CD57+ killer cell lectin-like receptor G1(KLRG-1)+ or C-C chemokine receptor 7 (CCR7)−CD45RA+ phenotype. These cells can also express T-cell immunoglobulin and mucin domain-containing 3 (Tim-3), T-cell immunoreceptor with Ig and ITIM domains (TIGIT), immunoglobulin-like transcript 2 (ILT2/CD85j), or other NK-like receptors. Unsettled is the issue of whether T cells really express exhausted markers such as programmed cell death protein 1 (PD-1) and lymphocyte-activation gene 3 (LAG-3) [85]. With aging, a progressive increase in terminal effector memory T (TEMRA) cells occurs [86]. These T cells, which are CCR7-, re-express the CD45RA marker and represent terminally differentiated effector cells that result from a protracted antigen exposure. Overall, TEMRA cells, characterized by a decline in proliferation potential, efficient cytotoxicity, pro-inflammatory activity, and high clonal expansion, are considered hallmarks of immunosenescence [87,88]. Finally, senescent T cells also become positive for senescence-associated β-galactosidase (saβ-gal) staining, upregulate p53, p21, and p16, downregulate cyclin-dependent kinase (Cdk) 2, Cdk6, and cyclin D3, and show SASP [85].
Since aging negatively affects the production of B cells in the bone marrow, the number of B-1 and B-2 cells is decreased in peripheral blood, counterbalanced by the increase in ABCs [52]. Although the phenotype of surface markers is still poorly defined, ABCs could be distinguished from other B-cell subsets because they express CD11b, CD11c, and T-bet markers, as well as innate activation stimuli, such as Toll-like receptor 7 signals, but not CD43 and CD5 molecules, which are present on B-1 cells [53].
Monitoring these biomarkers can provide insights into the aging immune system and help identify individuals who may be at increased risk for age-related immune dysfunctions and related health problems.

2.5. The Systems Immunology for the Study of Age-Related Immune System Variations

The “classic” study methods, resorting to the use of animal models, despite having highly contributed to unveiling relevant paradigms in immune system, have some imbedded limitations, such as genetic homogeneity, a synchronized day–night cycle, monotonous feeding, and exposure to a narrow spectrum of environmental stimuli. Therefore, animal modes lack those conditions that are so important for shaping the human immune repertoire [89]. Some investigations have used wild or pet store mice, which are maintained in hygienic conditions, which may recapitulate adult human immune challenges [90]. More recently, the issue of immune system variability has been addressed by a new approach, defined as “system immunology”, a branch of “systems biology” [91]. This methodology is based on the concept that various components of the immune system have a high degree of interdependence and interconnection. In a multihierarchical or multiscale organization, the components at a lower scale are integrated with the functional units of the immediately higher scale [92]. The variation in cell composition, plasma proteins, and functional responses in aging through a system immunology approach has been possible due to the introduction of high-throughput “omics” technologies [93]. This approach allows us to study, at both the single cell and population level, the behavior of genes and epigenetic modifications (genomics and epigenomics), mRNA (transcriptomics), and proteins (proteomics), and to quantify, in blood and tissues, the levels of immune system components, as well as the immune system cell markers, as summarized in Figure 3.
Advancements in mathematical and computational methods allow us to study the interactions within cellular and molecular immune system networks. This provides unprecedented information on the human immune response following vaccinations, infections, autoimmunity, and tumors [94]. Many of these improvements have been optimized for studies at the single cell level [95], especially to characterize immune system cells that are located in multiple sites, i.e., blood, primary and secondary lymphoid organs, respiratory and gastrointestinal tissues (lungs, intestine, pancreas, and liver), and barrier sites (skin and mucosal surfaces), where the composition, phenotype, function, and tissue-specific adaptations of different immune cell populations are known to differ [96]. One study that relies on use of these new technologies in addressing immunosenescence is the S3WP program, consisting of a cohort of 101 Swedish individuals between 50 and 65 years of age, followed longitudinally for 2 years. In this program, repeated immune cell profiles, proteomics, transcriptomics, lipidomics, metabolomics, and autoantibody data were integrated with information on gut microbiota composition, routine clinical chemistry, and various clinical parameters [97].

3. Aging of Immune System Components in Infectious Diseases

The interplay between immune senescence, inflammaging, and infections is complex and multifaceted, shaping the susceptibility, severity, and outcomes of infectious diseases in aging individuals. It has become a growing field of research aimed at clarifying whether pathogens accelerate the aging of immune system cells by inducing the expression of molecular determinants overlapping those associated with cellular aging, or whether the age-induced immune changes facilitate infection burden.
Infectious diseases, such as influenza and pneumonia, are among the main global infectious killers of the elderly, with a rate of 93.2 deaths per 100,000 in 2018 for those aged ≥65 years [98]. Moreover, infections by new pathogens, such as West Nile virus (WNV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for the coronavirus disease 2019 (COVID-19) pandemic, have shown heightened severity in the older population [99,100]. Overall changes in non-immune organs also concur with the age-related proneness to infectious diseases. For instance, in the lungs, decreased respiratory muscle strength reduces lung compliance, and impaired muco-ciliary function leads to inefficient clearance of infectious organisms [101]. Similarly, the decreased function of the epithelial barriers of the skin, lungs, and gastrointestinal tract favors the pathogen invasiveness [102]. Furthermore, neurocognitive modifications and increased vulnerability to metabolic encephalopathy contribute to the delayed recognition of infectious syndromes [103]. This is further aggravated by the frequent atypical clinical presentation of infections in frail older adults, often identified at the time when the underlying disease worsens or when an additional precipitating event occurs [104].
However, it is now recognized that age-related changes in immunity play a major role in determining the incidence and severity of infections and influencing spontaneous- or vaccine-induced immunity. In addition, persistent infections result in a prolonged antigenic overload of the immune system, leading to host T-cell exhaustion [105] and the promotion and/or acceleration the progressive accumulation of cells with an immunosenescent phenotype [106], thus easing the development of a pro-inflammatory milieu that favors aging-related diseases [107,108]. Therefore, both chronic infection and the related immune response contribute to accelerating aging [109]. This acceleration is anticipated to be largely variable, being related to the variety of inducing environmental factors, type, intensity, duration, and temporal sequence of antigen exposure, the so called “exposome” [110], implying that that aging is an individual phenomenon. Indeed, while chronological age is strongly correlated with the prevalence and severity of infections, it may not be the only cause of the higher susceptibility. In addition, the probability of a serious infection does not increase on a yearly basis, but it is closely related to concomitant chronic diseases that induce a worsening of the inflammatory state. Therefore, an elderly but healthy person may be more protected from serious infections than a younger person in poor health [111].
While several studies have focused on chronic diseases in the elderly, less is known about the epidemiological characteristics of their infectious diseases. In a French survey performed in 2005, the top five infections included, with decreasing incidence: urinary tract infection (UTI), respiratory tract infection, skin and soft tissue infections, surgical site infections, and bacteremia [112]. Also, intra-abdominal infections, such as cholecystitis, diverticulitis, appendicitis, abscesses, infectious endocarditis, bacterial meningitis, tuberculosis, and herpes zoster, have been reported to frequently occur in the elderly [113]. More recently, the availability of electronic records and health data platforms outlined the epidemiological characteristics and changes in infectious diseases among the elderly in the Shandong province of China [114]. This study outlined that respiratory infections have the highest incidence in this population, followed by mucocutaneous, blood- and sex-transmitted, gastrointestinal, and vector-borne infections. The most frequent respiratory diseases were influenza, pneumonia, pulmonary tuberculosis, cryptococcosis, whooping cough, and aspergillosis. Typhoid was by far the most frequent gastrointestinal infectious disease, followed by other infectious diarrheas, paratyphoid, ascariasis, bacteria and amoebic dysentery. Herpes zoster, viral conjunctivitis trachoma, common warts, and herpes simplex infections were prevalent among mucocutaneous diseases, while hepatitis B, syphilis, gonorrhea, and hepatitis C were listed as the most frequent blood- and sex-transmitted infections [114]. This study also reported that the incidence of infections was prevalent in elderly males.

3.1. Viral Infections

Viral infections represent one of the most important drivers of premature aging.
Human immunodeficiency virus (HIV) infection has been the first described as associated with immunosenescence [115]. Premature aging is also a consequence of highly active antiretroviral therapy, which has prolonged the survival of these patients despite a persistent inflammation, fueled by high levels of IL-6 and TNF-α, interferon (IFN)-α, reduced production of IL-2 [116,117,118], as well as by a variable decrease in the number of TRECs [37,119,120]. Using an assay that simultaneously measures TRECs and KRECs [43,49], we investigated the bone marrow and thymic output in treated HIV-infected patients and in those ones not requiring therapy. Cells containing KRECs remained unchanged after one year of therapy, then decreased to levels that were similar to those of untreated HIV+ patients. TRECs production increases following a successful antiretroviral therapy response, but never reaches the levels of HIV-1-infected patients not requiring therapy [121]. During this infection, an expansion of TEMRA cells expressing exhaustion markers has been described, strongly suggesting the involvement of T-cell-mediated immune responses in the infection associated immunosenescence. The inflammatory microenvironment of these patients may also drive TEMRA cell evolution towards non-specific senescence [88]. Very recently, the level of immunosenescence was investigated based on CD27 and CD57 expression on T cells in subjects living with HIV/AIDS. These subjects showed a lower proportion of T cells in the early stages of senescence and a higher proportion of T cells in the intermediate and final stages of aging [122]. The lowering of thymic output and the increase in TEMRA cells can be responsible for the observed oligoclonal expansions in both CD4+ and CD8+ T-cell compartments and a reduction in TCR diversity [123,124].
NK cell senescence in chronic HIV-infected patients is documented by the expansion of non-functional CD3−CD56−CD16+ NK cells displaying an activated profile, as indicated by the higher levels of cytokines and chemokines production, such as IL-4, IL-5, IL-6, IL-10, IL-12, IFN-α2, IFN-γ, TNF-α, RANTES, and MCP-1 [125]. Therefore, during HIV infection, the immunosenescence process is accelerated, resulting not only in an immunosuppressed state unable to contain HIV replication and spread, but also to a lower capacity to respond to new antigenic challenges favoring other infections and age-associated organ diseases [126].
For a discussion of the role of aging in response to severe acute respiratory syndrome coronavirus 2 infection, we recommend the reader refer to recent contributions [118,127,128].
As anticipated, the elderly population is more frequently prone to infections of the respiratory system caused by adenovirus, coronaviruses, human metapneumovirus, influenza A and B, parainfluenza, and respiratory syncytial virus (RSV). While in many countries, the vaccination rate is over 60% in subjects older than 65 years [129], influenza remains a serious health threat to this population. Disability and mortality outcomes are dependent on the level of frailty, cardiovascular events that are the most common extra-pulmonary complications, and diminished quality of life due to loss of independence following hospitalization. Unfortunately, large gaps exist between our understanding of the immune response to a natural influenza infection and vaccination, although immunity appears to be more robust and longer-lived in natural infections than following vaccinations [130]. Undoubtedly, a failure of innate immunity is also likely to play a role because of the decrease in macrophage functions [131,132], and of reduced uptake of opsonized bacteria by CD14+ monocytes [133]. Aging appears to affect DCs, in terms of a decrease in number, activity, and migration, determining poorer viral clearance as well as clinical outcomes and susceptibility to complications, which have been correlated to the levels of circulating inflammatory cytokines [134]. Although a diminished CD8+ T-cell immune response to influenza virus infection [135] has been demonstrated in the past in mice, only very recently has the role of CD8+ T cells been elucidated as a mechanism of respiratory viral infection severity in the elderly [136]. Single-cell profiling analysis demonstrated that CD8+ T cells specific for the major influenza HLA-A*02:01-M158-66 (A2/M158) epitope are similar in infants, children, and old humans. However, although CD8+ T cells of old adults displayed no signs of exhaustion, their suboptimal TCRαβ signatures led to less proliferation, polyfunctionality, avidity and recognition of peptide mutants. In particular, the reduced public TCRαβ clonotypes and TCRαβ diversity within older TCR repertoires explains why older adults, in the absence of pre-existing antibodies, are at higher risk of severe disease during influenza epidemics and pandemics [137]. Elderly people often have recurrent clinically mild RSV infections [138], which occasionally may cause altered airway resistance and the worsening of chronic obstructive lung disease [139], leading to serious or life-threatening pneumonia [140]. This population has low levels of serum neutralizing antibodies and IFN-γ and diminished in vitro cellular immune responses to RSV [141]. Data from a recent multiparametric immunological analysis suggest a primary role of cellular immunity in preventing symptomatic RSV infections in these patients. The study identified certain RSV-specific effector memory T-cell populations that are associated with the prevention of symptomatic infection, thus representing a potential marker of innate immunity to RSV in older age [142]. In addition, a systematic literature review of the risk factors for poor outcomes in RSV infection found a strong link between immunosenescence and preexisting co-morbidities (cardiac, pulmonary, and immunocompromising conditions, diabetes, and renal disease), and living conditions (socioeconomic status and nursing home residence) [143]. Other respiratory viruses, such as human metapneumovirus, human rhinovirus, and human parainfluenza virus, also lead to substantial morbidity and mortality in the elderly [144], but the impact of senescence and other factors modulating their severity in the elderly remain to be identified.
Despite being extensively investigated, no conclusive data is available regarding how cytomegalovirus (CMV) infection influences the immune response in the elderly and how it can trigger an increase in pro-inflammatory cytokines, promoting inflammaging [145]. Most likely, CMV infection contributes to age-associated changes in adaptive immunity by modulating the frequency and the cytotoxic capacity of NK cells. CMV seropositive elderly donors have a decreased percentage of both CD16− and CD16+ CD56bright NK cells, and an increase in the CD56−CD16+ subset [146]. An exploratory study demonstrated that CMV-specific CD8+ T cells of the elderly display a high expression of CD244, the T-cell differentiation marker of effector cells, suggesting the role of these lymphocytes in age-associated defective immune responses [147]. Chronic antigenic stimulation induced by persistent CMV infection drives a state of peripheral T-cell compartment exhaustion in older individuals, who exhibited an absolute increase in the effector memory CD4+ and CD8+ cells [148]. This increase occurs prevalently in the presence of elevated anti-CMV antibody titers. It is of interest that, although the number of CD8+ naïve T cells is lower in the blood, whether or not they are CMV-seropositive, only CMV-infected older adults have a lower number of naïve CD4+ T cells [148]. The accumulation of terminally differentiated, apoptosis-resistant, CMV-specific CD8+ lymphocytes is believed to reduce the overall TCR repertoire diversity [149,150]. This defect may impair the ability of older adults to respond to antigens they have never previously encountered. Accordingly, recent RNAseq data provide circumstantial evidence that aged subjects, and CMV-seropositive individuals in particular, exhibit blunted responses to neoantigens, partially due to reductions in the numbers of naïve T cells, resulting in a reduced TCR repertoire diversity [151].
In healthy donors >60 years of age, coinfection with CMV and Epstein–Barr virus (EBV) drives expansion of CD56- NK cells, characterized by reduced cytotoxic capacity and IFN-γ production [152]. Antigen-specific T cells against EBV undergo an age-related increase in the expression of markers associated with a differentiated phenotype, including KLRG-1, an increase in terminally differentiated T cells, and a decrease in the TCR repertoire diversity [153]. In infected patients, the expansion of viral-specific, exhausted, senescent CD8+CD28− T cells seems to play a central role in the onset of neoplastic lymphoproliferation, although the pathophysiology varies enormously among different disease entities [154]. However, information regarding the global burden of immunosenescence in lymphomagenesis is still scant, and requires a detailed analysis of different types of lymphoproliferation.
Long after hepatitis C clearance, predominantly male patients had increased plasma levels of SASP proteins, including IL-1α, IL-1RA, IL-8, IL-13, and IL-18. These changes have been associated with an increased risk of developing liver and non-hepatic diseases [155]. Infected patients likely produce an increased number of intrahepatic senescent, not functional T cells [156,157], which may promote the development of hepatocellular cancer as these T cells would be unable to eliminate premalignant senescent hepatocytes [158].
Varicella zoster virus (VZV) induces an expansion of NK cells displaying the terminally differentiated senescent marker CD57 [159], inhibiting their ability to secrete cytokines and to lyse virally infected target cells through NK cell-dependent cytotoxicity. In addition, the virus can interfere with the type 1 IFN pathway and the production of pro-inflammatory cytokines [160]. The incidence of herpes zoster, due to VZV infection, is also associated with an age-related decline in cell-mediated immunity against the virus, namely with the reduced frequency of virus-specific effector memory T cells [161], which appears to be the cause of the increased risk of post-herpetic neuralgia complications [162]. This is an important issue in view of the expected increase in population aging, leading to a higher incidence of herpes zoster cases in developed countries [163].
Other examples of the relationship between viruses and immunosenescence are measles, parvoviruses, and dengue viruses, as well as Merkel cell polyomavirus infections. Infection of human lung fibroblast cells by the measles virus generates cellular senescence, as documented by reduced cell proliferation, saβ-gal activity, increased expression of p53 and p21, as well as the induction of pro-inflammatory secretome with IL-8 or C-C motif chemokine ligand 5 expression [164]. Similarly, parvoviruses induce the expression of a pro-inflammatory secretome, including IL-8, IL-6, and IL-1β production [165]; dengue virus and Merkel cell polyomavirus elicit saβ-gal expression [166,167]. Whether these virus-induced senescence-mediated pathways is presently unknown.
Several signs of immunosenescence have been observed in the viral encephalitis caused by WNV [168], representing a worldwide health concern due to the prevalence, severity, and lack of efficient treatments for this disease. This virus is recognized by pathogen recognition receptors, the Toll-like receptors, which undergo an age-related decline [169]. Also, neutrophils, monocyte/macrophages, and DCs, the first barriers to this infection, show an age-related impairment [99]. In particular, an age-related up-regulation of AXL receptor tyrosine kinase, a molecule that, by regulating the blood–brain barrier permeability, facilitates viral uptake through phospholipid binding, has been reported. This receptor, potentially relevant for susceptibility to WNV, has been found in human DCs [170]. NK cells show age-related changes in phenotype and function [171].
Finally, it must be remembered that impairments of the immune system resembling those induced by aging can have an iatrogenic origin; this is the case, for example, of disease-modifying therapies for multiple sclerosis [172]. CD4+ T-cell lymphocytopenia, particularly within the central nervous system, low production of TRECs and KRECs, and T-cell repertoire restrictions increase the susceptibility of patients to infections, as reported for patients treated with natalizumab, who suffer of an earlier onset of progressive multifocal leukoencephalopathy, a rare but potentially fatal opportunistic infection caused by the JC virus [172,173,174].

3.2. Bacterial Infections

Several bacterial infections have been described as associated with the development of immunosenescence. This is the case with Mycobacterium tuberculosis (MTB). Although most individuals exposed to MTB manage to control tuberculosis (TB), which can remain in a latent form, approximately 5–10% of them develop an active disease [175]. Older adults are particularly susceptible to recurrences of dormant infections due to delayed diagnosis, co-morbidity, increased institutionalization, and immunosenescence [176]. Indeed, as with many other aging-related diseases, the diagnosis of active TB is difficult because of non-specific symptoms, such as unexplained fever and weight loss, which are less pronounced in this population [177]. In addition, age-related immunosenescence may reduce the overall sensitivity of traditional tuberculin skin tests and IFN-γ-releasing assays, which are recommended by the World Health Organization; this gap may be bypassed by the use of newer generation IFN-γ-releasing assays that show better performance in detecting TB infection among older adults [178,179,180]. Comorbidities, such as diabetes, could cause a ≥1.5-fold increased risk of developing TB [176]. The interactions between MTB and the host immune system are complex and only partially clarified, but the concomitant overall senescence of both innate and adaptive immunity appears to play a major role [181,182,183]. Changes in pulmonary IL-2 and TNF production may possibly impact granuloma formation and the maintenance of chronic infection. Similarly, an imbalanced pro- and anti-inflammatory factor pattern during aging may have a significant influence on the pulmonary macrophage functions relevant for the fusion of the phagosome with the lysosome, instrumental for eliminating MTB [184]. These data are in line with those obtained in mice, indicating that age induces an inflammatory pulmonary environment, turning the resident macrophages more prone to being infected by MTB [185]. Alterations in monocyte percentage and phenotype increase with age, suggesting a potential link with the elderly’s specific susceptibility to developing active TB [186]. Among older patients with TB, an age-associated over-representation of regulatory T cells, along with a significant reduction in the IFN-γ/IL-4 ratio, has also been reported, along with a significant reduction in other pro-inflammatory effector T-cell cytokines, such as IL-17A, IL-2, TNF-α, and polyfunctional (IFN-γ+ TNF-α+) T cells. This underscores the defective production of these effector cytokines, which may eventually lead to immunosuppression among older TB patients [187]. The studies demonstrating impaired adaptive T-cell immunity in old individuals with latent or active TB led to conflicting results, which do not allow us to identify immune age-associated changes as having a major pathogenic role [118,186]. Finally, it should be underlined that, despite the overall tuberculosis burden and rate of mortality in the elderly having declined in recent years [188], this pattern is expected to reverse, particularly for those aged ≥80 years [189]. Overall, due the low adherence to treatment and poorer tolerance to TB drugs, anticipatory diagnosis in the elderly represents an important challenge [176].
An additional leading cause of infection-related mortality in the elderly is Streptococcus pneumoniae infection [190]. In these patients, both antibody opsonic activities for all tested pneumococcal serotypes and phagocytic killing of pneumococci by neutrophils were significantly impaired [191]. The increased senescence markers, such as IL-1α/β, TNF-α, IL-6, and C-X-C motif chemokine ligand (CXCL)1, and cellular senescence facilitate bacterial adhesion to cells in the lungs [192], and may compromise upper respiratory mucosal immunity [193]. In addition, CD27+IgM+ B cells, which provide protection from Streptococcus pneumoniae and show age-related changes, may account for a reduced response to bacterial antigens [194].
Infections from Escherichia coli are the most common cause of recurring UTIs that continue to be a burden on aging females [195]. Older women are also more likely to be infected with the less common Gram-negative bacteria, such as Klebsiella pneumoniae, Proteus mirabilis, Enterobacter species, Citrobacter species, and Pseudomonas aeruginosa, as well as Gram-positive bacteria, including group B Streptococcus, other streptococci, Staphylococcus aureus, coagulase-negative staphylococci, and Enterococcus species [195,196]. The urinary bladder has physical barriers that prevent these infections, together with CD14+ monocyte-derived mononuclear phagocytes, and T cells [197]. The understanding of how immune senescence is related to UTI remains a challenge, although the data obtained in mice indicate a prevalent immune pathogenesis. It is of interest that, while T-cell responses can contribute to antigen-specific immunity in UTIs, bladder macrophages that in aged bladders begin to express CXCL13, may be responsible for inhibiting the development of adaptive immune responses [1,198]. In addition, macrophage phagocytic efficiency mediated via interaction with the NF-κB pathway during Pseudomonas aeruginosa infection has been found to be decreased [199]. The aged bladder produces high levels of the pro-inflammatory cytokines IL-6, IL-1β, and TNF-α [195], and is characterized by structures termed “tertiary lymphoid tissues” showing a germinal center with B-cell and T-cell zones and a follicular DC network. In addition, a redistribution of the B-cell pool from the periphery to the mucosal surface that alters the mucosal landscape has been described [195]. Interestingly, abundant γδ+ T cells were identified in the bladder [200], and experiments in mice demonstrated that these cells rapidly produce IL-17 upon infection, which promotes bacterial clearance [201]. Overall, the median percentage of γδ+ T cells decreases with increasing age [202].
Also, the overactive bladder, common in old women, appears to be correlated with inflammaging because the levels of nerve growth factor, C-C Motif Chemokine Ligand 2, and CXCL1 chemokines in the urine increase with age and disease severity [195].
Poor intestinal mucosal immunity represents a major factor leading to higher mortality from infections in aging [203] because it favors the changes in the human intestinal microbiota associated with inflammatory bowel diseases, irritable bowel syndrome, and metabolic disorders [204]. The altered balance between Gram-positive and Gram-negative intestinal bacteria may lead to the activation of DCs within the lamina propria of the intestine. As gut biodiversity decreases, potential pro-inflammatory microbes accumulate [205]. This imbalance starts a cascade of events inducing the release of pro-inflammatory cytokines, mainly IL-6 and IL-17. This, along with a decreased secretion of mucus and α-defensins by intestinal epithelial cells, further favors the entry of pathogens into mucosal layers [206]. Age-related alteration of the microbiota also leads to a decrease in the production of short-chain fatty acids, which may promote inflammation and cell vulnerability [207]. Dysregulation of the gut microbiota, more precisely the decrease in diversity of the microbiome associated with aging, appears also play a role in Clostridium difficile colitis, the major cause of gastrointestinal infections worldwide, occurring in up to 80% of cases in adults aged 65 [208,209]. Interestingly, patients who successfully eradicated Helicobacter pylori, another infection related to changes in the microbiota, had significantly lower histological markers of inflammation [210,211]. Finally, alteration in gut immune function has been proposed as one of the causes of small intestinal bacterial overgrowth (which implies excessive presence of bacteria) above 105–106 organisms/mL in small bowel aspirate [212]. This disease is common in the elderly, and is associated with chronic diarrhea, malabsorption, weight loss, and secondary nutritional deficiencies [213].
The Gram-negative anaerobic bacterium Porphyromonas gingivalis, responsible for a higher prevalence of periodontitis in the elderly population, is another pathogen associated with elevated expression of senescent cellular markers in immune cells, such as DCs. Indeed, bacterial invasion leads to an increase in the secretion of inflammatory exosomes which, in turn, amplify immune senescence [214].
Severe infections in the elderly can evolve into sepsis, a life-threatening event caused by host response failure, resulting in multiorgan collapse [215]. The outcomes of sepsis are worst in older adults, with higher rates of mortality, organ dysfunction, cognitive deficiency, and permanent disabilities [216]. These features can only partly be explained by age-related comorbidities [217], which are related to multiple organ failure produced by an excessive inflammatory response [218]. Bacteria highly responsible for sepsis are the Gram-positive Streptococcus pneumoniae, Streptococcus progenies, Streptococcus agalactiae, and Staphylococcus aureus; and the Gram-negative Neisseria meningitides, enteric (Escherichia coli, Klebsiella, Proteus, Enterobacter, Serratia, Citrobacter, and Salmonella), and non-enteric (Pseudomonas aeruginosa and Acinetobacter) [219].
The occurrence of sepsis arising from a concurrent excess of inflammation and immunosuppression [220] may also induce solid organ impairment [221], as in the case of blood–brain barrier disruption, local myocardial ischemia, or infarction secondary to preexisting coronaropathy. Liver damage appears in the early stages of sepsis in cirrhosis patients, in whom myeloid-derived suppressor cells are expanded. These cells, which inhibit the functions of DCs and macrophages [222], at the same time as reducing the diversity of NK cells, inhibit the Th1 response and induce Th2 and regulatory T-cell production [223]. Th2 cells have been shown to increase with aging [224], playing a paradoxical role in sepsis, since they may also increase the production of pro-inflammatory cytokines during emergency myelogenesis, and concomitantly be potently immunosuppressive [225]. Conclusively, our understanding of the relationship between aging and the onset of sepsis is incomplete and conflictual, as little or no association between age and inflammatory markers [226,227] or higher levels of these mediators among older septic patients [228] have been found.

3.3. Parasitic Infections

How age affects immune responses to lifelong parasitic infections is presently poorly understood. A supervised statistical learning technique indicated that older mice harbor a higher parasitic load than younger ones, due to the aging of adaptive immunity, characterized by reduced numbers of naïve T cells, poor T-cell responsiveness, and impaired antibody production [229]. In human cutaneous leishmaniasis, senescent CD56+CD57+ NK cells, together with CD4+ and effector memory CD8+ cells that re-express the CD45RA marker, play a role in the establishment and maintenance of tissue inflammation and are linked to lesion size [230,231]. RNA-seq data demonstrated that the cutaneous lesions exhibited a strong transcriptional co-induction of senescence-associated genes, and that the pro-inflammatory immune response is more strongly associated with the induction of senescent effector memory T cells re-expressing CD45RA [232]. In Chagas disease, a tropical disease representing a public health problem in developing countries, the infection by Trypanosoma cruzi causes excessive immune system stimulation that might elicit a progressive loss and collapse of immune functions. The induction of immune cells with senescent phenotypes may compromise the host’s capacity to control the magnitude of induced inflammation, predisposing infected hosts prematurely to immunosenescence [233], as demonstrated for CD4+ T cells. Indeed, in this infection, an increase in Ag-experienced IFN-γ-producing CD4+ T cells has been described [234]. Finally, there are data suggesting that the relationship among helminth parasite infection, immunity, and survival is not driven by genetics or early life environmental conditions, but rather by individual variations occurring late in life and, therefore, linked to immunosenescence. This was suggested mainly by a murine model performed in mice, in which Th2 function and anti-worm antibody production are compromised in old age [235].
Table 1 summarizes the principal age-related modifications of the innate and adaptive immune system occurring in age-related infectious diseases.

4. Immunosenescence and Vaccines

The interplay between immune senescence, inflammation, and infections highlights the importance of understanding age-related changes in the immune system and developing strategies to improve immune function in older individuals. The latter include the production of age-targeted vaccines. Vaccination that elicits immunological memory capable of protecting against subsequent infections is crucial in safeguarding the elderly, a population generally less responsive to both primary and booster challenges [134,236], and thus is less protected [237]. This was especially poignant in the wake of the COVID-19 pandemic, when the mortality rate was disproportionately high among the elderly [238]. The Advisory Committee on Immunization Practices of Centers for Disease Control and Prevention recommends that adults over 65 years obtain immunization against herpes zoster virus, Streptococcus pneumoniae, influenza viruses [104], and more recently, RSV in those over 60 years [239], and SARS-CoV-2 [104]. Moreover, tetanus and diphtheria vaccines were also recommended, despite responses to these vaccines often being impaired in older individuals [240].
The response to a vaccine is linked to several factors, such as the type, dose, and route of administration; the lack of, or incomplete, primary immunization; and the absence of regular boosters. Environmental factors, including exposure to pollutants and toxins, geographical location, seasons, the number of family members, composition of the microbiota, presence of co-infections, use of antibiotics, smoking, dietary intake, alcohol consumption, exercise, sleep patterns, and comorbidities of the host also play a role in vaccine response [241]. In addition, in older individuals, the response to vaccination is strongly influenced by immunosenescence and inflammaging. This last condition, through macrophage activation, creates a detrimental environment for the generation of a protective immune response to vaccinations, which is aggravated by the decreased antigen presentation capability of DCs to T cells [242]. Additionally, the production of cytokines is not optimal for adaptive immune response priming [243]. These responses in older individuals are also linked to the loss or decrease of the fine balance between the generation of inflammatory effector T cells and follicular helper T cells that mediate high-affinity antibody production, as well as the induction of long-lived memory cells for effective recall immunity [244]. In fact, during aging, this balance is reversed, and short-lived effector T-cell responses are prevalent over those of memory or follicular helper T cells [245]. Finally, vaccine-induced antibodies commonly show lower protective capacity [244], as suggested by the low production of antibodies in response to influenza vaccination in old rhesus macaques [246] and old individuals [247]. Similarly, IgA and IgM levels were found to be significantly lower in the elderly vaccinated against pneumonia [248,249,250], and antibodies against VZV are low in subjects ≥70 years old [251].
In view of the above, the currently available vaccines appear to provide short-term protection [241]. Therefore, new methodologies are needed to produce vaccines designed to optimally balance between immune stimulation and inflammatory status, and that are capable of inducing long-term immunological protection. These approaches include novel vaccine and adjuvant formulations, repeated heterologous booster injections, and alternative routes of administration [252]. New paradigms for the vaccine discovery and development process are based on “systems vaccinology,” which uses systems immunology technologies to probe the molecular networks that drive the immune response to vaccines. These advances, which incorporate immunobiography information with clinical, immunological and omics data, could allow stratifying subgroups of subjects and to identify those markers that could lead to the rational development of vaccines specifically designed for older adults [253,254]. The strengths of system vaccinology use are the possibility of exploiting results from previous studies on aging, in which the fixed variable is chronological age, with multiple longitudinal data, obtained from omics technologies, in which individuals are followed for a long time.

5. Limitations of Current Research on Immunosenescence, Future Research Directions and Potential Therapeutic Interventions

Although significant progress has been made in our understanding of immune features observed in the elderly in recent years, there are several common limitations in the studies regarding immunosenescence in both good health and diseases that should be considered. First, immune measures have usually been performed at a single point in time and, rarely, longitudinally. Therefore, there is still little information regarding the life-course trajectories of immune biomarkers or the differential rate of immunosenescence. Second, immunosenescence studies often lack appropriate controls. Third, characterizing cell phenotype in large-scale cohort studies is logistically and economically challenging, so the number of old subjects analyzed in different studies is usually limited. Fourth, a variety of particularities can arise in the context of the laboratory diagnostics of elderly people [255], and, therefore, it is possible to introduce technical errors due to transportation, pre-analytical mishandling, cell loss, and time involved to process samples. In particular, there are cell markers that are sensitive to the conditions in which they are collected, temperature, delay in processing, preservation method, duration of storage, and number of freeze–thaw cycles [256]. Fifth, although extensively studied, the knowledge of the exact molecular mechanisms related to immunosenescence remains limited, and molecular biomarkers for senescence remain lacking [15]. Sixth, there may be non-technical interferences, such as the frequent presence of acute diseases [257], which can momentarily alter immune features of old people. Finally, most studies have not considered sociodemographic differences in the aging population, while recent studies have demonstrated that individuals in more disadvantaged social positions experience greater levels of immunosenescence [258].
This review may also have some limitations. Because the literature related to immunosenescence has increased significantly in recent years and is continually updated, some information may have been lost, as well as information written in languages other than English.
Despite these limitations, the biases linked to individual variability and the conflicting results existing in the literature, the targeted and effective use of the large amount of information available today is the basis for the development of strategies aimed at fighting immunosenescence. At present, no preventive strategies capable of stemming the senescence of the immune system have been found. This is primarily because it is not yet well understood whether the observed immune function decline is a cause or a consequence of the interaction between the various systems within the aging of the organism. However, the identification of specific markers of cellular senescence will be crucial in the development of senolytic drugs targeting immune components. Several senolytic clinical trials have been completed, are current, or are planned for the near future [259], which include those focusing on immunosenescence or inflammaging [260,261,262]. While the first-generation senolytics demonstrated moderate efficacy when used in clinical trials, next-generation senolytics (CAR-T cells, antibody drug conjugates or vaccines) targeting specific proteins selectively expressed during senescence and, based on new technical developments, can pave the way for preclinical research [263]. Currently, the preclinical development of these strategies is challenging and aimed at avoiding strong side effects; however, the expected results are commensurate with the patients’ hopes regarding the treatments. Furthermore, stratification of the elderly population based on multiple biomarkers identified with the multi-omic approaches of system immunology, and the application of these techniques in the context of the mitochondrial genome [264] could help to recognize those patients who would benefit more from specific treatments, thus allowing more personalized approaches.
To our knowledge, no specific studies have evaluated the costs and burden of disease related to immunosenescence. However, it is important to underline that the diagnosis and treatment of chronic diseases related to age place a significant burden on National Healthcare System budgets [265]. Therefore, all strategies that will help to reduce immunosenescence, including the development of senolytics and successful vaccinations, have the potential to save many lives and to substantially reduce health care costs.
While current research on immunosenescence has provided valuable insights into age-related changes in the immune system, addressing existing limitations and pursuing innovative research directions are essential for developing effective therapeutic interventions to promote healthy aging and improve immune function in older adults. Collaboration across disciplines and integration of cutting-edge technologies will be key to advancing our understanding of immunosenescence and translating research findings into clinical practice.

6. Conclusions

The aged population worldwide is progressively increasing. According to the United Nations, the proportion of people over 65 will almost double between 2019 and 2050; therefore, age-related changes in the immune system are of enormous interest in the scientific and healthcare fields.
A large body of information has been accumulated regarding the still ill-defined link between bacterial infections and immunosenescence [266], indicating that aging affects the functions of immune system cells, resulting in an increase in infection severity, and that chronic viral infections can induce the senescence of immune cells.
Furthermore, it must be remembered that some younger individuals show features consistent with a pro-inflammatory and early immunosenescence state, which may predispose to an increased risk/severity of diseases [267]. On the contrary, there are elderly people, in particular centenarians, who harbor a unique, highly functional immune system that has successfully adapted to a history of insults, allowing for exceptional longevity [268]. These conditions can be related to the incapacity, at any age, to preserve and/or restore an optimal “immune resilience” when inflammatory or antigenic stressors occur [269]. Therefore, immune resilience would be a trait distinct from the processes that anchor immune status to chronological or biological age. Some individuals, especially males, may lack the ability to preserve optimal immune resilience when subjected to common inflammatory insults such as symptomatic viral infections.
Monitoring small populations of peripheral blood immune cells from clinical patient samples with new system immunology tools have enabled studies of cellular exhaustion leading to senescence directly in humans, especially during chronic infections. Despite this, to date, there are no precise data on the existence of common connections between immune senescence and specific infectious diseases. Mouse models have also been particularly informative in studies of aging [270], but they have not allowed us to establish to what extent these models faithfully recapitulate the mechanisms underlying immunosenescence processes in humans and their possible common effects in predisposing to infections. Similarly, clinical trials targeting aging in humans have shown promising but limited results [271]. In addition, all these studies did not consider potential changes in senescent cells residing in the target tissues of infections. A combination of multiple scientific approaches (e.g., multiparametric analyses, high-resolution omics technologies, systems biology and big data analytics) would enable a better understanding of common immune senescence features during human infections.
Although the full extent of the biological changes is largely unknown, several characteristic changes are typically and constantly observed in aging. Identifying hallmarks and characteristics associated with immunosenescence will be crucial for exploring their impact and significance, particularly in age-related infectious diseases.

Author Contributions

Conceptualization, E.Q.-R. and L.I.; writing—original draft preparation, L.I.; writing—review and editing, E.Q.-R., A.S., P.G.N. and L.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research received funding from 2nd bando Sanità; PNRR-Salute; Ministero della Salute, Italia.

Data Availability Statement

No new data were created.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wang, Y.; Dong, C.; Han, Y.; Gu, Z.; Sun, C. Immunosenescence, aging and successful aging. Front. Immunol. 2022, 13, 942796. [Google Scholar] [CrossRef] [PubMed]
  2. Rose, M.R. (Ed.) Evolutionary Biology of Aging. Oxford University Press: New York, NY, USA, 1991; ISBN 978-01-9509-530-2. [Google Scholar]
  3. Kirkwood, T.B.; Austad, S.N. Why do we age? Nature 2000, 408, 233–238. [Google Scholar] [CrossRef] [PubMed]
  4. Saborido, C.; García-Barranquero, P. Is aging a disease? The theoretical definition of aging in the light of the philosophy of medicine. J. Med. Philos. 2022, 47, 770–783. [Google Scholar] [CrossRef] [PubMed]
  5. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
  6. Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef] [PubMed]
  7. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef] [PubMed]
  8. Tuttle CS, L.; Waaijer ME, C.; Slee-Valentijn, M.S.; Stijnen, T.; Westendorp, R.; Maier, A.B. Cellular senescence and chronological age in various human tissues: A systematic review and meta-analysis. Aging Cell 2020, 19, e13083. [Google Scholar] [CrossRef] [PubMed]
  9. Xu, P.; Wang, M.; Song, W.M.; Wang, Q.; Yuan, G.C.; Sudmant, P.H.; Zare, H.; Tu, Z.; Orr, M.E.; Zhang, B. The landscape of human tissue and cell type specific expression and co-regulation of senescence genes. Mol. Neurodegener. 2022, 1, 5. [Google Scholar] [CrossRef]
  10. Low, E.; Alimohammadiha, G.; Smith, L.A.; Costello, L.F.; Przyborski, S.A.; von Zglinicki, T.; Miwa, S. How good is the evidence that cellular senescence causes skin ageing? Ageing Res. Rev. 2021, 71, 101456. [Google Scholar] [CrossRef]
  11. Liu, Y.; Zhang, Z.; Li, T.; Xu, H.; Zhang, H. Senescence in osteoarthritis: From mechanism to potential treatment. Arthritis Res. Ther. 2022, 24, 174. [Google Scholar] [CrossRef]
  12. Butcher, S.K.; Wang, K.; Lascelles, D.; Lord, J.M. Neutrophil ageing and immunosenescence. NeuroImmune Biol. 2004, 4, 41–55. [Google Scholar] [CrossRef]
  13. Aw, D.; Silva, A.B.; Palmer, D.B. Immunosenescence: Emerging challenges for an ageing population. Immunology 2007, 120, 435–446. [Google Scholar] [CrossRef] [PubMed]
  14. Bolton, C. An evaluation of the recognised systemic inflammatory biomarkers of chronic sub-optimal inflammation provides evidence for inflammageing (IFA) during multiple sclerosis (MS). Immun. Ageing 2021, 18, 18. [Google Scholar] [CrossRef] [PubMed]
  15. Liu, Z.; Liang, Q.; Ren, Y.; Guo, C.; Ge, X.; Wang, L.; Cheng, Q.; Luo, P.; Zhang, Y.; Han, X. Immunosenescence: Molecular mechanisms and diseases. Signal Transduct. Target. Ther. 2023, 8, 200. [Google Scholar] [CrossRef] [PubMed]
  16. Franceschi, C.; Bonafè, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef] [PubMed]
  17. Olivieri, F.; Marchegiani, F.; Matacchione, G.; Giuliani, A.; Ramini, D.; Fazioli, F.; Sabbatinelli, J.; Bonafè, M. Sex/gender-related differences in inflammaging. Mech. Ageing Dev. 2023, 211, 111792. [Google Scholar] [CrossRef] [PubMed]
  18. Sun, Y.; Coppé, J.P.; Lam, E.W. Cellular senescence: The sought or the unwanted? Trends Mol. Med. 2018, 24, 871–885. [Google Scholar] [CrossRef] [PubMed]
  19. Xia, S.; Zhang, X.; Zheng, S.; Khanabdali, R.; Kalionis, B.; Wu, J.; Wan, W.; Tai, X. An Update on inflamm-aging: Mechanisms, prevention, and treatment. J. Immunol. Res. 2016, 2016, 8426874. [Google Scholar] [CrossRef]
  20. Rossiello, F.; Jurk, D.; Passos, J.F.; d’Adda di Fagagna, F. Telomere dysfunction in ageing and age-related diseases. Nat. Cell Biol. 2022, 24, 135–147. [Google Scholar] [CrossRef] [PubMed]
  21. Fraile-Martinez, O.; De Leon-Oliva, D.; Boaru, D.L.; De Castro-Martinez, P.; Garcia-Montero, C.; Barrena-Blázquez, S.; García-García, J.; García-Honduvilla, N.; Alvarez-Mon, M.; Lopez-Gonzalez, L.; et al. Connecting epigenetics and inflammation in vascular senescence: State of the art, biomarkers and senotherapeutics. Front. Genet. 2024, 15, 1345459. [Google Scholar] [CrossRef] [PubMed]
  22. Arai, Y.; Martin-Ruiz, C.M.; Takayama, M.; Abe, Y.; Takebayashi, T.; Koyasu, S.; Suematsu, M.; Hirose, N.; von Zglinicki, T. Inflammation, but not telomere length, predicts successful ageing at extreme old age: A longitudinal study of semi-supercentenarians. EBioMedicine 2015, 2, 1549–1558. [Google Scholar] [CrossRef] [PubMed]
  23. Fulop, T.; Larbi, A.; Dupuis, G.; Le Page, A.; Frost, E.H.; Cohen, A.A.; Witkowski, J.M.; Franceschi, C. Immunosenescence and inflamm-aging as two sides of the same coin: Friends or foes? Front. Immunol. 2018, 8, 1960. [Google Scholar] [CrossRef] [PubMed]
  24. Santoro, A.; Bientinesi, E.; Monti, D. Immunosenescence and inflammaging in the aging process: Age-related diseases or longevity? Ageing Res. Rev. 2021, 71, 101422. [Google Scholar] [CrossRef] [PubMed]
  25. Fulop, T.; Larbi, A.; Pawelec, G.; Khalil, A.; Cohen, A.A.; Hirokawa, K.; Witkowski, J.M.; Franceschi, C. Immunology of aging: The birth of inflammaging. Clin. Rev. Allergy Immunol. 2023, 64, 109–122. [Google Scholar] [CrossRef] [PubMed]
  26. Bektas, A.; Schurman, S.H.; Sen, R.; Ferrucci, L. Human T cell immunosenescence and inflammation in aging. J. Leukoc. Biol. 2017, 102, 977–988. [Google Scholar] [CrossRef] [PubMed]
  27. Pawelec, G. Age and immunity: What is “immunosenescence”? Exp. Gerontol. 2018, 105, 4–9. [Google Scholar] [CrossRef]
  28. Chinn, I.K.; Blackburn, C.C.; Manley, N.R.; Sempowski, G.D. Changes in primary lymphoid organs with aging. Semin. Immunol. 2012, 24, 309–320. [Google Scholar] [CrossRef] [PubMed]
  29. Rodrigues, L.P.; Teixeira, V.R.; Alencar-Silva, T.; Simonassi-Paiva, B.; Pereira, R.W.; Pogue, R.; Carvalho, J.L. Hallmarks of aging and immunosenescence: Connecting the dots. Cytokine Growth Factor. Rev. 2021, 59, 9–21. [Google Scholar] [CrossRef] [PubMed]
  30. Chatta, G.S.; Andrews, R.G.; Rodger, E.; Schrag, M.; Hammond, W.P.; Dale, D.C. Hematopoietic progenitors and aging: Alterations in granulocytic precursors and responsiveness to recombinant human G-CSF, GM-CSF, and IL-3. J. Gerontol. 1993, 48, M207–M212. [Google Scholar] [CrossRef]
  31. Sabbatini, M.; Bona, E.; Novello, G.; Migliario, M.; Renò, F. Aging hampers neutrophil extracellular traps (NETs) efficacy. Aging Clin. Exp. Res. 2022, 34, 2345–2353. [Google Scholar] [CrossRef]
  32. van Beek, A.A.; Van den Bossche, J.; Mastroberardino, P.G.; de Winther, M.P.J.; Leenen, P.J.M. Metabolic alterations in aging Macrophages: Ingredients for inflammaging? Trends Immunol. 2019, 40, 113–127. [Google Scholar] [CrossRef] [PubMed]
  33. Taabazuing, C.Y.; Okondo, M.C.; Bachovchin, D.A. Pyroptosis and apoptosis pathways engage in bidirectional crosstalk in monocytes and macrophages. Cell Chem. Biol. 2017, 24, 507–514.e4. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, J.; Qiu, J.; Song, Y.; Liang, T.; Liu, S.; Ren, C.; Song, X.; Cui, L.; Sun, Y. Pyroptosis and degenerative diseases of the elderly. Cell Death Dis. 2023, 14, 94. [Google Scholar] [CrossRef] [PubMed]
  35. Guilliams, M.; Ginhoux, F.; Jakubzick, C.; Naik, S.H.; Onai, N.; Schraml, B.U.; Segura, E.; Tussiwand, R.; Yona, S. Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nat. Rev. Immunol. 2014, 14, 571–578. [Google Scholar] [CrossRef] [PubMed]
  36. Gounder, S.S.; Abdullah, B.J.J.; Radzuanb, N.E.I.B.M.; Zain, F.D.B.M.; Sait, N.B.M.; Chua, C.; Subramani, B. Effect of aging on NK cell population and their proliferation at ex vivo culture condition. Anal. Cell. Pathol. 2018, 2018, 7871814. [Google Scholar] [CrossRef] [PubMed]
  37. Douek, D.C.; McFarland, R.D.; Keiser, P.H.; Gage, E.A.; Massey, J.M.; Haynes, B.F.; Polis, M.A.; Haase, A.T.; Feinberg, M.B.; Sullivan, J.L.; et al. Changes in thymic function with age and during the treatment of HIV infection. Nature 1998, 396, 690–695. [Google Scholar] [CrossRef] [PubMed]
  38. Gulla, S.; Reddy, M.C.; Reddy, V.C.; Chitta, S.; Bhanoori, M.; Lomada, D. Role of thymus in health and disease. Int. Rev. Immunol. 2023, 42, 347–363. [Google Scholar] [CrossRef] [PubMed]
  39. Bains, I.; Yates, A.J.; Callard, R.E. Heterogeneity in thymic emigrants: Implications for thymectomy and immunosenescence. PLoS ONE 2013, 8, e49554. [Google Scholar] [CrossRef] [PubMed]
  40. Cunningham, C.A.; Helm, E.Y.; Fink, P.J. Reinterpreting recent thymic emigrant function: Defective or adaptive? Curr. Opin. Immunol. 2018, 51, 1–6. [Google Scholar] [CrossRef] [PubMed]
  41. Garcia-Prat, M.; Álvarez-Sierra, D.; Aguiló-Cucurull, A.; Salgado-Perandrés, S.; Briongos-Sebastian, S.; Franco-Jarava, C.; Martin-Nalda, A.; Colobran, R.; Montserrat, I.; Hernández-González, M.; et al. Extended immunophenotyping reference values in a healthy pediatric population. Cytom. B Clin. Cytom. 2019, 96, 223–233. [Google Scholar] [CrossRef] [PubMed]
  42. Steffens, C.M.; Al-Harthi, L.; Shott, S.; Yogev, R.; Landay, A. Evaluation of thymopoiesis using T cell receptor excision circles (TRECs): Differential correlation between adult and pediatric TRECs and naïve phenotypes. Clin. Immunol. 2000, 97, 95–101. [Google Scholar] [CrossRef]
  43. Sottini, A.; Serana, F.; Bertoli, D.; Chiarini, M.; Valotti, M.; Vaglio Tessitore, M.; Imberti, L. Simultaneous quantification of T-cell receptor excision circles (TRECs) and K-deleting recombination excision circles (KRECs) by real-time PCR. J. Vis. Exp. 2014, 94, 52184. [Google Scholar] [CrossRef]
  44. Adams, S.P.; Kricke, S.; Ralph, E.; Gilmour, N.; Gilmour, K.C. A comparison of TRECs and flow cytometry for naive T cell quantification. Clin. Exp. Immunol. 2018, 191, 198–202. [Google Scholar] [CrossRef]
  45. Weng, N.P. Numbers and odds: TCR repertoire size and its age changes impacting on T cell functions. Semin. Immunol. 2023, 69, 101810. [Google Scholar] [CrossRef]
  46. Qi, Q.; Liu, Y.; Cheng, Y.; Glanville, J.; Zhang, D.; Lee, J.Y.; Olshen, R.A.; Weyand, C.M.; Boyd, S.D.; Goronzy, J.J. Diversity and clonal selection in the human T-cell repertoire. Proc. Natl. Acad. Sci. USA 2014, 111, 13139–13144. [Google Scholar] [CrossRef] [PubMed]
  47. Derhovanessian, E.; Maier, A.B.; Beck, R.; Jahn, G.; Hähnel, K.; Slagboom, P.E.; de Craen, A.J.; Westendorp, R.G.; Pawelec, G. Hallmark features of immunosenescence are absent in familial longevity. J. Immunol. 2010, 185, 4618–4624. [Google Scholar] [CrossRef] [PubMed]
  48. Britanova, O.V.; Shugay, M.; Merzlyak, E.M.; Staroverov, D.B.; Putintseva, E.V.; Turchaninova, M.A.; Mamedov, I.Z.; Pogorelyy, M.V.; Bolotin, D.A.; Izraelson, M.; et al. Dynamics of individual T cell repertoires: From cord blood to centenarians. J. Immunol. 2016, 196, 5005–5013. [Google Scholar] [CrossRef] [PubMed]
  49. Tessitore, M.V.; Sottini, A.; Roccaro, A.M.; Ghidini, C.; Bernardi, S.; Martellosio, G.; Serana, F.; Imberti, L. Detection of newly produced T and B lymphocytes by digital PCR in blood stored dry on nylon flocked swabs. J. Transl. Med. 2017, 15, 70. [Google Scholar] [CrossRef] [PubMed]
  50. Frasca, D.; Landin, A.M.; Lechner, S.C.; Ryan, J.G.; Schwartz, R.; Riley, R.L.; Blomberg, B.B. Aging down-regulates the transcription factor E2A, activation-induced cytidine deaminase, and Ig class switch in human B cells. J. Immunol. 2008, 180, 5283–5290. [Google Scholar] [CrossRef]
  51. Dowery, R.; Benhamou, D.; Benchetrit, E.; Harel, O.; Nevelsky, A.; Zisman-Rozen, S.; Braun-Moscovici, Y.; Balbir-Gurman, A.; Avivi, I.; Shechter, A.; et al. Peripheral B cells repress B-cell regeneration in aging through a TNF-α/IGFBP-1/IGF-1 immune-endocrine axis. Blood 2021, 138, 1817–1829. [Google Scholar] [CrossRef]
  52. Ma, S.; Wang, C.; Mao, X.; Hao, Y.B. Cell dysfunction associated with aging and autoimmune diseases. Front. Immunol. 2019, 10, 318. [Google Scholar] [CrossRef]
  53. de Mol, J.; Kuiper, J.; Tsiantoulas, D.; Foks, A.C. The dynamics of B cell aging in health and disease. Front. Immunol. 2021, 12, 733566. [Google Scholar] [CrossRef]
  54. Fulop, T.; Witkowski, J.M.; Le Page, A.; Fortin, C.; Pawelec, G.; Larbi, A. Intracellular signalling pathways: Targets to reverse immunosenescence. Clin. Exp. Immunol. 2017, 187, 35–43. [Google Scholar] [CrossRef]
  55. Murakami, G.; Taniguchi, I. Histologic heterogeneity and intranodal shunt flow in lymph nodes from elderly subjects: A cadaveric study. Ann. Surg. Oncol. 2004, 11, 279S–284S. [Google Scholar] [CrossRef]
  56. Erofeeva, L.M.; Mnikhovich, M.V. Structural and functional changes in the mesenteric lymph nodes in humans during aging. Bull. Exp. Biol. Med. 2020, 168, 694–698. [Google Scholar] [CrossRef]
  57. Cakala-Jakimowicz, M.; Kolodziej-Wojnar, P.; Puzianowska-Kuznicka, M. Aging-related cellular, structural and functional changes in the lymph nodes: A significant component of immunosenescence? An Overview. Cells 2021, 10, 3148. [Google Scholar] [CrossRef]
  58. Denton, A.; Silva-Cayetano, A.; Dooley, J.; Hill, D.L.; Carr, E.J.; Robert, P.; Meyer-Hermann, M.; Liston, A.; Linterman, M.A. Intrinsic defects in lymph node stromal cells underpin poor germinal center responses during aging. bioRxiv. 2020. [Google Scholar] [CrossRef]
  59. Bronte, V.; Pittet, M.J. The spleen in local and systemic regulation of immunity. Immunity 2013, 39, 806–818. [Google Scholar] [CrossRef]
  60. Turner, V.M.; Mabbott, N.A. Influence of ageing on the microarchitecture of the spleen and lymph nodes. Biogerontology 2017, 18, 723–738. [Google Scholar] [CrossRef]
  61. Budamagunta, V.; Foster, T.C.; Zhou, D. Cellular senescence in lymphoid organs and immunosenescence. Aging 2021, 13, 19920–19941. [Google Scholar] [CrossRef]
  62. Sato, S.; Kiyono, H.; Fujihashi, K. Mucosal Immunosenescence in the gastrointestinal tract: A Mini-Review. Gerontology 2015, 61, 336–342. [Google Scholar] [CrossRef]
  63. Márquez, E.J.; Chung, C.H.; Marches, R.; Rossi, R.J.; Nehar-Belaid, D.; Eroglu, A.; Mellert, D.J.; Kuchel, G.A.; Banchereau, J.; Ucar, D. Sexual-dimorphism in human immune system aging. Nat. Commun. 2020, 11, 751. [Google Scholar] [CrossRef]
  64. Bongen, E.; Lucian, H.; Khatri, A.; Fragiadakis, G.K.; Bjornson, Z.B.; Nolan, G.P.; Utz, P.J.; Khatri, P. Sex differences in the blood transcriptome identify robust changes in immune cell proportions with aging and influenza infection. Cell Rep. 2019, 29, 1961–1973. [Google Scholar] [CrossRef]
  65. Pietschmann, P.; Gollob, E.; Brosch, S.; Hahn, P.; Kudlacek, S.; Willheim, M.; Woloszczuk, W.; Peterlik, M.; Tragl, K.H. The effect of age and gender on cytokine production by human peripheral blood mononuclear cells and markers of bone metabolism. Exp. Gerontol. 2003, 38, 1119–1127. [Google Scholar] [CrossRef]
  66. Sakiani, S.; Olsen, N.J.; Kovacs, W.J. Gonadal steroids and humoral immunity. Nat. Rev. Endocrinol. 2013, 9, 56–62. [Google Scholar] [CrossRef]
  67. Keestra, S.M.; Male, V.; Salali, G.D. Out of balance: The role of evolutionary mismatches in the sex disparity in autoimmune disease. Med. Hypotheses 2021, 151, 110558. [Google Scholar] [CrossRef]
  68. Aiello, A.; Farzaneh, F.; Candore, G.; Caruso, C.; Davinelli, S.; Gambino, C.M.; Ligotti, M.E.; Zareian, N.; Accardi, G. Immunosenescence and its hallmarks: How to oppose aging strategically? A review of potential options for therapeutic intervention. Front. Immunol. 2019, 10, 2247. [Google Scholar] [CrossRef]
  69. Hirokawa, K.; Utsuyama, M.; Hayashi, Y.; Kitagawa, M.; Makinodan, T.; Fulop, T. Slower immune system aging in women versus men in the Japanese population. Immun. Ageing 2013, 10, 19. [Google Scholar] [CrossRef]
  70. Dudkowska, M.; Janiszewska, D.; Karpa, A.; Broczek, K.; Dabrowski, M.; Sikora, E. The role of gender and labour status in immunosenescence of 65+ Polish population. Biogerontology 2017, 18, 581–590. [Google Scholar] [CrossRef]
  71. Piasecka, B.; Duffy, D.; Urrutia, A.; Quach, H.; Patin, E.; Posseme, C.; Bergstedt, J.; Charbit, B.; Rouilly, V.; MacPherson, C.R.; et al. Distinctive roles of age, sex, and genetics in shaping transcriptional variation of human immune responses to microbial challenges. Proc. Natl. Acad. Sci. USA 2018, 115, E488–E497. [Google Scholar] [CrossRef]
  72. Qu, K.; Zaba, L.C.; Giresi, P.G.; Li, R.; Longmire, M.; Kim, Y.H.; Greenleaf, W.J.; Chang, H.Y. Individuality and variation of personal regulomes in primary human T cells. Cell Syst. 2015, 1, 51–61. [Google Scholar] [CrossRef]
  73. Klein, S.L.; Morgan, R. The impact of sex and gender on immunotherapy outcomes. Biol. Sex Differ. 2020, 11, 24. [Google Scholar] [CrossRef]
  74. Brodin, P.; Jojic, V.; Gao, T.; Bhattacharya, S.; Angel, C.J.; Furman, D.; Shen-Orr, S.; Dekker, C.L.; Swan, G.E.; Butte, A.J.; et al. Variation in the human immune system is largely driven by non-heritable influences. Cell 2015, 160, 37–47. [Google Scholar] [CrossRef]
  75. Brodin, P. Systems-level patterns emerge. Nat. Rev. Immunol. 2019, 19, 87–88. [Google Scholar] [CrossRef]
  76. Olin, A.; Henckel, E.; Chen, Y.; Lakshmikanth, T.; Pou, C.; Mikes, J.; Gustafsson, A.; Bernhardsson, A.K.; Zhang, C.; Bohlin, K.; et al. Stereotypic immune system development in newborn children. Cell 2018, 174, 1277–1292.e14. [Google Scholar] [CrossRef]
  77. Alpert, A.; Pickman, Y.; Leipold, M.; Rosenberg-Hasson, Y.; Ji, X.; Gaujoux, R.; Rabani, H.; Starosvetsky, E.; Kveler, K.; Schaffert, S.; et al. A clinically meaningful metric of immune age derived from high-dimensional longitudinal monitoring. Nat. Med. 2019, 25, 487–495. [Google Scholar] [CrossRef]
  78. Yan, Z.; Maecker, H.T.; Brodin, P.; Nygaard, U.C.; Lyu, S.C.; Davis, M.M.; Nadeau, K.C.; Andorf, S. Aging and CMV discordance are associated with increased immune diversity between monozygotic twins. Immun. Ageing 2021, 18, 5. [Google Scholar] [CrossRef]
  79. Lakshmikanth, T.; Muhammad, S.A.; Olin, A.; Chen, Y.; Mikes, J.; Fagerberg, L.; Gummesson, A.; Bergström, G.; Uhlen, M.; Brodin, P. Human immune system variation during 1 Year. Cell Rep. 2020, 32, 107923. [Google Scholar] [CrossRef]
  80. Tran Van Hoi, E.; De Glas, N.A.; Portielje, J.E.A.; Van Heemst, D.; Van Den Bos, F.; Jochems, S.P.; Mooijaart, S.P. Biomarkers of the ageing immune system and their association with frailty—A systematic review. Exp. Gerontol. 2023, 176, 112163. [Google Scholar] [CrossRef]
  81. Alberro, A.; Iribarren-Lopez, A.; Sáenz-Cuesta, M.; Matheu, A.; Vergara, I.; Otaegui, D. Inflammaging markers characteristic of advanced age show similar levels with frailty and dependency. Sci. Rep. 2021, 11, 4358. [Google Scholar] [CrossRef]
  82. Xu, W.; Wong, G.; Hwang, Y.Y.; Larbi, A. The untwining of immunosenescence and aging. Semin. Immunopathol. 2020, 42, 559–572. [Google Scholar] [CrossRef]
  83. Medoro, A.; Saso, L.; Scapagnini, G.; Davinelli, S. NRF2 signaling pathway and telomere length in aging and age-related diseases. Mol. Cell. Biochem. 2023. [Google Scholar] [CrossRef]
  84. Vaiserman, A.; Krasnienkov, D. Telomere length as a marker of biological age: State-of-the-art, open issues, and future perspectives. Front. Genet. 2021, 11, 630186. [Google Scholar] [CrossRef]
  85. Zhang, J.; He, T.; Xue, L.; Guo, H. Senescent T cells: A potential biomarker and target for cancer therapy. EBioMedicine 2021, 68, 103409. [Google Scholar] [CrossRef]
  86. Verma, K.; Ogonek, J.; Varanasi, P.R.; Luther, S.; Bünting, I.; Thomay, K.; Behrens, Y.L.; Mischak-Weissinger, E.; Hambach, L. Human CD8+ CD57- TEMRA cells: Too young to be called “old”. PLoS ONE 2017, 12, e0177405. [Google Scholar] [CrossRef]
  87. Miron, M.; Meng, W.; Rosenfeld, A.M.; Dvorkin, S.; Poon, M.M.L.; Lam, N.; Kumar, B.V.; Louzoun, Y.; Luning Prak, E.T.; Farber, D.L. Maintenance of the human memory T cell repertoire by subset and tissue site. Genome Med. 2021, 13, 100. [Google Scholar] [CrossRef]
  88. Guo, L.; Liu, X.; Su, X. The role of TEMRA cell-mediated immune senescence in the development and treatment of HIV disease. Front. Immunol. 2023, 14, 1284293. [Google Scholar] [CrossRef]
  89. Yeh, Y.W.; Xiang, Z. Mouse hygiene status-A tale of two environments for mast cells and allergy. Allergol. Int. 2023, 73, 58–64. [Google Scholar] [CrossRef]
  90. Beura, L.K.; Hamilton, S.E.; Bi, K.; Schenkel, J.M.; Odumade, O.A.; Casey, K.A.; Thompson, E.A.; Fraser, K.A.; Rosato, P.C.; Filali-Mouhim, A.; et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature 2016, 532, 512–516. [Google Scholar] [CrossRef]
  91. Forlin, R.; James, A.; Brodin, P. Making human immune systems more interpretable through systems immunology. Trends Immunol. 2023, 44, 577–584. [Google Scholar] [CrossRef]
  92. Narang, V.; Decraene, J.; Wong, S.Y.; Aiswarya, B.S.; Wasem, A.R.; Leong, S.R.; Gouaillard, A. Systems immunology: A survey of modeling formalisms, applications and simulation tools. Immunol. Res. 2012, 53, 251–265. [Google Scholar] [CrossRef]
  93. Pulendran, B.; Davis, M.M. The science and medicine of human immunology. Science 2020, 369, eaay4014. [Google Scholar] [CrossRef]
  94. Davis, M.M.; Tato, C.M.; Furman, D. Systems immunology: Just getting started. Nat. Immunol. 2017, 18, 725–732. [Google Scholar] [CrossRef]
  95. Ginhoux, F.; Yalin, A.; Dutertre, C.A.; Amit, I. Single-cell immunology: Past, present, and future. Immunity 2022, 55, 393–404. [Google Scholar] [CrossRef]
  96. Poon, M.M.L.; Farber, D.L. The whole body as the system in systems immunology. iScience 2020, 23, 101509. [Google Scholar] [CrossRef]
  97. Tebani, A.; Gummesson, A.; Zhong, W.; Koistinen, I.S.; Lakshmikanth, T.; Olsson, L.M.; Boulund, F.; Neiman, M.; Stenlund, H.; Hellström, C.; et al. Integration of molecular profiles in a longitudinal wellness profiling cohort. Nat. Commun. 2020, 11, 4487. [Google Scholar] [CrossRef]
  98. Centers for Disease Control and Prevention. QuickStats: Death Rates* from Influenza and Pneumonia† Among Persons Aged ≥65 Years, by Sex and Age Group—National Vital Statistics System, United States. 2018. Available online: https://www.cdc.gov/mmwr/volumes/69/wr/mm6940a5.htm#:~:text=In%202018%2C%20the%20death%20rate,those%20aged%20%E2%89%A585%20years (accessed on 18 March 2024).
  99. Montgomery, R.R. Age-related alterations in immune responses to West Nile virus infection. Clin. Exp. Immunol. 2017, 187, 26–34. [Google Scholar] [CrossRef]
  100. Bartleson, J.M.; Radenkovic, D.; Covarrubias, A.J.; Furman, D.; Winer, D.A.; Verdin, E. SARS-CoV-2, COVID-19 and the ageing immune system. Nat. Aging 2021, 1, 769–782. [Google Scholar] [CrossRef]
  101. Cho, S.J.; Stout-Delgado, H.W. Aging and lung disease. Annu. Rev. Physiol. 2020, 82, 433–459. [Google Scholar] [CrossRef]
  102. Gomez, C.R.; Boehmer, E.D.; Kovacs, E.J. The aging innate immune system. Curr. Opin. Immunol. 2005, 17, 457–462. [Google Scholar] [CrossRef]
  103. Shinjyo, N.; Kita, K. Infection and immunometabolism in the central nervous system: A possible mechanistic link between metabolic imbalance and dementia. Front. Cell. Neurosci. 2021, 15, 765217. [Google Scholar] [CrossRef]
  104. Al-Jabri, M.; Rosero, C.; Saade, E.A. Vaccine-preventable diseases in older adults. Infect. Dis. Clin. N. Am. 2023, 37, 103–121. [Google Scholar] [CrossRef]
  105. Kahan, S.M.; Wherry, E.J.; Zajac, A.J. T cell exhaustion during persistent viral infections. Virology 2015, 479–480, 180–193. [Google Scholar] [CrossRef]
  106. Marrella, V.; Facoetti, A.; Cassani, B. Cellular Senescence in immunity against infections. Int. J. Mol. Sci. 2022, 23, 11845. [Google Scholar] [CrossRef]
  107. Wang, A.S.; Steers, N.J.; Parab, A.R.; Gachon, F.; Sweet, M.J.; Mysorekar, I.U. Timing is everything: Impact of development, ageing and circadian rhythm on macrophage functions in urinary tract infections. Mucosal Immunol. 2022, 15, 1114–1126. [Google Scholar] [CrossRef]
  108. Reyes, A.; Ortiz, G.; Duarte, L.F.; Fernández, C.; Hernández-Armengol, R.; Palacios, P.A.; Prado, Y.; Andrade, C.A.; Rodriguez-Guilarte, L.; Kalergis, A.M.; et al. Contribution of viral and bacterial infections to senescence and immunosenescence. Front. Cell. Infect. Microbiol. 2023, 13, 1229098. [Google Scholar] [CrossRef]
  109. Batista, M.A.; Calvo-Fortes, F.; Silveira-Nunes, G.; Camatta, G.C.; Speziali, E.; Turroni, S.; Teixeira-Carvalho, A.; Martins-Filho, O.A.; Neretti, N.; Maioli, T.U.; et al. Inflammaging in endemic areas for infectious diseases. Front. Immunol. 2020, 11, 579972. [Google Scholar] [CrossRef]
  110. Saavedra, D.; Añé-Kourí, A.L.; Barzilai, N.; Caruso, C.; Cho, K.H.; Fontana, L.; Franceschi, C.; Frasca, D.; Ledón, N.; Niedernhofer, L.J.; et al. Aging and chronic inflammation: Highlights from a multidisciplinary workshop. Immun. Ageing 2023, 20, 25. [Google Scholar] [CrossRef]
  111. Strulik, H.; Grossmann, V. The economics of aging with infectious and chronic diseases. Econ. Hum. Biol. 2023, 52, 101319. [Google Scholar] [CrossRef]
  112. Rothan-Tondeur, M.; Gavazzi, G.; Piette, F.; Lejeune, B.; de Wazières, B. L’observatoire du risque infectieux en gériatrie. Neurol. Psychiatr. Gériatr. 2005, 5, 20–25. [Google Scholar] [CrossRef]
  113. Yoshikawa, T.T. Epidemiology and unique aspects of aging and infectious diseases. Clin. Infect. Dis. 2000, 30, 931–933. [Google Scholar] [CrossRef] [PubMed]
  114. Du, W.Y.; Yin, C.N.; Wang, H.T.; Li, Z.W.; Wang, W.J.; Xue, F.Z.; Zhao, L.; Cao, W.C.; Cheeloo EcoHealth Consortium (CLEC). Infectious diseases among elderly persons: Results from a population-based observational study in Shandong province, China, 2013–2017. J. Glob. Health. 2021, 11, 08010. [Google Scholar] [CrossRef] [PubMed]
  115. Bender, B.S.H.I.V. Aging as a model for immunosenescence. J. Gerontol. A. Biol. Sci. Med. Sci. 1997, 52, M261–M263. [Google Scholar] [CrossRef] [PubMed]
  116. Deeks, S.G. HIV infection, inflammation, immunosenescence, and aging. Annu. Rev. Med. 2011, 62, 141–155. [Google Scholar] [CrossRef] [PubMed]
  117. Appay, V.; Rowland-Jones, S.L. Premature ageing of the immune system: The cause of AIDS? Trends Immunol. 2002, 23, 580–855. [Google Scholar] [CrossRef]
  118. Grifoni, A.; Alonzi, T.; Alter, G.; Noonan, D.M.; Landay, A.L.; Albini, A.; Goletti, D. Impact of aging on immunity in the context of COVID-19, HIV, and tuberculosis. Front. Immunol. 2023, 14, 1146704. [Google Scholar] [CrossRef] [PubMed]
  119. Hatzakis, A.; Touloumi, G.; Karanicolas, R.; Karafoulidou, A.; Mandalaki, T.; Anastassopoulou, C.; Zhang, L.; Goedert, J.J.; Ho, D.D.; Kostrikis, L.G. Effect of recent thymic emigrants on progression of HIV-1 disease. Lancet 2000, 355, 599–604. [Google Scholar] [CrossRef] [PubMed]
  120. Zhang, L.; Lewin, S.R.; Markowitz, M.; Lin, H.H.; Skulsky, E.; Karanicolas, R.; He, Y.; Jin, X.; Tuttleton, S.; Vesanen, M.; et al. Measuring recent thymic emigrants in blood of normal and HIV-1-infected individuals before and after effective therapy. J. Exp. Med. 1999, 190, 725–732. [Google Scholar] [CrossRef] [PubMed]
  121. Quiros-Roldan, E.; Serana, F.; Chiarini, M.; Zanotti, C.; Sottini, A.; Gotti, D.; Torti, C.; Caimi, L.; Imberti, L. Effects of combined antiretroviral therapy on B- and T-cell release from production sites in long-term treated HIV-1+ patients. J. Transl. Med. 2012, 10, 94. [Google Scholar] [CrossRef] [PubMed]
  122. Elias Junior, E.; Gubert, V.T.; Bonin-Jacob, C.M.; Puga, M.A.M.; Gouveia, C.G.; Sichinel, A.H.; Tozetti, I.A. CD57 T cells associated with immunosenescence in adults living with HIV or AIDS. Immunology 2024, 171, 146–153. [Google Scholar] [CrossRef] [PubMed]
  123. Heather, J.M.; Best, K.; Oakes, T.; Gray, E.R.; Roe, J.K.; Thomas, N.; Friedman, N.; Noursadeghi, M.; Chain, B. Dynamic perturbations of the T-cell receptor repertoire in chronic HIV infection and following antiretroviral therapy. Front. Immunol. 2016, 6, 644. [Google Scholar] [CrossRef] [PubMed]
  124. Turner, C.T.; Brown, J.; Shaw, E.; Uddin, I.; Tsaliki, E.; Roe, J.K.; Pollara, G.; Sun, Y.; Heather, J.M.; Lipman, M.; et al. Persistent T cell repertoire perturbation and T cell activation in HIV after long term treatment. Front. Immunol. 2021, 12, 634489. [Google Scholar] [CrossRef] [PubMed]
  125. Soares, L.S.; Espíndola, M.S.; Zambuzi, F.A.; Galvão-Lima, L.J.; Cacemiro, M.C.; Soares, M.R.; Santana, B.A.; Calado, R.T.; Bollela, V.R.; Frantz, F.G. Immunosenescence in chronic HIV infected patients impairs essential functions of their natural killer cells. Int. Immunopharmacol. 2020, 84, 106568. [Google Scholar] [CrossRef] [PubMed]
  126. Deeks, S.G.; Verdin, E.; McCune, J.M. Immunosenescence and HIV. Curr. Opin. Immunol. 2012, 24, 501–506. [Google Scholar] [CrossRef] [PubMed]
  127. Tizazu, A.M.; Mengist, H.M.; Demeke, G. Aging, inflammaging and immunosenescence as risk factors of severe COVID-19. Immun. Ageing 2022, 19, 53. [Google Scholar] [CrossRef]
  128. Asghari, F.; Asghary, A.; Majidi Zolbanin, N.; Faraji, F.; Jafari, R. Immunosenescence and Inflammaging in COVID-19. Viral. Immunol. 2023, 36, 579–592. [Google Scholar] [CrossRef] [PubMed]
  129. OECD Data. Influenza Vaccination Rates. Available online: https://data.oecd.org/healthcare/influenza-vaccination-rates.htm. (accessed on 18 March 2024).
  130. Patel, M.M.; York, I.A.; Monto, A.S.; Thompson, M.G.; Fry, A.M. Immune-mediated attenuation of influenza illness after infection: Opportunities and challenges. Lancet Microbe 2021, 2, e715–e725. [Google Scholar] [CrossRef]
  131. Plowden, J.; Renshaw-Hoelscher, M.; Engleman, C.; Katz, J.; Sambhara, S. Innate immunity in aging: Impact on macrophage function. Aging Cell 2004, 3, 161–167. [Google Scholar] [CrossRef]
  132. Albright, J.M.; Dunn, R.C.; Shults, J.A.; Boe, D.M.; Afshar, M.; Kovacs, E.J. Advanced age alters monocyte and macrophage responses. Antioxid. Redox. Signal. 2016, 25, 805–815. [Google Scholar] [CrossRef]
  133. Hearps, A.C.; Martin, G.E.; Angelovich, T.A.; Cheng, W.J.; Maisa, A.; Landay, A.L.; Jaworowski, A.; Crowe, S.M. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell 2012, 11, 867–875. [Google Scholar] [CrossRef]
  134. McElhaney, J.E.; Verschoor, C.P.; Andrew, M.K.; Haynes, L.; Kuchel, G.A.; Pawelec, G. The immune response to influenza in older humans: Beyond immune senescence. Immun. Ageing. 2020, 17, 10. [Google Scholar] [CrossRef] [PubMed]
  135. Jiang, J.; Fisher, E.M.; Murasko, D.M. CD8 T cell responses to influenza virus infection in aged mice. Ageing Res. Rev. 2011, 10, 422–427. [Google Scholar] [CrossRef] [PubMed]
  136. Parks, O.B.; Eddens, T.; Sojati, J.; Lan, J.; Zhang, Y.; Oury, T.D.; Ramsey, M.; Erickson, J.J.; Byersdorfer, C.A.; Williams, J.V. Terminally exhausted CD8+ T cells contribute to age-dependent severity of respiratory virus infection. Immun. Ageing 2023, 20, 40. [Google Scholar] [CrossRef] [PubMed]
  137. van de Sandt, C.E.; Nguyen, T.H.O.; Gherardin, N.A.; Crawford, J.C.; Samir, J.; Minervina, A.A.; Pogorelyy, M.V.; Rizzetto, S.; Szeto, C.; Kaur, J.; et al. Newborn and child-like molecular signatures in older adults stem from TCR shifts across human lifespan. Nat. Immunol. 2023, 24, 1890–1907. [Google Scholar] [CrossRef] [PubMed]
  138. Falsey, A.R.; Walsh, E.E. Respiratory syncytial virus infection in elderly adults. Drugs Aging 2005, 22, 577–587. [Google Scholar] [CrossRef] [PubMed]
  139. Mlinaric-Galinovic, G.; Falsey, A.R.; Walsh, E.E. Respiratory syncytial virus infection in the elderly. Eur. J. Clin. Microbiol. Infect. Dis. 1996, 15, 777–781. [Google Scholar] [CrossRef] [PubMed]
  140. Savic, M.; Penders, Y.; Shi, T.; Branche, A.; Pirçon, J.Y. Respiratory syncytial virus disease burden in adults aged 60 years and older in high-income countries: A systematic literature review and meta-analysis. Influenza Other Respir. Viruses 2023, 17, e13031. [Google Scholar] [CrossRef] [PubMed]
  141. Looney, R.J.; Falsey, A.R.; Walsh, E.; Campbell, D. Effect of aging on cytokine production in response to respiratory syncytial virus infection. J. Infect. Dis. 2002, 185, 682–685. [Google Scholar] [CrossRef] [PubMed]
  142. Salaun, B.; De Smedt, J.; Vernhes, C.; Moureau, A.; Öner, D.; Bastian, A.R.; Janssens, M.; Balla-Jhagjhoorsingh, S.; Aerssens, J.; Lambert, C.; et al. T cells, more than antibodies, may prevent symptoms developing from respiratory syncytial virus infections in older adults. Front. Immunol. 2023, 14, 1260146. [Google Scholar] [CrossRef] [PubMed]
  143. Njue, A.; Nuabor, W.; Lyall, M.; Margulis, A.; Mauskopf, J.; Curcio, D.; Kurosky, S.; Gessner, B.D.; Begier, E. Systematic literature review of risk factors for poor outcomes among adults with respiratory syncytial virus infection in high-income countries. Open Forum Infect. Dis. 2023, 10, ofad513. [Google Scholar] [CrossRef] [PubMed]
  144. Watson, A.; Wilkinson, T.M.A. Respiratory viral infections in the elderly. Ther. Adv. Respir. Dis. 2021, 15, 1753466621995050. [Google Scholar] [CrossRef] [PubMed]
  145. Palacios-Pedrero, M.Á.; Osterhaus, A.D.M.E.; Becker, T.; Elbahesh, H.; Rimmelzwaan, G.F.; Saletti, G. Aging and options to halt declining immunity to virus infections. Front. Immunol. 2021, 12, 681449. [Google Scholar] [CrossRef] [PubMed]
  146. Campos, C.; Pera, A.; Sanchez-Correa, B.; Alonso, C.; Lopez-Fernandez, I.; Morgado, S.; Tarazona, R.; Solana, R. Effect of age and CMV on NK cell subpopulations. Exp. Gerontol. 2014, 54, 130–137. [Google Scholar] [CrossRef] [PubMed]
  147. Pita-Lopez, M.L.; Gayoso, I.; DelaRosa, O.; Casado, J.G.; Alonso, C.; Muñoz-Gomariz, E.; Tarazona, R.; Solana, R. Effect of ageing on CMV-specific CD8 T cells from CMV seropositive healthy donors. Immun. Ageing 2009, 6, 11. [Google Scholar] [CrossRef] [PubMed]
  148. Wertheimer, A.M.; Bennett, M.S.; Park, B.; Uhrlaub, J.L.; Martinez, C.; Pulko, V.; Currier, N.L.; Nikolich-Žugich, D.; Kaye, J.; Nikolich-Žugich, J. Aging and cytomegalovirus infection differentially and jointly affect distinct circulating T cell subsets in humans. J. Immunol. 2014, 192, 2143–2155. [Google Scholar] [CrossRef]
  149. Khan, N.; Shariff, N.; Cobbold, M.; Bruton, R.; Ainsworth, J.A.; Sinclair, A.J.; Nayak, L.; Moss, P.A. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J. Immunol. 2002, 169, 1984–1992. [Google Scholar] [CrossRef] [PubMed]
  150. Attaf, M.; Malik, A.; Severinsen, M.C.; Roider, J.; Ogongo, P.; Buus, S.; Ndung’u, T.; Leslie, A.; Kløverpris, H.N.; Matthews, P.C.; et al. Major TCR repertoire perturbation by immunodominant HLA-B*44:03-restricted CMV-specific T cells. Front. Immunol. 2018, 9, 2539. [Google Scholar] [CrossRef] [PubMed]
  151. Nicoli, F.; Clave, E.; Wanke, K.; von Braun, A.; Bondet, V.; Alanio, C.; Douay, C.; Baque, M.; Lependu, C.; Marconi, P.; et al. Primary immune responses are negatively impacted by persistent herpesvirus infections in older people: Results from an observational study on healthy subjects and a vaccination trial on subjects aged more than 70 years old. EBioMedicine 2022, 76, 103852. [Google Scholar] [CrossRef]
  152. Müller-Durovic, B.; Grählert, J.; Devine, O.P.; Akbar, A.N.; Hess, C. CD56-negative NK cells with impaired effector function expand in CMV and EBV co-infected healthy donors with age. Aging 2019, 11, 724–740. [Google Scholar] [CrossRef] [PubMed]
  153. Lanfermeijer, J.; de Greef, P.C.; Hendriks, M.; Vos, M.; van Beek, J.; Borghans, J.A.M.; van Baarle, D. Age and CMV-infection jointly affect the EBV-specific CD8+ T-cell repertoire. Front. Aging 2021, 2, 665637. [Google Scholar] [CrossRef] [PubMed]
  154. Mancuso, S.; Carlisi, M.; Santoro, M.; Napolitano, M.; Raso, S.; Siragusa, S. Immunosenescence and lymphomagenesis. Immun. Ageing 2018, 15, 22. [Google Scholar] [CrossRef] [PubMed]
  155. Martín-Escolano, R.; Vidal-Alcántara, E.J.; Crespo, J.; Ryan, P.; Real, L.M.; Lazo-Álvarez, J.I.; Cabezas-González, J.; Macías, J.; Arias-Loste, M.T.; Cuevas, G.; et al. Immunological and senescence biomarker profiles in patients after spontaneous clearance of hepatitis C virus: Gender implications for long-term health risk. Immun. Ageing 2023, 20, 62. [Google Scholar] [CrossRef] [PubMed]
  156. Voehringer, D.; Blaser, C.; Brawand, P.; Raulet, D.H.; Hanke, T.; Pircher, H. Viral infections induce abundant numbers of senescent CD8 T cells. J. Immunol. 2001, 167, 4838–4843. [Google Scholar] [CrossRef] [PubMed]
  157. Giannakoulis, V.G.; Dubovan, P.; Papoutsi, E.; Kataki, A.; Koskinas, J. Senescence in HBV-, HCV- and NAFLD- mediated hepatocellular carcinoma and senotherapeutics: Current evidence and future perspective. Cancers 2021, 13, 4732. [Google Scholar] [CrossRef]
  158. Kang, T.W.; Yevsa, T.; Woller, N.; Hoenicke, L.; Wuestefeld, T.; Dauch, D.; Hohmeyer, A.; Gereke, M.; Rudalska, R.; Potapova, A.; et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 2011, 479, 547–551. [Google Scholar] [CrossRef] [PubMed]
  159. Campbell, T.M.; McSharry, B.P.; Steain, M.; Ashhurst, T.M.; Slobedman, B.; Abendroth, A. Varicella zoster virus productively infects human natural killer cells and manipulates phenotype. PLoS Pathog. 2018, 14, e1006999. [Google Scholar] [CrossRef] [PubMed]
  160. Gerada, C.; Campbell, T.M.; Kennedy, J.J.; McSharry, B.P.; Steain, M.; Slobedman, B.; Abendroth, A. Manipulation of the innate immune response by varicella zoster virus. Front. Immunol. 2020, 11, 1. [Google Scholar] [CrossRef] [PubMed]
  161. Levin, M.J. Immune senescence and vaccines to prevent herpes zoster in older persons. Curr. Opin. Immunol. 2012, 24, 494–500. [Google Scholar] [CrossRef]
  162. Johnson, R.W. Herpes Zoster and Postherpetic Neuralgia: A review of the effects of vaccination. Aging Clin. Exp. Res. 2009, 21, 236–243. [Google Scholar] [CrossRef] [PubMed]
  163. Curran, D.; Doherty, T.M.; Lecrenier, N.; Breuer, T. Healthy ageing: Herpes zoster infection and the role of zoster vaccination. NPJ Vaccines 2023, 8, 184. [Google Scholar] [CrossRef]
  164. Seoane, R.; Vidal, S.; Bouzaher, Y.H.; El Motiam, A.; Rivas, C. The interaction of viruses with the cellular senescence response. Biology 2020, 9, 455. [Google Scholar] [CrossRef] [PubMed]
  165. Arvia, R.; Zakrzewska, K.; Giovannelli, L.; Ristori, S.; Frediani, E.; Del Rosso, M.; Mocali, A.; Stincarelli, M.A.; Laurenzana, A.; Fibbi, G.; et al. Parvovirus B19 induces cellular senescence in human dermal fibroblasts: Putative role in systemic sclerosis-associated fibrosis. Rheumatology 2022, 61, 3864–3874. [Google Scholar] [CrossRef] [PubMed]
  166. AbuBakar, S.; Shu, M.H.; Johari, J.; Wong, P.F. Senescence affects endothelial cells susceptibility to dengue virus infection. Int. J. Med. Sci. 2014, 11, 538–544. [Google Scholar] [CrossRef] [PubMed]
  167. Siebels, S.; Czech-Sioli, M.; Spohn, M.; Schmidt, C.; Theiss, J.; Indenbirken, D.; Günther, T.; Grundhoff, A.; Fischer, N. Merkel cell polyomavirus DNA replication induces senescence in human dermal fibroblasts in a Kap1/Trim28-dependent manner. mBio 2020, 11, e00142-20. [Google Scholar] [CrossRef] [PubMed]
  168. Centers for Disease Control and Prevention. West Nile Virus. Available online: https://www.cdc.gov/westnile/statsmaps/historic-data.html?CDC_AA_refVal=https%3A%2F%2Fwww.cdc.gov%2Fwestnile%2Fstatsmaps%2FcumMapsData.html (accessed on 18 March 2024).
  169. Shaw, A.C.; Goldstein, D.R.; Montgomery, R.R. Age-dependent dysregulation of innate immunity. Nat. Rev. Immunol. 2013, 13, 875–887. [Google Scholar] [CrossRef] [PubMed]
  170. Qian, F.; Wang, X.; Zhang, L.; Lin, A.; Zhao, H.; Fikrig, E.; Montgomery, R. Impaired interferon signaling in dendritic cells from older donors infected in vitro with West Nile virus. J. Infect. Dis. 2011, 203, 1415–1424. [Google Scholar] [CrossRef] [PubMed]
  171. Yao, Y.; Strauss-Albee, D.M.; Zhou, J.Q.; Malawista, A.; Garcia, M.N.; Murray, K.O.; Blish, C.A.; Montgomery, R.R. The natural killer cell response to West Nile virus in young and old individuals with or without a prior history of infection. PLoS ONE 2017, 12, e0172625. [Google Scholar] [CrossRef] [PubMed]
  172. Mills, E.A.; Mao-Draayer, Y. Aging and lymphocyte changes by immunomodulatory therapies impact PML risk in multiple sclerosis patients. Mult. Scler. 2018, 24, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
  173. Prosperini, L.; Scarpazza, C.; Imberti, L.; Cordioli, C.; De Rossi, N.; Capra, R. Age as a risk factor for early onset of natalizumab-related progressive multifocal leukoencephalopathy. J. Neurovirol. 2017, 23, 742–749. [Google Scholar] [CrossRef] [PubMed]
  174. Bertoli, D.; Sottini, A.; Capra, R.; Scarpazza, C.; Bresciani, R.; Notarangelo, L.D.; Imberti, L. Lack of specific T- and B-cell clonal expansions in multiple sclerosis patients with progressive multifocal leukoencephalopathy. Sci. Rep. 2019, 9, 16605. [Google Scholar] [CrossRef] [PubMed]
  175. Behr, M.A.; Edelstein, P.H.; Ramakrishnan, L. Revisiting the timetable of tuberculosis. BMJ 2018, 362, k2738. [Google Scholar] [CrossRef] [PubMed]
  176. Teo, A.K.J.; Morishita, F.; Islam, T.; Viney, K.; Ong, C.W.M.; Kato, S.; Kim, H.; Liu, Y.; Oh, K.H.; Yoshiyama, T.; et al. Tuberculosis in older adults: Challenges and best practices in the Western Pacific Region. Lancet Reg. Health West. Pac. 2023, 36, 100770. [Google Scholar] [CrossRef] [PubMed]
  177. Rajagopalan, S. Tuberculosis and aging: A global health problem. Clin. Infect. Dis. 2001, 33, 1034–1039. [Google Scholar] [CrossRef] [PubMed]
  178. Kamiya, H.; Ikushima, S.; Kondo, K.; Satake, K.; Inomata, M.; Moriya, A.; Ando, T. Diagnostic performance of interferon-gamma release assays in elderly populations in comparison with younger populations. J. Infect. Chemother. 2013, 19, 217–222. [Google Scholar] [CrossRef] [PubMed]
  179. Kwon, Y.S.; Kim, Y.H.; Jeon, K.; Jeong, B.H.; Ryu, Y.J.; Choi, J.C.; Kim, H.C.; Koh, W.J. Factors that predict negative results of QuantiFERON-TB Gold In-Tube Test in patients with culture-confirmed tuberculosis: A Multicenter Retrospective Cohort Study. PLoS ONE 2015, 10, e0129792. [Google Scholar] [CrossRef] [PubMed]
  180. Fukushima, K.; Kubo, T.; Akagi, K.; Miyashita, R.; Kondo, A.; Ehara, N.; Takazono, T.; Sakamoto, N.; Mukae, H. Clinical evaluation of QuantiFERON®-TB Gold Plus directly compared with QuantiFERON®-TB Gold In-Tube and T-Spot®. TB for active pulmonary tuberculosis in the elderly. J. Infect. Chemother. 2021, 27, 1716–1722. [Google Scholar] [CrossRef] [PubMed]
  181. Byng-Maddick, R.; Noursadeghi, M. Does tuberculosis threaten our ageing populations? BMC Infect. Dis. 2016, 16, 119. [Google Scholar] [CrossRef] [PubMed]
  182. Menon, S.; Rossi, R.; Nshimyumukiza, L.; Wusiman, A.; Zdraveska, N.; Eldin, M.S. Convergence of a diabetes mellitus, protein energy malnutrition, and TB epidemic: The neglected elderly population. BMC Infect. Dis. 2016, 16, 361. [Google Scholar] [CrossRef]
  183. Rajagopalan, S. Tuberculosis in Older Adults. Clin. Geriatr. Med. 2016, 32, 479–491. [Google Scholar] [CrossRef]
  184. Asokan, S. Immune issues in elderly with TB. Indian J. Tuberc. 2022, 69 (Suppl. S2), S241–S245. [Google Scholar] [CrossRef] [PubMed]
  185. Canan, C.H.; Gokhale, N.S.; Carruthers, B.; Lafuse, W.P.; Schlesinger, L.S.; Torrelles, J.B.; Turner, J. Characterization of lung inflammation and its impact on macrophage function in aging. J. Leukoc. Biol. 2014, 96, 473–480. [Google Scholar] [CrossRef] [PubMed]
  186. Ault, R.; Dwivedi, V.; Koivisto, E.; Nagy, J.; Miller, K.; Nagendran, K.; Chalana, I.; Pan, X.; Wang, S.H.; Turner, J. Altered monocyte phenotypes but not impaired peripheral T cell immunity may explain susceptibility of the elderly to develop tuberculosis. Exp. Gerontol. 2018, 111, 35–44. [Google Scholar] [CrossRef] [PubMed]
  187. Namdeo, M.; Kandel, R.; Thakur, P.K.; Mohan, A.; Dey, A.B.; Mitra, D.K. Old age-associated enrichment of peripheral T regulatory cells and altered redox status in pulmonary tuberculosis patients. Eur. J. Immunol. 2020, 50, 1195–1208. [Google Scholar] [CrossRef] [PubMed]
  188. Hase, I.; Toren, K.G.; Hirano, H.; Sakurai, K.; Horne, D.J.; Saito, T.; Narita, M. Pulmonary tuberculosis in older adults: Increased mortality related to tuberculosis within two months of treatment initiation. Drugs Aging 2021, 38, 807–815. [Google Scholar] [CrossRef] [PubMed]
  189. Jang, S.Y.; Kim, M.J.; Cheong, H.K.; Oh, I.H. Estimating disability-adjusted life years due to tuberculosis in Korea through to the year 2040. Int. J. Environ. Res. Public Health 2020, 17, 5960. [Google Scholar] [CrossRef] [PubMed]
  190. Chen, H.; Matsumoto, H.; Horita, N.; Hara, Y.; Kobayashi, N.; Kaneko, T. Prognostic factors for mortality in invasive pneumococcal disease in adult: A system review and meta-analysis. Sci. Rep. 2021, 11, 11865. [Google Scholar] [CrossRef] [PubMed]
  191. Simell, B.; Vuorela, A.; Ekström, N.; Palmu, A.; Reunanen, A.; Meri, S.; Käyhty, H.; Väkeväinen, M. Aging reduces the functionality of anti-pneumococcal antibodies and the killing of Streptococcus pneumoniae by neutrophil phagocytosis. Vaccine 2011, 29, 1929–1934. [Google Scholar] [CrossRef] [PubMed]
  192. Shivshankar, P.; Boyd, A.R.; Le Saux, C.J.; Yeh, I.T.; Orihuela, C.J. Cellular senescence increases expression of bacterial ligands in the lungs and is positively correlated with increased susceptibility to pneumococcal pneumonia. Aging Cell 2011, 10, 798–806. [Google Scholar] [CrossRef] [PubMed]
  193. Weight, C.M.; Jochems, S.P.; Adler, H.; Ferreira, D.M.; Brown, J.S.; Heyderman, R.S. Insights into the effects of mucosal epithelial and innate immune dysfunction in older people on host interactions with Streptococcus pneumoniae. Front. Cell. Infect. Microbiol. 2021, 11, 651474. [Google Scholar] [CrossRef] [PubMed]
  194. Dunn-Walters, D.K.; O’Hare, J.S. Older Human B Cells and Antibodies. In Handbook of Immunosenescence: Basic Understanding and Clinical Implications; Fulop, T., Franceschi, C., Hirokawa, K., Pawelec, G., Eds.; Springer Nature: Cham, Switzerland, 2018; pp. 785–819. [Google Scholar] [CrossRef]
  195. Ligon, M.M.; Joshi, C.S.; Fashemi, B.E.; Salazar, A.M.; Mysorekar, I.U. Effects of aging on urinary tract epithelial homeostasis and immunity. Dev. Biol. 2023, 493, 29–39. [Google Scholar] [CrossRef] [PubMed]
  196. Thomas-White, K.J.; Kliethermes, S.; Rickey, L.; Lukacz, E.S.; Richter, H.E.; Moalli, P.; Zimmern, P.; Norton, P.; Kusek, J.W.; Wolfe, A.J.; et al. Evaluation of the urinary microbiota of women with uncomplicated stress urinary incontinence. Am. J. Obstet. Gynecol. 2017, 216, 55.e1–55.e16. [Google Scholar] [CrossRef] [PubMed]
  197. Munoz, J.A.; Uhlemann, A.C.; Barasch, J. Innate bacteriostatic mechanisms defend the urinary tract. Annu. Rev. Physiol. 2022, 84, 533–558. [Google Scholar] [CrossRef] [PubMed]
  198. Mora-Bau, G.; Platt, A.M.; van Rooijen, N.; Randolph, G.J.; Albert, M.L.; Ingersoll, M.A. Macrophages subvert adaptive immunity to urinary tract infection. PLoS Pathog. 2015, 11, e1005044. [Google Scholar] [CrossRef] [PubMed]
  199. Li, H.; Luo, Y.F.; Wang, Y.S.; Yang, Q.; Xiao, Y.L.; Cai, H.R.; Xie, C.M. Using ROS as a second messenger, NADPH oxidase 2 mediates macrophage senescence via interaction with NF-κB during Pseudomonas aeruginosa infection. Oxid. Med. Cell. Longev. 2018, 2018, 9741838. [Google Scholar] [CrossRef] [PubMed]
  200. Matsukawa, M.; Kumamoto, Y.; Hirose, T.; Matsuura, A. Tissue gamma/delta T cells in experimental urinary tract infection relationship between other immuno-competent cells. Kansenshogaku Zasshi 1994, 68, 1498–1511. [Google Scholar] [CrossRef] [PubMed]
  201. Sivick, K.E.; Schaller, M.A.; Smith, S.N.; Mobley, H.L. The innate immune response to uropathogenic Escherichia coli involves IL-17A in a murine model of urinary tract infection. J. Immunol. 2010, 184, 2065–2075. [Google Scholar] [CrossRef] [PubMed]
  202. Singh, P.; Szaraz-Szeles, M.; Mezei, Z.; Barath, S.; Hevessy, Z. Age-dependent frequency of unconventional T cells in a healthy adult Caucasian population: A combinational study of invariant natural killer T cells, γδ T cells, and mucosa-associated invariant T cells. Geroscience 2022, 44, 2047–2060. [Google Scholar] [CrossRef] [PubMed]
  203. Schmucker, D.L.; Heyworth, M.F.; Owen, R.L.; Daniels, C.K. Impact of aging on gastrointestinal mucosal immunity. Dig. Dis. Sci. 1996, 41, 1183–1193. [Google Scholar] [CrossRef] [PubMed]
  204. Madhogaria, B.; Bhowmik, P.; Kundu, A. Correlation between human gut microbiome and diseases. Infect. Med. 2022, 1, 180–191. [Google Scholar] [CrossRef] [PubMed]
  205. Ren, J.; Li, H.; Zeng, G.; Pang, B.; Wang, Q.; Wei, J. Gut microbiome-mediated mechanisms in aging-related diseases: Are probiotics ready for prime time? Front. Pharmacol. 2023, 14, 1178596. [Google Scholar] [CrossRef] [PubMed]
  206. Dumic, I.; Nordin, T.; Jecmenica, M.; Stojkovic Lalosevic, M.; Milosavljevic, T.; Milovanovic, T. Gastrointestinal Tract Disorders in Older Age. Can. J. Gastroenterol. Hepatol. 2019, 2019, 6757524. [Google Scholar] [CrossRef] [PubMed]
  207. De Vuyst, L.; Leroy, F. Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production. Int. J. Food Microbiol. 2011, 149, 73–80. [Google Scholar] [CrossRef] [PubMed]
  208. Kelly, C.P.; LaMont, J.T. Clostridium difficile--more difficult than ever. N. Engl. J. Med. 2008, 359, 1932–1940. [Google Scholar] [CrossRef] [PubMed]
  209. Yoshikawa, T.T.; Norman, D.C. Geriatric infectious diseases: Current concepts on diagnosis and management. J. Am. Geriatr. Soc. 2017, 65, 631–641. [Google Scholar] [CrossRef] [PubMed]
  210. Kokkola, A.; Sipponen, P.; Rautelin, H.; Härkönen, M.; Kosunen, T.U.; Haapiainen, R.; Puolakkainen, P. The effect of Helicobacter pylori eradication on the natural course of atrophic gastritis with dysplasia. Aliment. Pharmacol. Ther. 2002, 16, 515–520. [Google Scholar] [CrossRef] [PubMed]
  211. Shin, C.M.; Kim, N.; Park, J.H.; Lee, D.H. Changes in gastric corpus microbiota with age and after Helicobacter pylori eradication: A long-term follow-up study. Front. Microbiol. 2021, 11, 621879. [Google Scholar] [CrossRef] [PubMed]
  212. Dukowicz, A.C.; Lacy, B.E.; Levine, G.M. Small intestinal bacterial overgrowth: A comprehensive review. Gastroenterol. Hepatol. 2007, 3, 112–122. [Google Scholar]
  213. Parlesak, A.; Klein, B.; Schecher, K.; Bode, J.C.; Bode, C. Prevalence of small bowel bacterial overgrowth and its association with nutrition intake in nonhospitalized older adults. J. Am. Geriatr. Soc. 2003, 51, 768–773. [Google Scholar] [CrossRef] [PubMed]
  214. Elsayed, R.; Elashiry, M.; Liu, Y.; El-Awady, A.; Hamrick, M.; Cutler, C.W. Porphyromonas gingivalis provokes exosome secretion and paracrine immune senescence in bystander dendritic cells. Front. Cell. Infect. Microbiol. 2021, 11, 669989. [Google Scholar] [CrossRef] [PubMed]
  215. Rudd, K.E.; Johnson, S.C.; Agesa, K.M.; Shackelford, K.A.; Tsoi, D.; Kievlan, D.R.; Colombara, D.V.; Ikuta, K.S.; Kissoon, N.; Finfer, S.; et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: Analysis for the Global Burden of Disease Study. Lancet 2020, 395, 200–211. [Google Scholar] [CrossRef] [PubMed]
  216. Rowe, T.A.; McKoy, J.M. Sepsis in older adults. Infect. Dis. Clin. N. Am. 2017, 31, 731–742. [Google Scholar] [CrossRef] [PubMed]
  217. Yang, Y.; Yang, K.S.; Hsann, Y.M.; Lim, V.; Ong, B.C. The effect of comorbidity and age on hospital mortality and length of stay in patients with sepsis. J. Crit. Care 2010, 25, 398–405. [Google Scholar] [CrossRef] [PubMed]
  218. Giamarellos-Bourboulis, E.J. Immune-mediated inflammatory diseases as long-term sepsis complications: Long-term persistence of host dysregulation? J. Intern. Med. 2024, 295, 123–125. [Google Scholar] [CrossRef] [PubMed]
  219. Alebachew, G.; Teka, B.; Endris, M.; Shiferaw, Y.; Tessema, B. Etiologic agents of bacterial sepsis and their antibiotic susceptibility patterns among patients living with Human Immunodeficiency Virus at Gondar University Teaching Hospital, Northwest Ethiopia. Biomed. Res. Int. 2016, 2016, 5371875. [Google Scholar] [CrossRef] [PubMed]
  220. Liu, D.; Huang, S.Y.; Sun, J.H.; Zhang, H.C.; Cai, Q.L.; Gao, C.; Li, L.; Cao, J.; Xu, F.; Zhou, Y.; et al. Sepsis-induced immunosuppression: Mechanisms, diagnosis and current treatment options. Mil. Med. Res. 2022, 9, 56. [Google Scholar] [CrossRef] [PubMed]
  221. Lu, X.; Lu, Y.Q. Editorial: Immunosenescence after sepsis. Front. Immunol. 2023, 14, 1177148. [Google Scholar] [CrossRef] [PubMed]
  222. Sehgal, R.; Maiwall, R.; Rajan, V.; Islam, M.; Baweja, S.; Kaur, N.; Kumar, G.; Ramakrishna, G.; Sarin, S.K.; Trehanpati, N. Granulocyte-Macrophage Colony-Stimulating Factor modulates myeloid-derived suppressor cells and Treg activity in decompensated cirrhotic patients with sepsis. Front. Immunol. 2022, 313, 828949. [Google Scholar] [CrossRef]
  223. Malavika, M.; Sanju, S.; Poorna, M.R.; Vishnu Priya, V.; Sidharthan, N.; Varma, P.; Mony, U. Role of myeloid derived suppressor cells in sepsis. Int. Immunopharmacol. 2022, 104, 108452. [Google Scholar] [CrossRef] [PubMed]
  224. Venet, F.; Monneret, G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat. Rev. Nephrol. 2018, 14, 121–137. [Google Scholar] [CrossRef] [PubMed]
  225. Cuenca, A.G.; Delano, M.J.; Kelly-Scumpia, K.M.; Moreno, C.; Scumpia, P.O.; Laface, D.M.; Heyworth, P.G.; Efron, P.A.; Moldawer, L.L. A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma. Mol. Med. 2011, 17, 281–292. [Google Scholar] [CrossRef] [PubMed]
  226. Marik, P.E.; Zaloga, G.P. NORASEPT II Study Investigators. North American Sepsis Trial II. The effect of aging on circulating levels of proinflammatory cytokines during septic shock. Norasept II Study Investigators. J. Am. Geriatr. Soc. 2001, 49, 5–9. [Google Scholar] [CrossRef] [PubMed]
  227. Pinheiro da Silva, F.; Zampieri, F.G.; Barbeiro, D.F.; Barbeiro, H.V.; Goulart, A.C.; Torggler Filho, F.; Velasco, I.T.; da Cruz Neto, L.M.; de Souza, H.P.; Machado, M.C. Septic shock in older people: A prospective cohort study. Immun. Ageing 2013, 10, 21. [Google Scholar] [CrossRef] [PubMed]
  228. Ginde, A.A.; Blatchford, P.J.; Trzeciak, S.; Hollander, J.E.; Birkhahn, R.; Otero, R.; Osborn, T.M.; Moretti, E.; Nguyen, H.B.; Gunnerson, K.J.; et al. Age-related differences in biomarkers of acute inflammation during hospitalization for sepsis. Shock 2014, 42, 99–107. [Google Scholar] [CrossRef] [PubMed]
  229. Babayan, S.A.; Sinclair, A.; Duprez, J.S.; Selman, C. Chronic helminth infection burden differentially affects haematopoietic cell development while ageing selectively impairs adaptive responses to infection. Sci. Rep. 2018, 8, 3802. [Google Scholar] [CrossRef] [PubMed]
  230. Covre, L.P.; Martins, R.F.; Devine, O.P.; Chambers, E.S.; Vukmanovic-Stejic, M.; Silva, J.A.; Dietze, R.; Rodrigues, R.R.; de Matos Guedes, H.L.; Falqueto, A.; et al. Circulating senescent T cells are linked to systemic inflammation and lesion size during human cutaneous leishmaniasis. Front. Immunol. 2019, 9, 3001. [Google Scholar] [CrossRef] [PubMed]
  231. Covre, L.P.; Devine, O.P.; Garcia de Moura, R.; Vukmanovic-Stejic, M.; Dietze, R.; Ribeiro-Rodrigues, R.; Guedes, H.L.M.; Lubiana Zanotti, R.; Falqueto, A.; Akbar, A.N.; et al. Compartmentalized cytotoxic immune response leads to distinct pathogenic roles of natural killer and senescent CD8+ T cells in human cutaneous leishmaniasis. Immunology 2020, 159, 429–440. [Google Scholar] [CrossRef] [PubMed]
  232. Fantecelle, C.H.; Covre, L.P.; Garcia de Moura, R.; Guedes, H.L.M.; Amorim, C.F.; Scott, P.; Mosser, D.; Falqueto, A.; Akbar, A.N.; Gomes, D.C.O. Transcriptomic landscape of skin lesions in cutaneous leishmaniasis reveals a strong CD8+ T cell immunosenescence signature linked to immunopathology. Immunology 2021, 164, 754–765. [Google Scholar] [CrossRef]
  233. Guimarães-Pinto, K.; Ferreira, J.R.M.; da Costa, A.L.A.; Morrot, A.; Freire-de-Lima, L.; Decote-Ricardo, D.; Freire-de-Lima, C.G.; Filardy, A.A. Cellular stress and senescence induction during Trypanosoma cruzi infection. Trop. Med. Infect. Dis. 2022, 7, 129. [Google Scholar] [CrossRef] [PubMed]
  234. Albareda, M.C.; Olivera, G.C.; Laucella, S.A.; Alvarez, M.G.; Fernandez, E.R.; Lococo, B.; Viotti, R.; Tarleton, R.L.; Postan, M. Chronic human infection with Trypanosoma cruzi drives CD4+ T cells to immune senescence. J. Immunol. 2009, 183, 4103–4108. [Google Scholar] [CrossRef] [PubMed]
  235. Froy, H.; Sparks, A.M.; Watt, K.; Sinclair, R.; Bach, F.; Pilkington, J.G.; Pemberton, J.M.; McNeilly, T.N.; Nussey, D.H. Senescence in immunity against helminth parasites predicts adult mortality in a wild mammal. Science 2019, 365, 1296–1298. [Google Scholar] [CrossRef] [PubMed]
  236. Fulop, T.; Larbi, A.; Pawelec, G.; Cohen, A.A.; Provost, G.; Khalil, A.; Lacombe, G.; Rodrigues, S.; Desroches, M.; Hirokawa, K.; et al. Immunosenescence and altered vaccine efficiency in older subjects: A myth difficult to change. Vaccines 2022, 10, 607. [Google Scholar] [CrossRef] [PubMed]
  237. Demicheli, V.; Jefferson, T.; Di Pietrantonj, C.; Ferroni, E.; Thorning, S.; Thomas, R.E.; Rivetti, A. Vaccines for preventing influenza in the elderly. Cochrane Database Syst. Rev. 2018, 2, CD004876. [Google Scholar] [CrossRef] [PubMed]
  238. Yang, X.; Yu, Y.; Xu, J.; Shu, H.; Xia, J.; Liu, H.; Wu, Y.; Zhang, L.; Yu, Z.; Fang, M.; et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: A single-centered, retrospective, observational study. Lancet Respir. Med. 2020, 8, 475–481. [Google Scholar] [CrossRef] [PubMed]
  239. Melgar, M.; Britton, A.; Roper, L.E.; Talbot, H.K.; Long, S.S.; Kotton, C.N.; Havers, F.P. Use of Respiratory Syncytial Virus Vaccines in Older Adults: Recommendations of the Advisory Committee on Immunization Practices—United States, 2023. MMWR Morb. Mortal. Wkly. Rep. 2023, 72, 793–801. [Google Scholar] [CrossRef] [PubMed]
  240. Weinberger, B.; Schirmer, M.; Matteucci Gothe, R.; Siebert, U.; Fuchs, D.; Grubeck-Loebenstein, B. Recall responses to tetanus and diphtheria vaccination are frequently insufficient in elderly persons. PLoS ONE 2013, 8, e82967. [Google Scholar] [CrossRef] [PubMed]
  241. Soegiarto, G.; Purnomosari, D. Challenges in the vaccination of the elderly and strategies for improvement. Pathophysiology 2023, 30, 155–173. [Google Scholar] [CrossRef] [PubMed]
  242. Murdaca, G.; Paladin, F.; Martino, G.; Gangemi, S. Impact of immunosenescence on viral Infections with an emphasis on COVID-19. Front. Biosci. 2023, 28, 225. [Google Scholar] [CrossRef] [PubMed]
  243. Crooke, S.N.; Ovsyannikova, I.G.; Poland, G.A.; Kennedy, R.B. Immunosenescence and human vaccine immune responses. Immun. Ageing 2019, 16, 25. [Google Scholar] [CrossRef] [PubMed]
  244. Gustafson, C.E.; Kim, C.; Weyand, C.M.; Goronzy, J.J. Influence of immune aging on vaccine responses. J. Allergy Clin. Immunol. 2020, 145, 1309–1321. [Google Scholar] [CrossRef] [PubMed]
  245. Linterman, M.A. Age-dependent changes in T follicular helper cells shape the humoral immune response to vaccination. Semin. Immunol. 2023, 69, 101801. [Google Scholar] [CrossRef] [PubMed]
  246. Coe, C.L.; Lubach, G.R.; Kinnard, J. Immune senescence in old and very old rhesus monkeys: Reduced antibody response to influenza vaccination. Age 2012, 34, 1169–1177. [Google Scholar] [CrossRef] [PubMed]
  247. Murasko, D.M.; Bernstein, E.D.; Gardner, E.M.; Gross, P.; Munk, G.; Dran, S.; Abrutyn, E. Role of humoral and cell-mediated immunity in protection from influenza disease after immunization of healthy elderly. Exp. Gerontol. 2002, 37, 427–439. [Google Scholar] [CrossRef] [PubMed]
  248. Ademokun, A.; Wu, Y.C.; Martin, V.; Mitra, R.; Sack, U.; Baxendale, H.; Kipling, D.; Dunn-Walters, D.K. Vaccination-induced changes in human B-cell repertoire and pneumococcal IgM and IgA antibody at different ages. Aging Cell 2011, 10, 922–930. [Google Scholar] [CrossRef] [PubMed]
  249. Schenkein, J.G.; Nahm, M.H.; Dransfield, M.T. Pneumococcal vaccination for patients with COPD: Current practice and future directions. Chest 2008, 133, 767–774. [Google Scholar] [CrossRef] [PubMed]
  250. Schenkein, J.G.; Park, S.; Nahm, M.H. Pneumococcal vaccination in older adults induces antibodies with low opsonic capacity and reduced antibody potency. Vaccine 2008, 26, 5521–5526. [Google Scholar] [CrossRef] [PubMed]
  251. Levin, M.J.; Oxman, M.N.; Zhang, J.H.; Johnson, G.R.; Stanley, H.; Hayward, A.R.; Caulfield, M.J.; Irwin, M.R.; Smith, J.G.; Clair, J.; et al. Varicella-zoster virus-specific immune responses in elderly recipients of a herpes zoster vaccine. J. Infect. Dis. 2008, 197, 825–835. [Google Scholar] [CrossRef] [PubMed]
  252. Arrazola Martínez, M.P.; Eiros Bouza, J.M.; Plans Rubió, P.; Puig-Barberà, J.; Ruiz Aragón, J.; Torres Lana, A.J. Efficacy, effectiveness and safety of the adjuvanted influenza vaccine in the population aged 65 or over. Rev. Esp. Quimioter. 2023, 36, 334–345. [Google Scholar] [CrossRef] [PubMed]
  253. Ciabattini, A.; Nardini, C.; Santoro, F.; Garagnani, P.; Franceschi, C.; Medaglini, D. Vaccination in the elderly: The challenge of immune changes with aging. Semin. Immunol. 2018, 40, 83–94. [Google Scholar] [CrossRef] [PubMed]
  254. Pulendran, B.; S Arunachalam, P.; O’Hagan, D.T. Emerging concepts in the science of vaccine adjuvants. Nat. Rev. Drug Discov. 2021, 20, 454–475. [Google Scholar] [CrossRef] [PubMed]
  255. Schuff-Werner, P. Challenges of laboratory diagnostics in the elderly. J. Lab. Med. 2018, 42, 105–107. [Google Scholar] [CrossRef]
  256. Agrawal, L.; Engel, K.B.; Greytak, S.R.; Moore, H.M. Understanding preanalytical variables and their effects on clinical biomarkers of oncology and immunotherapy. Semin. Cancer. Biol. 2018, 52, 26–38. [Google Scholar] [CrossRef] [PubMed]
  257. Palleschi, L.; Galdi, F.; Pedone, C. Acute medical illness and disability in the elderly. Geriatr. Care 2018, 4, 7561. [Google Scholar] [CrossRef]
  258. Noppert, G.A.; Stebbins, R.C.; Dowd, J.B.; Aiello, A.E. Sociodemographic differences in population-level immunosenescence in older age. MedRxiv 2022. Preprint. [Google Scholar] [CrossRef]
  259. Chaib, S.; Tchkonia, T.; Kirkland, J.L. Cellular senescence and senolytics: The path to the clinic. Nat. Med. 2022, 28, 1556–1568. [Google Scholar] [CrossRef] [PubMed]
  260. An Open-Label Intervention Trial to Reduce Senescence and Improve Frailty in Adult Survivors of Childhood Cancer. Available online: https://ncorp.cancer.gov/find-a-study/find-a-study.php?id=NCT04733534 (accessed on 18 March 2024).
  261. Targeting Pro-Inflammatory Cells in Idiopathic Pulmonary Fibrosis: A Human Trial (IPF). Available online: https://clinicaltrials.gov/study/NCT02874989 (accessed on 18 March 2024).
  262. Malavolta, M.; Giacconi, R.; Brunetti, D.; Provinciali, M.; Maggi, F. Exploring the relevance of senotherapeutics for the current SARS-CoV-2 emergency and similar future global health threats. Cells 2020, 9, 909. [Google Scholar] [CrossRef] [PubMed]
  263. Lelarge, V.; Capelle, R.; Oger, F.; Mathieu, T.; Le Calvé, B. Senolytics: From pharmacological inhibitors to immunotherapies, a promising future for patients’ treatment. NPJ Aging 2024, 10, 12. [Google Scholar] [CrossRef] [PubMed]
  264. Silverstein, A.R.; Flores, M.K.; Miller, B.; Kim, S.J.; Yen, K.; Mehta, H.H.; Cohen, P. Mito-Omics and immune function: Applying novel mitochondrial omic techniques to the context of the aging immune system. Transl. Med. Aging 2020, 4, 132–140. [Google Scholar] [CrossRef] [PubMed]
  265. Rafferty, E.; Paulden, M.; Buchan, S.A.; Robinson, J.L.; Bettinger, J.A.; Kumar, M.; Svenson, L.W.; MacDonald, S.E. Canadian Immunization Research Network (CIRN) investigators. Evaluating the individual healthcare costs and burden of disease associated with RSV across age groups. Pharmacoeconomics 2022, 40, 633–645. [Google Scholar] [CrossRef] [PubMed]
  266. Mathiasen, S.L.; Gall-Mas, L.; Pateras, I.S.; Theodorou, S.D.P.; Namini, M.R.J.; Hansen, M.B.; Martin, O.C.B.; Vadivel, C.K.; Ntostoglou, K.; Butter, D.; et al. Bacterial genotoxins induce T cell senescence. Cell Rep. 2021, 35, 109220. [Google Scholar] [CrossRef] [PubMed]
  267. Bauer, M.E.; Wieck, A.; Petersen, L.E.; Baptista, T.S. Neuroendocrine and viral correlates of premature immunosenescence. Ann. N. Y. Acad. Sci. 2015, 1351, 11–21. [Google Scholar] [CrossRef] [PubMed]
  268. Karagiannis, T.T.; Dowrey, T.W.; Villacorta-Martin, C.; Montano, M.; Reed, E.; Belkina, A.C.; Andersen, S.L.; Perls, T.T.; Monti, S.; Murphy, G.J.; et al. Multi-modal profiling of peripheral blood cells across the human lifespan reveals distinct immune cell signatures of aging and longevity. EBioMedicine 2023, 90, 104514. [Google Scholar] [CrossRef] [PubMed]
  269. Ahuja, S.K.; Manoharan, M.S.; Lee, G.C.; McKinnon, L.R.; Meunier, J.A.; Steri, M.; Harper, N.; Fiorillo, E.; Smith, A.M.; Restrepo, M.I.; et al. Immune resilience despite inflammatory stress promotes longevity and favorable health outcomes including resistance to infection. Nat. Commun. 2023, 14, 3286. [Google Scholar] [CrossRef] [PubMed]
  270. Yousefzadeh, M.J.; Melos, K.I.; Angelini, L.; Burd, C.E.; Robbins, P.D.; Niedernhofer, L.J. Mouse models of accelerated cellular senescence. Methods Mol. Biol. 2019, 1896, 203–230. [Google Scholar] [CrossRef] [PubMed]
  271. Nielsen, J.L.; Bakula, D.; Scheibye-Knudsen, M. Clinical Trials Targeting Aging. Front. Aging 2022, 3, 820215. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the principal age-related diseases.
Figure 1. Schematic representation of the principal age-related diseases.
Microorganisms 12 00775 g001
Figure 2. Main qualitative age-associated changes in the cells of innate and adaptive immunity [13,28,29,32,35,37,39,40,41,42,43,44,45,46,49,50,51].
Figure 2. Main qualitative age-associated changes in the cells of innate and adaptive immunity [13,28,29,32,35,37,39,40,41,42,43,44,45,46,49,50,51].
Microorganisms 12 00775 g002
Figure 3. Overview of the principal organs, immune components, and targets of omics profiling technologies that can be used to characterize the age-related modification of the immune system at population and single-cell levels. ATAC-seq: assay for transposase-accessible chromatin using sequencing; BS-Seq: bisulfite sequencing; ChiP-seq chromatin immunoprecipitation assays combined with DNA sequencing; scMS: single-cell metabolomics; scRNA-seq: single-cell RNA sequencing; cyTOF: cytometry by time of flight; smFISH: single-molecule fluorescence in situ hybridization; and MS-PTM-MS: tandem mass spectrometry (MS/MS) of posttranslational modifications (PTM).
Figure 3. Overview of the principal organs, immune components, and targets of omics profiling technologies that can be used to characterize the age-related modification of the immune system at population and single-cell levels. ATAC-seq: assay for transposase-accessible chromatin using sequencing; BS-Seq: bisulfite sequencing; ChiP-seq chromatin immunoprecipitation assays combined with DNA sequencing; scMS: single-cell metabolomics; scRNA-seq: single-cell RNA sequencing; cyTOF: cytometry by time of flight; smFISH: single-molecule fluorescence in situ hybridization; and MS-PTM-MS: tandem mass spectrometry (MS/MS) of posttranslational modifications (PTM).
Microorganisms 12 00775 g003
Table 1. Main age-associated immune defects observed in the principal infectious diseases affecting old individuals.
Table 1. Main age-associated immune defects observed in the principal infectious diseases affecting old individuals.
Infective AgentsInnate ImmunityAdaptive Immunity
References References
viralHIVIL-6 and TNF-α elevation[116,118]decreased thymic output[37,119,120]
IFN-α increase and IL-2 reduction expansion of TEMRA cells[88]
expansion of non-functional CD3-CD56-CD16+ NK cells [125]elevated KRECs in patients not needing therapy[121]
decreased T-cell diversity[123]
diminished CD8+ T-cell response in aged mice[135]
reduced public CD8+ TCRαβ clonotypes and TCRαβ diversity[137]
Influenza virusdecrease in macrophage peritoneal phagocytic function [131,132]
reduced uptake of bacteria by monocytes[133]
decreased abundance, activity and migration of DCs[134]
RSVlow levels of serum neutralizing antibody and IFN-γ[141]RSV-specific effector memory T cells prevent symptomatic infection[142]
CMV increased CD8+ CD244+ effector T cells[147]
decreased CD16-/CD16+CD56bright and increase in CD56−CD16+ NK cells percentage[146]lower number of naïve CD4+ and increased effector memory CD4+ and CD8+ T cells[148]
reduced TCR repertoire diversity[151]
EBVexpansion of CD56- NK cells with reduced cytotoxic capacity and IFN-γ production[152]increase in terminally differentiated T cells and decrease in TCR repertoire diversity[153]
expansion of viral-specific exhausted, senescent CD8+ CD28− T cells[154]
HCVhigh plasma levels of SASP proteins[155]increase in intrahepatic senescent, not functional T cells[156,157]
HZVinterferes with the type 1 IFN pathway and the production of pro-inflammatory cytokines[160]reduced frequency of virus-specific effector memory T cells[161]
increases in CD57+ NK cells [159]
Measles virus and parvovirus induction of pro-inflammatory secretome-related factors [164]
WWNV impairment of neutrophils, monocyte/macrophages, DCs, and NK cells[169]
JCV CD4+ T-cell lymphocytopenia, low production of TRECs and KRECs and TCR repertoire restrictions in natalizumab-treated patients[172,173,174]
bacterialMycobacterium tuberculosisalterations in monocyte proportion and phenotype [186]impaired adaptive T-cell immunity[118,186]
reduction in IFN-γ/IL-4 ratio and other pro-inflammatory, such as IL-17A, IL-2, TNF-α[187]reduction in regulatory T cells and polyfunctional IFN-γ+TNF-α+ T cells[187]
imbalanced pro- and anti-inflammatory factor pattern and changes in IL-2 and TNF-α production in the lung [184]
Streptococcus pneumoniaelow opsonic activities of antibodies and phagocytic killing of neutrophils [191]changes in CD27+IgM+ B cells[194]
increase in senescence markers (IL-1α/β, TNF-α, IL-6, and CXCL1) [192]
Escherichia coli and other bacteria inducing urinary tract infectionshigh levels of pro-inflammatory cytokines (IL-6, IL-1β, and TNF-α)[195]formation of bladder tertiary lymphoid tissues and redistribution of B-cell pools from the periphery to mucosal surface that alter the mucosal landscape[195]
aged bladder CXCL13+ macrophages may be responsible for inhibiting development of the adaptive immune responses[198]
decreased macrophage phagocytosis[199]
Gram-positive and Gram-negative intestinal bacteria disequilibriumactivation of DCs [205]
release of pro-inflammatory cytokines, mainly IL-6 and IL-17[206]
Porphyromonas gingivalissenescent cellular markers in DCs[214]
Gram-positive and Gram-negative induced sepsisexpansion of myeloid-derived suppressor cells, inhibiting the function of DCs and macrophages in cirrhosis patients [222]inhibition of Th1 response and induction of Th2 and regulatory T-cell productions[222]
parasiticLeishmaniaexpansion of senescent CD56+ CD57+ NK cells[231]expansion of CD57+ CD4+ lymphocytes [230]
expansion of effector memory CD8+ T cells that re-express CD45RA marker[231]
increased transcriptions of senescence-associated genes in the cutaneous lesions[232]
Trypanosoma cruzicompromised capacity to control the magnitude of inflammation[233]increase in antigen-experienced IFN-γ-producing CD4+ T cells [234]
Helminths compromised Th2 function in mice [235]
HIV: human immunodeficiency virus; RSV: respiratory syncytial virus; CMV: cytomegalovirus; EBV: Epstein–Barr virus; HCV: hepatitis C virus; HZV: herpes zoster virus; WNV: West Nile virus; JCV: JC virus.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Quiros-Roldan, E.; Sottini, A.; Natali, P.G.; Imberti, L. The Impact of Immune System Aging on Infectious Diseases. Microorganisms 2024, 12, 775. https://doi.org/10.3390/microorganisms12040775

AMA Style

Quiros-Roldan E, Sottini A, Natali PG, Imberti L. The Impact of Immune System Aging on Infectious Diseases. Microorganisms. 2024; 12(4):775. https://doi.org/10.3390/microorganisms12040775

Chicago/Turabian Style

Quiros-Roldan, Eugenia, Alessandra Sottini, Pier Giorgio Natali, and Luisa Imberti. 2024. "The Impact of Immune System Aging on Infectious Diseases" Microorganisms 12, no. 4: 775. https://doi.org/10.3390/microorganisms12040775

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop