Translational Research in Gerontology and Geriatrics - Reviews
Submitted: 2022-10-07
Published: 2023-04-05

Healthy aging: when periodontal health matters

Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”; The authors contributed equally.
Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy; The authors contributed equally.
Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy
Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy
Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy
Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy
Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy
Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy
Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy
Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy
Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy
Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy
Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy. Corrisponding author - alessandro.cannavo@unina.it
periodontitis immunosenescence inflammaging aging oral health

Abstract

Providing care for the elderly has been considered a significant challenge for modern medicine. As age progresses, diseases become more frequent and severe than those observed at a younger age. This is particularly relevant for infectious diseases, typical in the elderly and usually associated with poor outcomes. Moreover, when persisting and diffusing into the bloodstream (i.e. bacteremia), these infections keep up with the demand for immune cells’ response and consequently increase the concentration of inflammatory markers systemically. This phenomenon is known as “inflammaging”, which potentially triggers or facilitates the development and progression of several age-related disorders, such as cancer, cardiovascular and neurodegenerative diseases. Periodontal disease is one of the most prominent among the disparate number of causal factors responsible for bacteremia and low-grade systemic inflammation in the aging population. This inflammatory disorder is triggered by a dysbiosis of certain bacterial species that activates a massive local toxic deleterious immune response leading to non-reversible damage of supportive tissues surrounding the teeth. In chronic, oral pathogens and their toxic factors can penetrate the bloodstream contributing to systemic inflammation. Based on this premise, it seems evident that maintaining oral health in the elderly is vital not just for owning healthy mouth but also because it contributes to a healthy aging. This review provides an updated account of molecular insights into the bidirectional association between oral health and “successful” aging.

INTRODUCTION

In the last century, the portion of older adults globally has increased rapidly, with the number of people aged over 60 years old reaching almost 22% in 2050 1. Of course, prolonged life expectancy is associated with an augmented risk of chronic degenerative disorders frequently observed in older populations, with national healthcare systems facing this evolution with enormous costs 2-4. According to Franceschi et al. 5, centenarians representing a model of “successful” or healthy aging usually exhibit medical histories with remarkably low incidence rates of common age-related disorders such as cardiovascular-related diseases (CVDs), diabetes, Parkinson’s and Alzheimer’s disease, and cancer 4,5. Thus, great interest has been raised in understandingthe molecular basis of successful aging and identifyinga potential strategy to achieve it. What differentiates the unsuccessful aging process from a successful one is undoubtedly a different response to a condition known as “inflammaging”. This low-grade chronic inflammatory process, resulting from the long-term stimulation of the innate immune system, contributes to the onset of age-related pathologies. In contrast, in subjects who age successfully (with no co-morbidities), like centenarians, it has been suggested that this inflammatory status is counterbalanced by anti-inflammaging mechanisms 6-9. Of note, several are the cellular and molecular mechanisms involved in inflammaging, and among these, dysbiosis (an imbalance in the host-microbial community) appears to play a pivotal role 6,10. This phenomenon is facilitated by physiologic and pathophysiologic changes occurring with aging, like an impairment in immune system functionality (immunosenescence) that enable host-microbiome alteration and contribute to the incidence and severity of infections, such as periodontitis 11-13. Global data indicate that these oral disorders are highly prevalent among older adults (60 years and older), and due to their infective and inflammatory nature, they represent a significant public health issue in aging populations 14-17. For instance, recent studies depicted a catastrophic situation with 68% of adults ≥ 65 years of age affected with chronic periodontitis in the USA 18,19. In addition, several experimental and clinical data demonstrated that various age-related systemic diseases are strictly associated (in a bidirectional manner) with periodontitis 20,21. Hence, it is more than plausible to speculate that controlling the development and progression of such oral disease may significantly decelerate biological aging 22.

Herein, in this Review, we discuss how periodontitis influences overall health status, representing a causal risk factor of accelerated aging. In particular, we mainly focused on the strict causal relationship between immunosenescence and periodontal disease and the consequent related effects on the development and progression of age-related disorders.

IMMUNOSENESCENCE AND INFLAMMAGING

Biologically, aging is associated with a physiological process of tissue degeneration related to chronic low-grade systemic inflammation 6,23,24 is characterized by high circulating cytokine levels such as tumor necrosis factor α (TNF), interleukin (IL)-6 and IL-1, and anti-inflammatory mediators, including the IL-1 receptor antagonist, and the soluble TNF receptors 1 and 2, despite the absence of general pathophysiological stress or infection 25,26. This condition, known as inflammaging, has been associated with accelerated aging and age-related disorders that increase morbidity and mortality in the elderly population 27,28. Among the numerous processes implicated in inflammaging, immunosenescence is for sure one of the most significant pathological drivers 29. The existence of this process involves both cells of adaptative immunity like T and B cells and those of innate immunity, including natural killer (NK), neuthrophils, monocytes and macrophages.

T-CELLS

T-cells are involved in adaptive immunity and are critical for generating and maintaining long-term immunological memory and protection 30,31. These cells direct robust humoral immunity (CD4+ T cells) and mediate cytotoxic responses (CD8+ T cells). However, as age progresses, some well-established defects in these populations of T-cells occur 30,31. For instance, as demonstrated by Cho, with increasing age, hematopoietic stem cells (HSCs), have a decreased capacity to produce lymphoid progenitors (T, B, and NK cells) and are more prone to generate myeloid progenitors (megakaryocytes, erythroid, myeloid and some dendritic cells) 32. Consequently, the HSCs are more prone to generate myeloid progenitors (megakaryocytes, erythroid, myeloid, and some dendritic cells) 32. Moreover, in humans and mice, the involution of the thymus, a specialized central lymphoid organ located in the thorax, has been associated with a decreased output of naïve T cells 31. As a result, circulating naïve T-cells undergo homeostatic proliferation into unprimed memory-like CD8 T cells, called “virtual memory” (VM) cells 33,34.

Further to these factors, significant evidence suggests that persistent latent infections (i.e. cytomegalovirus [CMV]) are associated with the deleterious modifications observed in the T-cell compartment with age 35,36. Indeed, studies reveal that chronic CMV infection impacts the memory compartment and stimulates the expansion of CMV-specific memory CD8+ T-cells 37.

Altogether these mechanisms lead to a decreased response to infections and reduced vaccination efficiency in the older population. Most of these effects have been attributed to the age-associated loss in T cells of the CD28 molecule 38-41. This costimulatory molecule interacts with ligands expressed on antigen-presenting cells (APCs), stimulating T-cell activation and proliferation 41. From a functional standpoint, loss of CD28 leads to several upstream and downstream outcomes, including decreased ability to secrete IL-2, reduced T-cell repertoire diversity, decreased telomerase activity, proliferation, and survival upon antigen exposure 42-45. Although the mechanisms responsible for the loss in CD28 with age are not fully comprehended, several studies suggest a potential implication for TNF-α. Indeed, this factor can significantly inhibit the expression of CD28, and its levels are increased in older adults with chronic inflammaging 46.

In addition, as demonstrated by Patrick et al., the downregulation of CD28, along with the reduced expression of CD27, is accompanied by the loss of human telomerase reverse transcriptase (hTERT), leading to a decreased telomerase activity and increased telomers frailty 47.

Of note, memory CD8+ T cells also acquire CD57, a glycoepitope well-recognized marker of replicative senescence 48, and as reported by Martínez-Zamudio et al. develop high levels of senescence-associated β-galactosidase (SA-βGal) activity 49.

B-CELLS

B-cells are responsible for developing long-lasting protective antibody responses and immunological memory following infections or vaccination. Hence, a defect in the function of these cells negatively impacts their capacity to trigger a correct primary or secondary response 46,50. B cell development depends on lymphoid-biased hematopoietic stem cells (HSC), which develop into pro-B cells, pre-B cells, and immature B cells in the bone marrow. Once they exit from the bone marrow, these immature cells complete their maturation. These mature cells are composed of two peripheral pools of follicular or marginal zone B cells 51. Of note, total peripheral B cell count, which has been associated with impaired bone marrow production in the bone marrow, progressively decline with aging 52. This phenomenon is at least in part dependent on the above-mentioned age-associated switching of HSCs from lymphoid- to myeloid-biased cells that lead to a reduction in both T and B-cells production 32.

In addition, as suggested by Stephan et al. 53,54 an altered function of bone marrow stromal cells (BMSCs) and pro-B cells impacts mature B-cells production. Indeed, BMSCs produce the IL-7, which is essential for the transition of pro-B into pre-B cells, and as demonstrated by these authors, BMSCs from aged mice produce less IL-7. In addition, pro-B-cells from aging mice appear less responsive to IL-7 signaling.

As demonstrated in aged mice, the residual pools of pro-B cells present lowered levels of the surrogate light chain (SLC) proteins λ5 and VpreB 55-57. This effect overlaps with declines in the expression of E47/E2A 56,58-60 and Early B-cell factor (EBF) 61,62. Both these factors regulate the SLC gene expression 63. The reduction in SLC causes a loss of pre-B cell receptors (pre-BCR), restraining the expansion and development of pre-B cells and reducing the production of B cells with normal functions 57.

Moreover, the increased secretion of TNF-α by old follicular B cells 64 yields the apoptosis of SLC+ pro-B cells in the bone marrow, consequently leading to the accumulation of SLC deficient B cells that inhibit the production of immature B cells 65. The signaling pathways mentioned above drastically interfere with the development of defensive humoral immunity in response to infectious pathogens in the aged population.

NK CELLS

Natural killer (NK) cells are innate lymphoid cells that comprise 10~15% of the circulating lymphocyte population and play an important role in early defense against pathogens and tumor cells 66. These cytotoxic cells can be subdivided into different subsets based on the expression of the surface markers CD16 (also known as Fc-gamma receptor [FcγRIIIA]) and CD56 (also known as a neural cell adhesion molecule [N-CAM]).

The majority (about 90%) of circulating NK cells have a low-density expression of CD56dim that, accordingly to Lanier and coworkers, are mature NK cells with higher cytotoxic capacity 67. On the contrary, almost 10% of NK cells that express CD56bright are more immature and secrete cytokines and chemokines, including IFN-γ 68,69. With age, there is a progressive decline in the CD56bright population and an accumulation of the CD56dim population that begins to express CD57 (CD56dimCD57+ NK cells) 70. This pool of CD56dimCD57+ cells shows high cytolytic capacity but reduced responsiveness to cytokines 71. Moreover, impairment of NK cell cytotoxicity on a per-cell basis has also been reported 72. Importantly, this effect has been partly attributed to the decreased expression of the NK protein 30 (NKp30), also known as natural cytotoxicity receptor 3 (NCR3 or CD337), in elderly individuals 73,74. In centenarians, this NK subset maintains well-preserved cytotoxicity, which presumably helps this population to achieve advanced age in good conditions 75.

NEUTROPHILS

Polymorphonuclear neutrophils (PMNs) play a pivotal role in the innate immune system representing one of the first lines of defense against pathogens. PMNs migrate from the blood to sites of inflammation and infection, where they recognize and phagocyte the invading microorganisms to kill them via different cytotoxic mechanisms, including the generation of reactive oxygen (ROS) and nitrogen (RNS) species and the release of proteases and antimicrobial peptides 76,77. In addition, PMNs can form neutrophil extracellular traps (NETs) that entrap and kill pathogens via oxidative and non-oxidative mechanisms 78. Notably, in the absence of specific stimuli, their lifespan is relatively short, however, pro-inflammatory stimuli like bacterial lipopolysaccharide (LPS) can significantly raise it 79. Even though aging, their number is preserved 80,81, some relevant age-related perturbations in neutrophil function, including dysfunctional phagocytic and chemotactic abilities, have been described 82-84. In this regard, a reduction in free radical ROS production by neutrophils has been reported in older adults. Importantly, ROS are produced after oxidative bursts in phagosomes and are pivotally involved in the microbicidal function of these cells via induction of NET formation 85,86. Because of these age-related effects on neutrophil function, there is a compromised ability of these cells to serve as primary responders to infections 84-88.

MONOCYTES AND MACROPHAGES

Like neutrophils, monocytes, and macrophages are phagocytic cells critically involved in the innate response against infection. Importantly, circulating monocytes represent the precursors to tissue-resident macrophages that, upon differentiation, act as one of the leading cells deputed to host defense in response to exogenous agents 89,90. However, several defects in monocyte and macrophage function, primarily due to cellular signaling dysregulation, have been reported with age. Importantly, in monocytes, these defects are particularly evident upon TLRs stimulation. For instance, Nyugen and coworkers 91 reported that monocytes isolated from older individuals exhibited a decreased expression of TLR1, resulting in impaired IL-6 and TNF-α production that negatively impacted the phagocytosis activity of these cells. In line with these data, Metcalf et al. 92 reported that in response to TLR4 and TLR7/8 stimulation, human monocytes from adults and old subjects showed significant differences at the transcriptional and functional levels, not observed in the absence of a stimulus. These authors demonstrated the impaired production of interferons (IFN-α and -γ), IL-1β, and chemokines like CCL20 and CCL8. Of note, the basis for part of these age-related defects in macrophages has been attributed to an altered expression of TLRs on the cell membrane of these cells with advancing age 93. However, in their report, Boehmer and coworkers 94 observed that TLRs expression was not impacted in aging macrophages. Therefore, these authors suggested that part of the functional defects observed in macrophages, like altered cytokine production, were mainly related to intracellular signaling. Specifically, these authors reported that in response to LPS, there was impaired phosphorylation of mitogen-activated protein kinases (MAPK) like p38 and JNK. Despite these controversial results, other reports have suggested that defects at the level of TLR expression or intracellular signaling appear to be highly related to the increased susceptibility to and severity of microbial infections in the elderly population 95-98. However, Pattabiraman et al. analyzing the responses elicited by a wide array of TLR agonists in distinct populations of murine macrophages provided data suggesting that extensive changes in TLR responsiveness are not associated with age. Of note, these authors reported that effects seen in response to TLRs stimulation could not account in full for the altered inflammatory status and cytokine production/release typical of immunosenescent macrophages. Therefore, these studies demand more investigations to elucidate the mechanisms responsible for “macrophaging” 99. In this context, recent reports demonstrated that inflammaging could expand the content of activated M2-like macrophages that increase the inflammatory status of tissues and express several senescence markers. Therefore, this indicates that aging in macrophages impacts many processes, including TLR signaling, phagocytosis, and polarization.

INFLAMMAGING AND FRAILTY

Immunosenescence and inflammaging represent crucial contributors to age- and frailty-related ailments affecting whole systemic health 100-102.

Frailty is a complex physiological syndrome characterized by increased susceptibility to stressors and reduced physiological reserves that, from a biological point of view, is driven by a gradual and lifelong accumulation of molecular defects, including those affecting the immune system.

In this regard, several proofs have corroborated such a theory 103. For instance, Leng et al. 104 provided the first evidence of a direct link between frailty and inflammation, showing that community-dwelling older frail individuals presented with higher serum IL-6 levels than non-frail subjects. In addition, the longitudinal InCHIANTI study showed that high levels of inflammatory molecules like IL-6, IL-1, and CRP are associated with poor overall physical performance and reduced muscle strength 105. In line with this, the Longitudinal Aging Study of Amsterdam (LASA) identified CRP as a risk factor for frailty 101,106.

Analogously, the Women’s Health and Aging Study found that IL-6 levels are higher in frail individuals than in non-frail counterparts 107. Finally, the Newcastle 85+ study has confirmed the importance of these inflammatory markers in frailty 108.

To date, several reports demonstrated that in frail subjects, pro-inflammatory secretion appears to be driven by an altered functionality of immune cells (i.e., immunosenescence) in response to a chronic infection. Indeed, Leng and coworkers 109, showed that peripheral blood mononuclear cells (PBMC) from older frail adults, after persistent stimulation with bacterial LPS, proliferated less and augmented the release of pro-inflammatory cytokines. Further, Qu et al. 110 demonstrated that monocytes from frail older individuals exposed to LPS presented an increased expression of genes encoding cytokines and chemokines compared to non-frail counterparts. For their part, Schmaltz et al. 111 demonstrated that Cytomegalovirus (CMV) infection was associated with physical frailty and that IL-6 enhanced the extent of such association. Finally, Kawamura and colleagues 112 demonstrated that chronic exposure of mice to the LPS of Porphyromonas gingivalis (P. gingivalis), one of the major pathogenic factors for periodontitis 113,114 increased muscle atrophy participating in the development of physical frailty and sarcopenia.

Overall, the aforementioned mechanisms support the interrelation between the impairment of the functionality of immune cells (immunosenescence), the increased incidence and severity of infections observed in older subjects, and the impact on frailty 103.

AGING AND PERIODONTITIS

The oral cavity is one of the main portals of entry for several microorganisms (bacteria, viruses, and fungi) composing the human microbiome 115. The oral microbiome, the so-called Oralome, is a fundamental component of the human microbiome and is composed of approximately 800-1000 microbial species, with oral bacteria (commensal and opportunistic) representing the main constituents 115,116. Within the mouth, have been identified different habitats for oral microorganisms. For instance, it has been calculated that one milliliter of human saliva holds approximately 100 million bacterial cells (planktonic free-floating phase). Of note, these planktonic species represent the primary source of bacteria able to colonize the diverse soft and hard surfaces in the oral cavity. In this regard, mucosal sites (shedding surfaces) present monolayers of bacteria that regularly desquamate, like cheek and palate, or stable multilayers of biofilm-like bacteria (tongue) 113. Alternatively, non-shedding ones, including natural and artificial teeth, orthodontic appliances, and tooth fillings, are colonized by a film of bacteria and sugars (bio-film) 114. Different studies have demonstrated that several external factors such as diet, stress, smoke, alcohol, and food planes are associated with oral microbiome alteration, impacting the stability between commensal and pathogenic microorganisms forming the dental plaque, thus leading to oral disorders like periodontitis 117. Importantly different bacterial “complexes” that compose this biofilm have been identified by Haffajee et al. 118 and Socransky and colleagues 119 with progression from facultative to anaerobe species finally responsible for gingivitis and periodontitis 118-120. In this regard, these authors described three main complexes: the green/yellow complex that comprises mainly streptococci; the orange complex, with Fusobacterium nucleatum being the most important 121 and the red complex that includes the gram-negative periodontal pathogens Porphyromonas (P.) gingivalis, Tannerella forsythia, and Treponema denticola 113,114. Mechanistically, the dysbiosis of periodontal pathogenic bacteria triggers a massive harmful local immune response. In addition, the augmented concentration of bacterial virulence factors, such as the LPS, stimulates the production of cytokines and inflammatory mediators (e.g., ILs, prostaglandin E2 (PGE2) and TNF-α) that contribute to alveolar bone reabsorption by osteoclasts 122. Due to gingival tissue and bone destruction, oral pathogens and their toxic factors spread into the bloodstream (bacteremia), leading to systemic inflammation. Therefore, periodontitis represents a perfect model of inflammaging with chronic infections induced by oral pathogens dysbiosis that negatively impacts the health status of the young and old population, promoting accelerated aging. In this regard, levels of inflammatory mediators, including C-reactive protein (CRP), TNF-α, and IL-6, which are elevated in periodontitis, represent the linchpin for disorders like diabetes, cardiovascular and neurodegenerative diseases development that have been previously reviewed by us 113,123. Moreover, it has been recently suggested that periodontal disease is a risk factor for complications of SARS-CoV-2 infection 124,125.

Importantly, a healthy immune system can prevent and tolerate the occurrence of constant acute inflammatory reactions despite a great oral microbial load. However, defects in host immunoregulation, like those observed with aging, are the major mechanism contributing to periodontitis. Indeed, several reports demonstrated the strict correlation between immunosenescence and periodontitis. For instance, Bodineau and colleagues 126 reported a substantial reduction in the ratio of gingival CD4+lymphocyte subset when comparing older with younger patients with chronic periodontitis. On the other hand, Clark et al. 127 analyzed the effects of aging on macrophages and reported that the age-related alteration in macrophages’ function is responsible for the higher prevalence of this disorder and reduced recovery in old individuals compared to their younger counterparts. Along the same lines, a study from Liang et al. 128, examining young and old macrophages under resting conditions or following infection with P. gingivalis demonstrated that aged macrophages presented with elevated expression of surface receptors that amplify inflammation, like the triggering receptor expressed on myeloid cells (TREM]-1) and the C5a anaphylatoxin receptor (C5aR). Interestingly, P. gingivalis, through direct activation of C5aR, disarms and subverts host immunity, further supporting the possibility that immunosenescence can contribute to the persistence of microbial communities that drive dysbiotic disorders and increase the susceptibility to infections 129.

THE IMPORTANCE OF ORAL HEALTH IN OLDER PERSONS

The presence of periodontitis affects human health in two ways: the spreading of bacteria into the bloodstream and inflammaging. These motivations represent an additional reason not to neglect the periodontal state of the elderly and to push national systems toward a periodontitis prevention campaign for the whole population. Indeed, among the interventions to prevent and decrease infections in older adults, emerging evidence demonstrated that the treatment of periodontitis is efficient in positively impacting general health 130.

In this context, the standard treatment is to control the infection, thus preventing local inflammation. This effect can be achieved by removing the plaque by professional supra and subgingival debridement, rigorous home dental care, and correcting bone defects caused by periodontitis by employing periodontal regenerative surgery 131. Currently, regenerative periodontal therapy is conducted by eliminating pathogens and applying biomaterials. However, recent studies support the usage of Human dental pulp stem cells (hDPSCs), one of the readily available sources of multipotent mesenchymal stem cells (MSCs), positively impacting not only the regeneration of periodontium (via a natural process) but resolving the infection and then inflammation thanks to their immunomodulatory role 131,132. Therefore, periodontal treatment represents a valid strategy to counteract the adverse effects of the inflammaging triggered by oral dysbiosis. For instance, several studies demonstrated to positive effects of periodontal treatment on endothelial dysfunction 133-136. This conditionoccurs during the aging process and is related to oxidative stress and vascular inflammation 137, and several pre-clinical and clinical studies have found a direct association with periodontitis. For instance, P. gingivalis can increase the endothelial monocyte chemoattractant protein-1 (MCP-1) expression and induce a rise in endothelin-1 levels and release 138,139. Notably, in these studies, it has been demonstrated that periodontal treatment resulted in benefits in oral health that were accompanied with improvement in endothelial function and a reduction of circulating inflammatory markers. Beneficial effects associated with periodontal treatment were also observed in patients with type 2 diabetes 140,141 and rheumatoid arthritis 142. Notably, these disorders, whose incidence increases with age 143,144, have been directly associated with periodontitis, also in a bidirectional manner 113,123,145 and interestingly, it has been suggested that both these disorders can develop because of premature aging (immunosenescence) of the immune system 144,146.

Further, a recent study from Schwahn and coworkers 147 reported that periodontal treatment had a favorable effect on Alzheimer’s disease, the most prevalent form of dementia in the elderly, reducing brain atrophy. In this regard, we have previously reported a bidirectional link between Alzheimer’s disease and periodontitis 123. Indeed, while oral pathogens’ dysbiosis and their toxic proteins represent a severe risk factor for neurodegeneration and neuroinflammation, the decline of cognitive functions in Alzheimer’s disease patients negatively impacts proper oral hygiene practices, thus favoring periodontitis and tooth loss.

On the same premise, recent findings suggest that improvement in oral health contributes to the prevention of Sarcopenia, a typical syndrome of old individuals characterized by a significant loss of muscle mass and function 148. In this regard, Han et al. 149, in a cross-sectional study, reported that periodontitis was significantly associated with Sarcopenia. In line with this study, Abe et al. 148 reported that improving oral health among community-dwelling older adults contributes to preventing Sarcopenia and Diabetes. Although experimental evidence concerning the correlation between periodontitis and Sarcopenia is limited to date, studies like the one by Kawamura et al. 112, discussed above support such relationship.

CONCLUSIVE REMARKS

This narrative Review article aimed to condense the current knowledge regarding the bidirectional association between oral health and healthy aging (Fig. 1). As discussed, immunosenescence is a major cause of augmented incidence and severity of pathogens infections, consequently leading to inflammaging in a vicious cycle that fuels itself with age. In this regard, we focused on periodontitis, a chronic inflammatory disease with high incidence in the elderly, that is characterized by a dysbiosis of pathogenic bacteria that stimulate a local and systemic inflammation representing the best example of a noxious age-related process that worsens if untreated. Therefore, adequate countermeasures aiming at counteracting the adverse effects of oral dysbiosis on systemic health should be implemented at an early age in line with the idea that “prevention is better than cure”. Of note, nutritional interventions, amelioration of social conditions, and correct lifestyle are undoubtedly considered optimal strategies to prevent oral disorders, given that proper oral hygiene exists. Thus, oral health status may be a predictor of longevity, and this hypothesis has been supported by recent studies showing centenarians had a lower rate of edentulism and prevalence of severe periodontal disease than their younger counterparts 150,151. For all these reasons, several initiatives have been activated with which the Italian Society of Gerontology and Geriatrics (SIGG) should associate itself, like the one recently launched by The Gerontological Society of America called “Oral Health An Essential Element of Healthy Aging” (). This program has the goals of supporting older adults to keep their oral health as part of a healthy aging process and to help researchers, practitioners, educators, and policymakers to identify areas of research/activity on the subject of “oral health in older adults”.

Conflict of interest statement

The authors declare no conflict of interest.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author contributions

CR, AV: wrote and revised the manuscript; DL, GS, GC, FDL, MRL, AP: wrote the manuscript; GR, NF, SR: revised the manuscript; RV, AC: wrote, revised, and edited the manuscript.

Ethical consideration

Not applicable.

Figures and tables

Figure 1.Schematic representation summarizing the relationship between immunosenescence and periodontal disease and their effects on the development and progression of age-related disorders. As age progress, immunosenescence negatively impact on immune cells functionality and number thus augmenting the incidence and severity of oral pathogens dysbiosis and infection with consequent local and systemic inflammation thus fueling the inflammaging process. Red arrows indicate reduced.Green arrows indicate increased.

References

  1. Rahimi Foroushani A, Estebsari F, Mostafaei D. The effect of health-promoting intervention on healthy lifestyle and social support in elders: a clinical trial study. Iran Red Crescent Med J. 2014; 16:e18399. DOI
  2. Dominguez LJ, Veronese N, Baiamonte E. Healthy aging and dietary patterns. Nutrients. 2022; 14:889. DOI
  3. Brown GC. Living too long: the current focus of medical research on increasing the quantity, rather than the quality, of life is damaging our health and harming the economy. EMBO Rep. 2015; 16:137-141. DOI
  4. Borras C, Ingles M, Mas-Bargues C. Centenarians: an excellent example of resilience for successful ageing. Mech Ageing Dev. 2020; 186:111199. DOI
  5. Franceschi C, Passarino G, Mari D. Centenarians as a 21st century healthy aging model: a legacy of humanity and the need for a world-wide consortium (WWC100+). Mech Ageing Dev. 2017; 165:55-58. DOI
  6. Franceschi C, Garagnani P, Parini P. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol. 2018; 14:576-590. DOI
  7. Strindhall J, Nilsson B-O, Löfgren S. No immune risk profile among individuals who reach 100 years of age: findings from the Swedish NONA immune longitudinal study. Exp Gerontol. 2007; 42:753-761. DOI
  8. Franceschi C, Bonafè M. Centenarians as a model for healthy aging. Biochem Soc Trans. 2003; 31:457-461. DOI
  9. Arai Y, Martin-Ruiz CM, Takayama M. Inflammation, but not telomere length, predicts successful ageing at extreme old age: a longitudinal study of semi-supercentenarians. EBioMedicine. 2015; 2:1549-1558. DOI
  10. Ebersole JL, Graves CL, Gonzalez OA. Aging, inflammation, immunity and periodontal disease. Periodontol 2000. 2016; 72:54-75. DOI
  11. Hajishengallis G. The inflammophilic character of the periodontitis-associated microbiota. Mol Oral Microbiol. 2014; 29:248-257. DOI
  12. Jin LJ, Lamster IB, Greenspan JS. Global burden of oral diseases: emerging concepts, management and interplay with systemic health. Oral Dis. 2016; 22:609-619. DOI
  13. Papapanou PN, Susin C. Periodontitis epidemiology: is periodontitis under-recognized, over-diagnosed, or both?. Periodontol 2000. 2017; 75:45-51. DOI
  14. Marcenes W, Kassebaum BE, Flaxman A. Global burden of oral conditions in 1990-2010: a systematic analysis. J Dent Res. 2013; 92:592-597.
  15. Kassebaum NJ, Bernabe E, Dahiya M. Global burden of untreated caries: a systematic review and metaregression. J Dent Res. 2015; 94:650-658.
  16. Kassebaum NJ, Bernabe E, Dahiya M. Global burden of severe periodontitis in 1990-2010: a systematic review and meta-regression. J Dent Res. 2014; 93:1045-1053.
  17. Eke PI, Thornton-Evans GO, Wei L. Periodontitis in U.S. adults: National Health and Nutrition Examination Survey 2009-2014. J Am Dent Ass. 2018; 149:576-588.
  18. Michaud DS, Fu Z, Shi J. Periodontal disease, tooth loss, and cancer risk. Epidemiol Rev. 2017; 39:49-58. DOI
  19. Eke PI, Dye BA, Wei L. Update on prevalence of periodontitis in adults in the United States: NHANES 2009 to 2012. J Periodontol. 2015; 86:611-622. DOI
  20. Ebersole JL, Dawson DA, Emecen Huja P. Age and periodontal health - immunological view. Curr Oral Health Rep. 2018; 5:229-241. DOI
  21. Ebersole JL, Lambert J, Bush H. Serum nutrient levels and aging effects on periodontitis. Nutrients. 2018; 10:1986. DOI
  22. Nguyen LM, Chon JJ, Kim EE. Biological aging and periodontal disease: analysis of NHANES (2001-2002). JDR Clin Trans Res. 2022; 7:145-153. DOI
  23. López-Otín C, Blasco MA, Partridge L. The Hallmarks of aging. Cell. 2013; 153:1194-1217. DOI
  24. Kinn PM, Holdren GO, Westermeyer BA. Age-dependent variation in cytokines, chemokines, and biologic analytes rinsed from the surface of healthy human skin. Sci Rep. 2015; 5:10472. DOI
  25. Fabbri E, An Y, Zoli M. Aging and the burden of multimorbidity: associations with inflammatory and anabolic hormonal biomarkers. J Gerontol A Biol Sci Med Sci. 2015; 70:63-70. DOI
  26. Ballou SP, Lozanski FB, Hodder S. Quantitative and qualitative alterations of acute-phase proteins in healthy elderly persons. Age Ageing. 1996; 25:224-230. DOI
  27. Ferrucci L, Fabbri E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol. 2018; 15:505-522. DOI
  28. Howcroft TK, Campisi J, Louis GB. The role of inflammation in age-related disease. Aging. 2013; 5:84-93. DOI
  29. Fulop T, Larbi A, Dupuis G. Immunosenescence and Inflamm-aging as two sides of the same coin: friends or foes?. Front Immunol 201810. 8:1960. DOI
  30. Tu W, Rao S. Mechanisms underlying T cell immunosenescence: aging and cytomegalovirus infection. Front Microbiol. 2016; 7:2111. DOI
  31. Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nat Immunol. 2004; 5:133-139.
  32. Cho RH, Sieburg HB, Muller-Sieburg CE. A new mechanism for the aging of hematopoietic stem cells: aging changes the clonal composition of the stem cell compartment but not individual stem cells. Blood. 2008; 111:5553-5561.
  33. Nikolich-Zugich J. Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nature Rev Immunol. 2008; 8:512-522. DOI
  34. Goronzy JJ, Fang F, Cavanagh MM. Naive T cell maintenance and function in human aging. J Immunol. 2015; 194:4073-4080. DOI
  35. Wertheimer AM, Bennett MS, Park B. Aging and cytomegalovirus infection differentially and jointly affect distinct circulating T cell subsets in humans. J Immunol. 2014; 192:2143-2155. DOI
  36. Pawelec G, Derhovanessian E. Role of CMV in immune senescence. Virus Res. 2011; 157:175-179. DOI
  37. Kim J, Kim AR, Shin EC. Cytomegalovirus infection and memory T cell inflation. Immune Netw. 2015; 15:186-190. DOI
  38. Xu W, Wong G, Hwang YY. The untwining of immunosenescence and aging. Semin Immunopathol. 2020; 42:559-572. DOI
  39. Linehan E, Fitzgerald DC. Ageing and the immune system: focus on macrophages. Eur J Microbiol Immunol (Bp). 2015; 5:14-24. DOI
  40. Fagnoni FF, Vescovini R, Mazzola M. Expansion of cytotoxic CD8+ CD28- T cells in healthy ageing people, including centenarians Immunology. 1996; 88:501-507. DOI
  41. Effros RB, Boucher N, Porter V. Decline in CD28+ T cells in centenarians and in long-term T cell cultures: a possible cause for both in vivo and in vitro immunosenescence. Exp Gerontol. 1994; 29:601-609. DOI
  42. Azuma M, Phillips JH, Lanier LL. CD28- T lymphocytes. Antigenic and functional properties. J Immunol. 1993; 150:1147-1159.
  43. Batliwalla FJ, Monteiro D, Serrano P. Oligoclonality of CD8+ T cells in health and disease: aging, infection, or immune regulation?. Hum Immunol. 1996; 48:68-76. DOI
  44. Valenzuela HF, Effros RB. Divergent telomerase and CD28 expression patterns in human CD4 and CD8 T cells following repeated encounters with the same antigenic stimulus. Clin Immunol. 2002; 105:117-125. DOI
  45. Topp MS, Riddell SR, Akatsuka Y. Restoration of CD28 expression in CD28- CD8+ memory effector T cells reconstitutes antigen-induced IL-2 production. J Exp Med. 2003; 198:947-955. DOI
  46. Crooke SN, Ovsyannikova IG, Poland GA. Immunosenescence: a systems-level overview of immune cell biology and strategies for improving vaccine responses. Exp Gerontol. 2019; 124:110632. DOI
  47. Patrick MS, Cheng NL, Kim J. Human T cell differentiation negatively regulates telomerase expression resulting in reduced activation-induced proliferation and survival. Front Immunol. 2019; 10:1993. DOI
  48. Wood KL, Twigg HL, Doseff AI. Dysregulation of CD8+ lymphocyte apoptosis, chronic disease, and immune regulation. Front Biosci (Landmark Ed). 2009; 14:3771-3781. DOI
  49. Martínez-Zamudio RI, Dewald HK, Vasilopoulos T. Senescence-associated β-galactosidase reveals the abundance of senescent CD8+ T cells in aging humans. Aging Cell. 2021; 20:e13344. DOI
  50. Cancro MP, Hao Y, Scholz JL. B cells and aging: molecules and mechanisms. Trends Immunol. 2009; 30:313-318. DOI
  51. LeBien TW, Tedder TF. B lymphocytes: how they develop and function. Blood. 2008; 112:1570. DOI
  52. Zharhary D. Age-related changes in the capability of the bone marrow to generate B cells. J Immunol. 1988; 141:1863-1869.
  53. Stephan RP, Lill-Elghanian DA, Witte PL. Development of B cells in aged mice: decline in the ability of pro-B cells to respond to IL-7 but not to other growth factors. J Immunol. 1997; 158:1598-1609.
  54. Stephan RP, Reilly CR, Witte PL. Impaired ability of bone marrow stromal cells to support B-lymphopoiesis with age. Blood. 1998; 91:75-88.
  55. Sherwood EM, Blomberg BB, Xu W. Senescent BALB/c mice exhibit decreased expression of lambda5 surrogate light chains and reduced development within the pre-B cell compartment. J Immunol. 1998; 161:4472-4475.
  56. Sherwood EM, Xu W, King AM. The reduced expression of surrogate light chains in B cell precursors from senescent BALB/c mice is associated with decreased E2A proteins. Mech Ageing Dev. 2000; 118:45-59. DOI
  57. Alter-Wolf S, Blomberg BB, Riley RL. Deviation of the B cell pathway in senescent mice is associated with reduced surrogate light chain expression and altered immature B cell generation, phenotype, and light chain expression. J Immunol. 2009; 182:138-147. DOI
  58. Frasca D, Nguyen D, Riley RL. Decreased E12 and/or E47 transcription factor activity in the bone marrow as well as in the spleen of aged mice. J Immunol. 2003; 170:719-726. DOI
  59. Van der Put E, Frasca D, King AM. Decreased E47 in senescent B cell precursors is stage specific and regulated posttranslationally by protein turnover. J Immunol. 2004; 173:818-827. DOI
  60. King AM, Van der Put E, Blomberg BB. Accelerated Notch-dependent degradation of E47 proteins in aged B cell precursors is associated with increased ERK MAPK activation. J Immunol. 2007; 178:3521-3529. DOI
  61. Lescale C, Dias S, Maës J. Reduced EBF expression underlies loss of B-cell potential of hematopoietic progenitors with age. Aging Cell. 2010; 9:410-419. DOI
  62. Riley RL. Impaired B lymphopoiesis in old age: a role for inflammatory B cells?. Immunol Res. 2013; 57:361-369. DOI
  63. Sigvardsson M, O’Riordan M, Grosschedl R. EBF and E47 collaborate to induce expression of the endogenous immunoglobulin surrogate light chain genes. Immunity. 1997; 7:25-36. DOI
  64. Frasca D, Romero M, Diaz A. A molecular mechanism for TNF-α-mediated downregulation of B cell responses. J Immunol. 2012; 188:279. DOI
  65. Michelle R, Sarah A, Kelly MA. In aged mice, low surrogate light chain promotes pro-B-cell apoptotic resistance, compromises the PreBCR checkpoint, and favors generation of autoreactive, phosphorylcholine-specific B cells. Aging Cell. 2015; 14:382-390. DOI
  66. Solana R, Tarazona R, Gayoso I. Innate immunosenescence: effect of aging on cells and receptors of the innate immune system in humans. Semin Immunol. 2012; 24:331-341. DOI
  67. Lanier LL, Le AM Civin CI. The relationship of CD16 (Leu-11) and Leu-19 (NKH-1) antigen expression on human peripheral blood NK cells and cytotoxic T lymphocytes. J Immunol. 1986; 136:4480-4486.
  68. Cooper MA, Fehniger TA, Turner SC. Human natural killer cells: a unique innate immunoregulatory role for the CD56(bright) subset. Blood. 2001; 97:3146-3151. DOI
  69. Cooper MA, Fehniger TA, Fuchs A. NK cell and DC interactions. Trends Immunol. 2004; 25:47-52. DOI
  70. Solana R, Campos C, Pera A. Shaping of NK cell subsets by aging. Curr Opin Immunol. 2014; 29:56-61. DOI
  71. Lopez-Vergès S, Milush JM, Pandey S. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood. 2010; 116:3865-3874. DOI
  72. Hazeldine J, Lord JM. The impact of ageing on natural killer cell function and potential consequences for health in older adults. Ageing Res Rev. 2013; 12:1069-1078. DOI
  73. Le Garff-Tavernier M, Beziat V, Decocq J. Human NK cells display major phenotypic and functional changes over the life span. Aging Cell. 2010; 9:527-535. DOI
  74. Almeida-Oliveira A, Smith-Carvalho M, Porto LC. Age-related changes in natural killer cell receptors from childhood through old age. Hum Immunol. 2011; 72:319-329. DOI
  75. Sansoni P, Brianti V, Fagnoni F. NK cell activity and T-lymphocyte proliferation in healthy centenarians. Ann N Y Acad Sci. 1992; 663:50550-7. DOI
  76. Wessels I, Jansen J, Rink L. Immunosenescence of polymorphonuclear neutrophils. Scientific World J. 2010; 10:145-160. DOI
  77. Rosales C, Demaurex N, Lowell CA. Neutrophils: Their role in innate and adaptive immunity. J Immunol Res. 2016; 2016:1469780. DOI
  78. Zawrotniak M, Rapala-Kozik M. Neutrophil extracellular traps (NETs) - formation and implications. Acta Biochim Pol. 2013; 60:277-284.
  79. Hazeldine J, Lord JM. Innate immunesenescence: underlying mechanisms and clinical relevance. Biogerontology. 2015; 16:187-201. DOI
  80. Born J, Uthgenannt D, Dodt C. Cytokine production and lymphocyte subpopulations in aged humans. An assessment during nocturnal sleep. Mech Ageing Dev. 1995; 84:113-126. DOI
  81. Butcher SK, Killampalli V, Lascelles D. Raised cortisol: DHEAS ratios in the elderly after injury: potential impact upon neutrophil function and immunity. Aging Cell. 2005; 4:319-324. DOI
  82. Wenisch C, Patruta S, Daxböck F. Effect of age on human neutrophil function. J Leukoc Biol. 2000; 67:40-45. DOI
  83. Oh SJ, Lee JK, Shin OS. Aging and the immune system: the impact of immunosenescence on viral infection, immunity and vaccine immunogenicity. Immune Netw. 2019; 19:e37. DOI
  84. Niwa Y, Kasama T, Miyachi Y. Neutrophil chemotaxis, phagocytosis and parameters of reactive oxygen species in human aging: cross-sectional and longitudinal studies. Life Sci. 1989; 44:1655-1664. DOI
  85. Hazeldine J, Harris P, Chapple IL. Impaired neutrophil extracellular trap formation: a novel defect in the innate immune system of aged individuals. Aging Cell 201413.690-698. DOI
  86. Lord JM, Butcher S, Killampali V. Neutrophil ageing and immunesenescence. Mech Ageing Dev. 2001; 122:1521-1535. DOI
  87. Simell B, Vuorela A, Ekström N. Aging reduces the functionality of anti-pneumococcal antibodies and the killing of Streptococcus pneumoniae by neutrophil phagocytosis. Vaccine. 2011; 29:1929-1934. DOI
  88. Butcher SK, Chahal H, Nayak L. Senescence in innate immune responses: reduced neutrophil phagocytic capacity and CD16 expression in elderly humans. J Leukoc Biol. 2001; 70:881-886.
  89. Geissmann F, Manz MG, Jung S. Development of monocytes, macrophages, and dendritic cells. Science. 2010; 327:656-661. DOI
  90. Salminen A. Immunosuppressive network promotes immunosenescence associated with aging and chronic inflammatory conditions. J Mol Med (Berl). 2021; 99:1553-1569. DOI
  91. Nyugen J, Agrawal S, Gollapudi S. Impaired functions of peripheral blood monocyte subpopulations in aged humans. J Clin Immunol. 2010; 30:806-813. DOI
  92. Metcalf TU, Wilkinson PA, Cameron MJ. Human monocyte subsets are transcriptionally and functionally altered in aging in response to pattern recognition receptor agonists. J Immunol. 2017; 199:1405-1417. DOI
  93. Renshaw M, Rockwell J, Engleman C. Cutting edge: impaired Toll-like receptor expression and function in aging. J Immunol. 2002; 169:4697-4701. DOI
  94. Boehmer ED, Goral J, Faunce DE. Age-dependent decrease in Toll-like receptor 4-mediated proinflammatory cytokine production and mitogen-activated protein kinase expression. J Leukoc Biol. 2004; 75:342-349. DOI
  95. Sun Y, Li H, Yang MF. Effects of aging on endotoxin tolerance induced by lipopolysaccharides derived from Porphyromonas gingivalis and Escherichia coli. PLoS One. 2012; 7:e39224. DOI
  96. van Duin D, Mohanty S, Thomas V. Age-associated defect in human TLR-1/2 function. J Immunol. 2007; 178:970-975. DOI
  97. Yoon P, Keylock KT, Hartman ME. Macrophage hypo-responsiveness to interferon-gamma in aged mice is associated with impaired signaling through Jak-STAT. Mech Ageing Dev. 2004; 125:137-143. DOI
  98. Schütze S, Kaufmann A, Bunkowski S. Interferon-gamma impairs phagocytosis of Escherichia coli by primary murine peritoneal macrophages stimulated with LPS and differentially modulates proinflammatory cytokine release. Cytokine X. 2021; 3:100057. DOI
  99. Sebastián C, Espia M, Serra M. MacrophAging: a cellular and molecular review. Immunobiology. 2005; 210:121-126. DOI
  100. Niebla-Cárdenas A, Bareke H, Juanes-Velasco P. Translational research into frailty from bench to bedside: salivary biomarkers for inflammaging. Exp Gerontol. 2023; 171:112040. DOI
  101. Fulop T, McElhaney J, Pawelec G. Frailty, inflammation and immunosenescence. Interdiscip Top Gerontol Geriatr. 2015; 41:26-40. DOI
  102. Franceschi C, Bonafè M, Valensin S. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann NY Acad Sci. 2000; 908:244-254. DOI
  103. Cannavo A, Carandina A, Corbi G. Are skeletal muscle changes during prolonged space flights similar to those experienced by frail and sarcopenic older adults?. Life (Basel). 2022; 12:2139. DOI
  104. Leng S, Chaves P, Koenig K. Serum interleukin-6 and hemoglobin as physiological correlates in the geriatric syndrome of frailty: a pilot study. J Am Geriatr Soc. 2002; 50:1268-1271. DOI
  105. Bandeen-Roche K, Walston JD, Huang Y. Measuring systemic inflammatory regulation in older adults: evidence and utility. Rejuvenation Res. 2009; 12:403-410. DOI
  106. Puts MT, Visser M, Twisk JW. Endocrine and inflammatory markers as predictors of frailty. Clin Endocrinol (Oxf). 2005; 63:403-411. DOI
  107. Volpato S, Guralnik JM, Ferrucci L. Cardiovascular disease, interleukin-6, and risk of mortality in older women: the women’s health and aging study. Circulation. 2001; 103:947-953. DOI
  108. Collerton J, Martin-Ruiz C, Davies K. Frailty and the role of inflammation, immunosenescence, and cellular ageing in the very old: cross-sectional findings from the Newcastle 85+ Study. Mech Ageing Dev. 2012; 133:456-466. DOI
  109. Leng SX, Yang H, Walston JD. Decreased cell proliferation and altered cytokine production in frail older adults. Aging Clin Exp Res. 2004; 16:249-252. DOI
  110. Qu T, Walston JD, Yang H. Upregulated ex vivo expression of stress-responsive inflammatory pathway genes by LPS-Challenged CD14(+) monocytes in frail older adults. Mech Ageing Dev. 2009; 130:161-166. DOI
  111. Schmaltz HN, Fried LP, Xue QL. Chronic cytomegalovirus 919 infection and inflammation are associated with prevalent frailty in community-dwelling older women. J Am Geriatr Soc. 2005; 53:747-754. DOI
  112. Kawamura N, Ohnuki Y, Matsuo I. Effects of chronic Porphyromonas gingivalis lipopolysaccharide infusion on skeletal muscles in mice. J Physiol Sci. 2019; 69:503-511. DOI
  113. Liccardo D, Cannavo A, Spagnuolo G. Periodontal disease: a risk factor for diabetes and cardiovascular disease. Int J Mol Sci. 2019; 20:E1414. DOI
  114. Del Giudice C, Vaia E, Liccardo D. Infective endocarditis: a focus on oral microbiota. microorganisms. 2021; 9:1218. DOI
  115. Radaic A, Kapila YL. The oralome and its dysbiosis: new insights into oral microbiome-host interactions. Comput Struct Biotechnol J. 2021; 19:1335-1360. DOI
  116. Kolenbrander PE, Palmer RJ, Periasamy S. Oral multispecies biofilm development and the key role of cell-cell distance. Nat Rev Microbiol. 2010; 8:471-480. DOI
  117. Buduneli N. Environmental factors and periodontal microbiome. Periodontol 2000. 2020; 85:1-12.
  118. Haffajee AD, Socransky S, Feres M. Dental plaque revisited. Oral biofilms in health and disease. BioLine: Cardiff, UK; 1999.
  119. Socransky SS, Haffajee AD, Cugini MA. Microbial complexes in subgingival plaque. J Clin Periodontol. 1998; 25:134-144. DOI
  120. Theilade E, Wright WH, Jensen SB. Experimental gingivitis in man. II. A longitudinal clinical and bacteriological investigation. J Periodontal Res. 1966; 1:1-13. DOI
  121. Kolenbrander PE, Andersen RN, Moore LV. Coaggregation of Fusobacterium nucleatum, Selenomonas flueggei, Selenomonas infelix, Selenomonas noxia, and Selenomonas sputigena with strains from 11 genera of oral bacteria. Infect Immun. 1989; 57:3194-3203. DOI
  122. Hong CY, Lin SK, Kok SH. The role of lipopolysaccharide in infectious bone resorption of periapical lesion. J Oral Pathol Med. 2004; 33:162-169. DOI
  123. Liccardo D, Marzano F, Carraturo F. Potential bidirectional relationship between periodontitis and Alzheimer’s disease. Front Physiol. 2020; 11:683. DOI
  124. Gupta S, Räisänen IT, Sorsa T. Periodontitis as a risk of hospitalization and death by SARS-CoV-2. Int J Public Health. 2022; 67:1605156. DOI
  125. Basso L, Chacun D, Sy K. Periodontal diseases and COVID-19: a scoping review. Eur J Dent. 2021; 15:768-775. DOI
  126. Bodineau A, Coulomb B, Tedesco AC. Increase of gingival matured dendritic cells number in elderly patients with chronic periodontitis. Arch Oral Biol. 2009; 54:12-16. DOI
  127. Clark D, Halpern B, Miclau T. The contribution of macrophages in old mice to periodontal disease. J Dent Res. 2021; 100:1397-1404. DOI
  128. Liang S, Domon H, Hosur KB. Age-related alterations in innate immune receptor expression and ability of macrophages to respond to pathogen challenge in vitro. Mech Ageing Dev. 2009; 130:538-546. DOI
  129. Maekawa T, Krauss JL, Abe T. Porphyromonas gingivalis manipulates complement and TLR signaling to uncouple bacterial clearance from inflammation and promote dysbiosis. Cell Host Microbe. 2014; 15:768-778. DOI
  130. Jeffcoat MK, Jeffcoat RL, Gladowski PA. Impact of periodontal therapy on general health: evidence from insurance data for five systemic conditions. Am J Prev Med. 2014; 47:166-174. DOI
  131. Tatullo M, Riccitiello F, Rengo S. Management of endodontic and periodontal lesions: the role of regenerative dentistry and biomaterials. Dent J (Basel). 2020; 8:32. DOI
  132. Marrelli M, Falisi G, Apicella A. Behaviour of dental pulp stem cells on different types of innovative mesoporous and nanoporous silicon scaffolds with different functionalizations of the surfaces. J Biol Regul Homeost Agents. 2015; 29:991-997.
  133. Seinost G, Wimmer G, Skerget M. Periodontal treatment improves endothelial dysfunction in patients with severe periodontitis. Am Heart J. 2005; 149:1050-1054. DOI
  134. Elter JR, Hinderliter AL, Offenbacher S. The effects of periodontal therapy on vascular endothelial function: a pilot trial. Am Heart J. 2006; 151:47. DOI
  135. Tonetti MS, D’Aiuto F, Nibali L. Treatment of periodontitis and endothelial function. N Engl J Med. 2007; 356:911-920. DOI
  136. Vidal F, Cordovil I, Figueredo CM. Non-surgical periodontal treatment reduces cardiovascular risk in refractory hypertensive patients: a pilot study. J Clin Periodontol. 2013; 40:681-687. DOI
  137. Rodríguez-Mañas L, El-Assar M, Vallejo S. Endothelial dysfunction in aged humans is related with oxidative stress and vascular inflammation. Aging Cell. 2009; 8:226-238. DOI
  138. Hashizume T, Kurita-Ochiai T, Yamamoto M. Porphyromonas gingivalis stimulates monocyte adhesion to human umbilical vein endothelial cells. FEMS Immunol Med Microbiol. 2011; 62:57-65. DOI
  139. Ansai T, Yamamoto E, Awano S. Effects of periodontopathic bacteria on the expression of endothelin-1 in gingival epithelial cells in adult periodontitis. Clin Sci (Lond). 2002; 103:327S-331S. DOI
  140. D’Aiuto F, Gkranias N, Bhowruth D. Systemic effects of periodontitis treatment in patients with type 2 diabetes: a 12 month, single-centre, investigator-masked, randomised trial. TASTE Group. Lancet Diabetes Endocrinol. 2018; 6:954-965. DOI
  141. Teshome A, Yitayeh A. The effect of periodontal therapy on glycemic control and fasting plasma glucose level in type 2 diabetic patients: systematic review and meta-analysis. BMC Oral Health. 2016; 17:31. DOI
  142. Calderaro DC, Corrêa JD, Ferreira GA. Influence of periodontal treatment on rheumatoid arthritis: a systematic review and meta-analysis. Rev Bras Reumatol Engl Ed. 2017; 57:238-244. DOI
  143. Longo M, Bellastella G, Maiorino MI. Diabetes and aging: from treatment goals to pharmacologic therapy. Front Endocrinol (Lausanne). 2019; 10:45. DOI
  144. Chalan P, van den Berg A, Kroesen BJ. Rheumatoid arthritis, immunosenescence and the hallmarks of aging. Curr Aging Sci. 2015; 8:131-146. DOI
  145. González-Febles J, Sanz M. Periodontitis and rheumatoid arthritis: what have we learned about their connection and their treatment?. Periodontol 2000. 202; 87:181-203. DOI
  146. Fülöp T, Dupuis G, Witkowski JM. The role of immunosenescence in the development of age-related diseases. Rev Invest Clin. 2016; 68:84-91.
  147. Schwahn C, Frenzel S, Holtfreter B. Effect of periodontal treatment on preclinical Alzheimer’s disease – results of a trial emulation approach. Alzheimers Dement. 2022; 18:127-141. DOI
  148. Abe T, Tominaga K, Ando Y. S.. Number of teeth and masticatory function are associated with sarcopenia and diabetes mellitus status among community-dwelling older adults: a Shimane CoHRE study. PLoS One. 2021; 16:e0252625. DOI
  149. Han CH, Chung JH. Association between sarcopenia and tooth loss. Ann Geriatr Med Res. 2018; 22:145-150. DOI
  150. Sekundo C, Langowski E, Kilian S. Periodontal and peri-implant diseases in centenarians. J Clin Periodontol. 2020; 47:1170-1179. DOI
  151. Sekundo C, Langowski E, Kilian S. Oral health and functional capacity of centenarians. Sci Rep. 202; 10:22215. DOI

Affiliations

Carlo Rengo

Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”; The authors contributed equally.

Alessandra Valletta

Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy; The authors contributed equally.

Daniela Liccardo

Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy

Gianrico Spagnuolo

Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy

Graziamaria Corbi

Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy

Francesca De Luca

Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy

Maria Rosaria Lauria

Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy

Alessia Perrotta

Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy

Giuseppe Rengo

Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy

Nicola Ferrara

Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy

Sandro Rengo

Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy

Rossella Valletta

Department of Neurosciences, Reproductive and Odontostomatological Sciences, "Federico II" University of Naples, Naples, Italy

Alessandro Cannavo

Department of Translational Medical Sciences, "Federico II" University of Naples, Naples, Italy. Corrisponding author - alessandro.cannavo@unina.it

Copyright

© JOURNAL OF GERONTOLOGY AND GERIATRICS , 2023

How to Cite

[1]
Rengo, C., Valletta, A., Liccardo, D., Spagnuolo, G., Corbi, G., De Luca, F., Lauria, M.R., Perrotta, A., Rengo, G., Ferrara, N., Rengo, S., Valletta, R. and Cannavo, A. 2023. Healthy aging: when periodontal health matters. JOURNAL OF GERONTOLOGY AND GERIATRICS. 72, 1 (Apr. 2023), 11-23. DOI:https://doi.org/10.36150/2499-6564-N580.
  • Abstract viewed - 820 times
  • PDF downloaded - 143 times