Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jun 1.
Published in final edited form as: Periodontol 2000. 2022 Mar 4;89(1):215–230. doi: 10.1111/prd.12421

Maladaptive trained immunity and clonal hematopoiesis as potential mechanistic links between periodontitis and inflammatory comorbidities

George Hajishengallis 1,*, Xiaofei Li 1, Kimon Divaris 2,3, Triantafyllos Chavakis 4
PMCID: PMC9018499  NIHMSID: NIHMS1771490  PMID: 35244943

Abstract

Periodontitis is bidirectionally associated with systemic inflammatory disorders. The prevalence and severity of this oral disease and linked comorbidities increases with aging. Here we review two newly emerged concepts, trained innate immunity (TII) and clonal hematopoiesis of indeterminate potential (CHIP), which together support a potential hypothesis on how periodontitis affects and is affected by comorbidities and why the susceptibility to periodontitis and comorbidities increases with aging. Given that chronic diseases are largely triggered by the action of inflammatory immune cells, modulation of their bone marrow precursors, the hematopoietic stem and progenitor cells (HSPCs), may affect multiple disorders that emerge as comorbidities. Such alterations in HSPCs can be mediated by TII and/or CHIP, two non-mutually exclusive processes sharing a bias for enhanced myelopoiesis and production of innate immune cells with heightened proinflammatory potential. TII is a state of elevated immune responsiveness based on innate immune (epigenetic) memory. Systemic inflammation can initiate TII in the bone marrow via sustained rewiring of HSPCs, which thereby display a skewing towards the myeloid lineage, resulting in generation of hyper-reactive or ‘trained’ myeloid cells. CHIP arises from aging-related somatic mutations in HSPCs which confer a survival and proliferation advantage to the mutant HSPCs and give rise to an outsized fraction of hyper-inflammatory mutant myeloid cells in the circulation and tissues. This review discusses emerging evidence that supports the notion that TII and CHIP may underlie a causal and age-related association between periodontitis and comorbidities. A holistic mechanistic understanding of the periodontitis-systemic disease connection may offer novel diagnostic and therapeutic targets for treating inflammatory comorbidities.

1. Introduction

Periodontal disease is an exemplar of a dysregulated balance in the crosstalk between the local subgingival microbiome and the immune response of the host that is induced to ostensibly control the pathogen challenge. In periodontitis patients, however, the inflammatory response not only fails to mediate protective immunity, but also fuels the selective expansion of pathogenic species leading to dysbiosis 1. Indeed, the inflammatory destruction of connective and bone tissue is not simply a hallmark of the disease but also generates a nutritionally favorable environment (enriched with degraded proteins such as collagen and heme-containing compounds, which are sources of amino-acids and iron, respectively) for periodontitis-associated ‘inflammophilic’ species 2. The flourishing of periodontitis-associated microbial communities under inflammatory conditions exacerbates the inflammatory destruction process, generating a feed-forward loop that promotes the persistence of the dysbiotic microbiome and the chronicity of periodontitis. This oral disease has a significant socioeconomic impact 3,4 and the global prevalence of its severe form is approximately 10% of the adult population 5.

Multiple clinical studies have associated periodontitis with certain inflammatory disorders (comorbidities), such as, cardiovascular disease (CVD), rheumatoid arthritis (RA), psoriasis, inflammatory bowel disease (IBD), type 2 diabetes (T2D), osteoporosis, Alzheimer’s disease, and non-alcoholic fatty liver disease (NAFLD) 613. In this regard, periodontal disease shares inflammatory effector mechanisms with these comorbidities; for instance, interleukin (IL)-23– and IL-17–driven destructive inflammation participates in the pathogenesis of not only periodontitis but also of RA, psoriasis and IBD 1419. Periodontitis also has common genetic or environmental risk factors (e.g., age, socioeconomic status, smoking, diabetes, obesity, hypertension) with other chronic inflammatory disorders 2024. Nevertheless, an independent association between periodontitis and comorbidities persists even upon adjusting known confounding factors 2533. Consistent with this, the activity of certain comorbid conditions (or surrogate markers thereof) and markers of systemic inflammation (e.g., IL-1, IL-6, C-reactive protein, fibrinogen and neutrophil counts in the blood) are improved following successful local periodontal treatment 13,3448. These clinical studies together with emerging collective evidence from preclinical models suggest that periodontitis exerts a systemic effect on overall health above and beyond the local effect in the affected periodontal tissue 49.

In this chapter, we review and discuss emerging evidence in support of potential causal mechanisms that may explain how periodontitis affects and is affected by comorbidities and why the susceptibility to periodontitis and linked comorbidities increases with aging. Specifically, we focus on mechanisms involving maladaptive adaptation of bone marrow hematopoietic stem and progenitor cells (HSPCs) in the context of trained immunity and clonal hematopoiesis 5052. These concepts will be expounded upon after a brief review of evidence establishing the capacity of periodontitis to cause systemic inflammation.

2. Periodontitis-associated systemic inflammation

Studies from independent clinical research groups show that periodontitis is associated with low-grade systemic inflammation (Figure 1). Indeed, as compared with healthy individuals, periodontitis patients display higher levels of IL-1, IL-6, C-reactive protein, and fibrinogen, as well as increased numbers of neutrophils 4345,5364. Other reports document increased serum levels of TNF, IFNγ, IL-17, and oncostatin M in patients with periodontitis 6568. Interestingly, there is significant correlation between the levels of certain inflammatory cytokines (such as TNF, IL-6, oncostatin) in local gingival crevicular fluid samples and corresponding systemic (serum) samples 46,68,69. In both GCF and serum, inflammatory cytokines are found in higher levels in periodontitis relative to health and generally decrease after successful non-surgical periodontal treatment 46,68. The notion that local, tooth-related lesions contribute to the systemic inflammatory burden is also supported by the conclusions of a meta-analysis that apical periodontitis is linked with higher serum levels of C-reactive protein, IL-6, and the central complement component C3, the concentration of which is significantly reduced after the treatment and resolution of apical periodontitis 70.

Figure 1. Periodontitis-related systemic inflammation and inflammatory modulation of bone marrow progenitor cells.

Figure 1.

In periodontitis, bacteria from the tooth-associated dysbiotic biofilm may translocate through the ulcerated periodontal pockets into the blood circulation, resulting in bacteremias and systemic inflammation. Systemic inflammation, in turn, may cause inflammatory modulation of hematopoietic stem and progenitor cells promoting increased myelopoiesis. HSC, hematopoietic stem cell; MPP, multipotent progenitor; GMP, granulocyte-monocyte progenitor.

The periodontitis potential to trigger systemic inflammation is also in keeping with the beneficial actions of local periodontitis treatment on improving systemic inflammatory markers discussed above, 13,3448 as well as with the results of a prospective study of 11,869 individuals (mean age 50) 71. This report demonstrated that poor maintenance of oral hygiene (never or rarely brushing teeth) correlated with enhanced systemic concentrations of fibrinogen and C-reactive protein and increased risk for cardiovascular disease events 71. Although periodontitis is related to elevated risk of common systemic comorbidities 7,9,29,30,7277, the reverse is also true, i.e., periodontitis becomes more prevalent in those patients with systemic comorbidities, such as, RA, cardiometabolic conditions, and IBD 9,26,75,7886. Consistent with this, systemic inflammation (using fibrinogen and white blood cell counts as markers) has been dose-dependently and longitudinally linked with periodontitis in an 11-year prospective study of 1784 participants 87. Therefore, periodontitis and linked comorbidities appear to be bidirectionally associated.

Regarding how periodontitis could affect systemic health, two major mechanisms have been suggested that are consistent with clinical findings. First, periodontal bacteria or their products (such as, LPS and proteases), may reach the blood circulation via the ulcerated epithelium of the periodontal pockets causing systemic complications 6,55,8892. The surface area covered by the epithelium of the periodontal pockets in periodontitis patients is large and estimated at 8 to 20 cm2 93. Consistently, periodontitis is often linked with bacteremias (Figure 1), the occurrence of which may correlate with the severity of clinical periodontal inflammation 55,9498. Second, in severe periodontitis, innate and adaptive immune cells within the connective tissues subjacent to periodontal pockets secrete abundant levels of pro-inflammatory cytokines (e.g., IL-1β, IL-6, G-CSF 99,100), which may thus enter the bloodstream. In this regard, viral infections in the oral cavity that affect the periodontium may modify the periodontal host response in ways that amplify local inflammatory responses and hence increase the systemic inflammatory burden 101,102. Thus, at least in principle, periodontitis-associated systemic inflammation could arise from the spillover of inflammatory cytokines derived from the periodontium to the circulation or induced by the hematogenous translocation of periodontal bacteria and products thereof. The periodontitis-associated systemic Inflammatory response, and particularly IL-6, can additionally cause an acute phase response in the liver, leading to elevated C-reactive protein and fibrinogen that can contribute to systemic complications, such as atherogenesis 55,58,63,64.

Given that chronic inflammatory diseases are, in large part, driven by the action of inflammatory immune cells, it can be reasoned that systemic inflammation-induced alterations in their bone marrow progenitors are likely to reciprocally influence multiple chronic inflammatory disorders that may emerge as comorbidities 51. Alterations in HSPCs in the bone marrow leading to the generation of progeny with augmented proinflammatory potential can occur as a result of trained innate immunity (TII) 50,103 or somatic mutations occurring in the context of the aging-related clonal hematopoiesis of indeterminate potential (CHIP) 104106 (Figure 2). TII and CHIP are not mutually exclusive and, although distinct phenomena, share a bias for enhanced myelopoiesis (over other lineages) resulting in the production of mature myeloid cells with enhanced proinflammatory capacity 51,52. These events, in turn, can influence the initiation and/or the progression of periodontitis and comorbid inflammatory conditions, as discussed in the following sections. Before presenting evidence and engaging in discussions of how TII and CHIP affect periodontitis and associated comorbidities, we provide a brief background on these two important concepts in the context of bone marrow myelopoiesis.

Figure 2. A vicious feed-forward loop linking periodontitis to inflammation-adapted HSPCs in the context of trained immunity and clonal hematopoiesis.

Figure 2.

The ability of bone marrow HSPCs to not only sense but also adapt to pro-inflammatory stimuli underlies the induction of trained immunity, leading to sustained enhanced myelopoiesis and generation of myeloid cells with heightened immune preparedness and responsiveness. Clonal hematopoiesis of indeterminate potential (CHIP) results from age-related somatic mutations in HSPCs that endow the mutant HSPCs with a proliferative advantage, especially under inflammatory conditions, thereby generating an outsized fraction of hyper-inflammatory mutant myeloid cells. Periodontitis-associated systemic inflammation may thus contribute to induction of trained innate immunity and shaping of an inflammatory bone marrow microenvironment conducive for the selective expansion of CHIP-mutant HSC clones. The generated hyper-responsive or hyper-inflammatory myeloid cells are recruited to sites of infection and inflammation and can thus exacerbate periodontitis, which in turn can exert increased inflammatory influence on the bone marrow. Thus, a self-sustained feed-forward loop may be generated between inflammation-adapted HSPCs and periodontitis that could contribute to the chronicity of this oral disorder.

3. Trained innate immunity

The bone marrow is the epicenter of hematopoiesis where the self-renewable hematopoietic stem cell (HSC), also known as long-term (LT)-HSC, has the ability to produce all types of mature blood cells 107. LT-HSCs can differentiate via short-term HSCs (ST-HSCs) to multi-potent progenitors (MPPs). LT-HSCs, ST-HSCs and MPPs comprise the so-called ‘hematopoietic stem and progenitor cells’ (HSPCs) 107109. HSPCs express TLRs and growth factor and cytokine receptors (e.g., receptors for IL-1β, IL-6, M-CSF) 107,109 and can thus respond to systemic inflammation (or infection) by initiating increased production of myeloid cells 51,110 (Figure 1). The standard differentiation pathway for myelopoiesis downstream of MPP cells includes common myeloid progenitors (CMP) and granulocyte macrophage progenitors (GMP), the precursors of monocytes/macrophages and neutrophils.

Under the influence of inflammation, HSPCs may become adapted for long-term enhanced proliferation and skewed differentiation toward the myeloid lineage. This sustained inflammatory modulation of HSPCs is based on enduring cell-autonomous (metabolic, transcriptional, and epigenetic) mechanisms that underlie the induction and persistence of TII, a relatively recent concept 51,111113. Indeed, historically, immunological memory has been viewed as an exclusive ‘privilege’ of the adaptive immune system. However, owing to advances in the past decade, it is now appreciated that innate immune cells can also build and maintain ‘memory’ of earlier inflammatory events, such as those associated with infectious stimuli (e.g., the major fungal cell wall constituent β-glucan) or vaccination (e.g., the bacillus Calmette–Guérin [BCG] vaccine). In this manner, innate immune cells exhibit hyper-responsiveness upon re-challenge; in other words, are in a position to respond faster and stronger to future challenges. However, the anamnestic response is not specific for the original stimulus and thus can be elicited against the same or even unrelated stimulus 114116 (Figure 3). This elevated state of inflammatory activation that is based on non-specific innate immune memory is designated ‘trained innate immunity’ (TII) 50. Accordingly, TII is defined as a non-specific memory elicited irrespective of adaptive immunity upon earlier encounters, and which promotes augmented responsiveness to ensuing homologous or heterologous challenges 50,51,103.

Figure 3. Trained innate immunity.

Figure 3.

Trained innate immunity represents non-specific immune memory which can be induced, independent of adaptive immunity, upon an earlier microbial challenge and can be recalled at later time points in response to a new challenge that is not necessarily the same with the primary one. The induction of the memory or trained phenotype can be initiated in the bone marrow via long-lived metabolic, epigenetic, and transcriptional adaptations in hematopoietic stem and progenitor cells, which in turn give rise to trained mature myeloid progeny with augmented immune and inflammatory responsiveness.

The long-term effects of TII on blood myeloid cells that have relatively limited lifespan (only a few days) in the circulation 117 are explained by the demonstration that TII can be initiated at the level of the bone marrow through long-lived metabolic and transcriptional alterations in HSPCs, the precursors of differentiated innate immune cells 111,112. Specifically, TII induces long-lasting alterations in HSPCs, which, through enhanced myeloid-biased differentiation, can give rise to increased numbers of ‘trained’ myeloid cells 51,112.

At least in preclinical models where trained myelopoiesis is induced by β-glucan, the underlying mechanism involves inflammatory IL-1β-signaling in HSPCs associated with higher IL-1β levels in the bone marrow. Moreover, the activation of HSPCs by β-glucan–induced TII involves their metabolic reprogramming and enhanced GM-CSF/CD131 signaling. Together, these immunometabolic alterations in HSPCs drive myelopoiesis 112. Integral to the long-lasting effects of TII in the bone marrow are inflammation-induced epigenetic modifications in LT-HSCs. Indeed, LT-HSCs in mice trained with LPS can retain epigenetic memory (increased chromatin openness and accessibility) of earlier infections or inflammatory challenges for at least 12 weeks, thereby potentiating myeloid differentiation and innate immune responses following a future challenge 113. The initiation of TII with persistent transcriptomic myeloid skewing of HSPCs in the bone marrow leading to long-lived and increased responsiveness of peripheral myeloid cells to heterologous stimuli was also shown in healthy human individuals upon BCG vaccination118. Overall, therefore, the induction of innate immune memory, upon exposure to microbial and/or inflammatory stimuli, entails metabolic, epigenetic and transcriptional reprogramming which can persist for months 103,119123 (Figure 3).

TII initiated by modulation of LT-HSCs can give rise to trained myeloid cell populations (neutrophils, monocytes/macrophages) that can replenish and fortify the innate immune system upon stress conditions, such as those related to infections or with inflammatory, tumorigenic or chemotherapeutic challenges 111113,124. Consequently, TII is thought to represent an evolutionarily maintained beneficial response that promotes host survival upon re-infection or other future challenges that can be potentially detrimental to the host 103,125128. However, TII may also promote aberrant immune responses that aggravate chronic inflammatory diseases 51,129131. In this regard, TII in the bone marrow can contribute to perpetuation of inflammation both quantitatively and qualitatively, i.e., by increasing the production of myeloid cells which at the same time have enhanced inflammatory responsiveness 51,132,133 (Figure 2).

4. Clonal hematopoiesis of indeterminate potential

In common with other chronic inflammatory diseases, an individual’s susceptibility to periodontitis increases with aging 134138. The term “inflamm-aging” was coined to describe the aging-related elevation of low-grade chronic systemic inflammation (e.g., IL-1β- and IL-6), which coincides with an increase in co-morbidities 139,140. Although the concept of inflamm-aging is well documented epidemiologically, its etiology has been largely unknown 140145. A potential mechanistic explanation for the association of aging with inflammation is CHIP 104106.

CHIP defines an aging-related condition, whereby somatic mutations that are usually related to myeloid malignancies accrue in HSPCs; these mutations confer a survival/proliferation advantage to the mutant HSPC, thereby enabling its clonal expansion and the generation of mutant progeny cells. The latter represent a separate fraction of leukocytes in the peripheral blood. Importantly, however, individuals with CHIP do not have clinical signs of a hematologic disorder, for instance cytopenias 106,146 (Figure 4). The increased mutation accumulation by HSCs with aging may, at least in part, result from the fact that aged HSCs are more resistant to DNA damage-induced apoptosis than young HSCs, owing to decreased DNA damage repair pathways in the former 147. Therefore, aged HSCs may not die despite suffering DNA damage, hence accumulating mutations 147.

Figure 4. CHIP may potentially affect multiple inflammatory disorders.

Figure 4.

With advancing age, HSPCs progressively acquire somatic mutations that confer increased self-renewal capacity and myeloid differentiation bias, giving rise to mutant myeloid progeny with enhanced pro-inflammatory potential, as compared to their counterparts from normal HSPC clones. This condition, which - in the absence of apparent hematological malignancy - is designated clonal hematopoiesis of indeterminate potential (CHIP) has been epidemiologically and experimentally linked with atherosclerotic cardiovascular disease. However, the generation and release of hyper-inflammatory myeloid cells in the context of CHIP may contribute to inflamm-aging and elevate systemic inflammation, which in turn could contribute to the pathogenesis of additional inflammatory pathologies, such as periodontitis and rheumatoid arthritis.

Although the formal definition of CHIP is the presence of expanded blood cell clones harboring a somatic mutation related to a myeloid malignancy without overt hematological symptoms or malignancy 106, individuals with CHIP have an increased risk of developing hematologic malignant disorders, as well as atherosclerosis and diabetes mellitus 146,148152. More recently, CHIP in postmenopausal women was associated with natural (that is, non-surgical) premature menopause 153. Moreover, CHIP is suspected to also contribute to aging-related inflammatory disorders 51 (Figure 4). In fact, the “indeterminate potential” part of the term denotes the uncertainty of how CHIP may develop clinically, in part because distinct environmental stress factors (e.g., an inflammatory bone marrow microenvironment) may be needed to cooperate with the underlying somatic mutations for the emergence of pathological phenotypes 51,154156.

By definition, CHIP mutations are detected at a variant allele frequency (VAF) of 2% or greater. At this consensus cutoff value, CHIP occurs in >10% of individuals older than 65 with progressively increased prevalence with further aging 52,106. CHIP-related mutations mostly involve disruptive somatic lesions (nonsynonymous, nonsense, frameshift, or splice-site disruption) in the affected genes 148. Using an ultra-sensitive sequencing method, a recent study detected CHIP in >20% of individuals 60 to 69 years of age and in 3% of individuals aged 20 to 29 years 157. Although CHIP prevalence is minimal in young ages, the risk it confers to young individuals for progression of hematologic malignancies and cardiovascular disease is similar to that in the elderly 106.

The most common CHIP-related mutations affect three genes that encode epigenetic modifiers, namely, TET2 (Ten-eleven translocation methylcytosine dioxygenase 2), DNMT3A (DNA methyltransferase 3A), and ASXL1 (Associated sex combs-like 1) 105,106,146,148,158. Together, mutations in TET2 and DNMT3A, which mediate cytosine methylation and demethylation, respectively, represent approximately two thirds of acquired CHIP mutations. The mutant alleles usually present with loss of function 52,106,146,148,158,159.

As TET2 functions as a demethylase160162, loss-of-function mutations cause hypermethylation of DNA at sites involved in gene expression regulation, such as transcription factor–binding sites 160,161. The epigenetic dysregulation owing to TET2 mutations leads to survival advantage of clonal HSCs, which also are skewed towards the myeloid lineage 163168. When the function of DNMT3A is impaired by inactivating CHIP-related mutations, all hematopoietic lineages appear to be affected in humans; consistent with this observation, genetic ablation of Dnmt3a in mice leads to elevated risk of both myeloid and lymphoid malignancies 168172. Moreover, hematopoietic-specific deletion of Asxl1 in mice results in cytopenias affecting all lineages and 173 loss-of-function mutations in ASXL1 predominantly affect the trimethylation of lysine 27 of histone 3 174.

Aging HSPCs with loss-of-function mutations in TET2 display enhanced self-renewal and preferential commitment to the myeloid lineage, giving rise to mature myeloid cells with enhanced proinflammatory activity 149,163166,175. An inflammatory environment in the bone marrow, due to inflamm-aging or underlying inflammatory comorbidities, favors the expansion of CHIP clones 155. Specifically, both IL-6 and TNF support the survival of TET2-deficient HSPCs 176,177. In contrast, under similar inflammatory conditions, TET2-sufficient HSPCs are suppressed and/or are led to exhaustion 51,104,176178. In other words, TET2-mutant clones appear to have a greater fitness over normal clones in an inflammatory bone marrow environment. Moreover, the increased proliferation of CHIP-mutant HSPCs in an inflammatory environment associated with atherosclerosis accelerates the emergence of somatic CHIP-driver mutations, hence further promoting CHIP 179. Overall, inflammation in the bone marrow may promote the emergence of CHIP mutant clones, potentially predisposing to the increased risk for pathological conditions in CHIP carriers 176178.

5. Maladaptive trained immunity and periodontitis-associated comorbidities

As discussed above, TII is thought to represent an evolutionarily conserved beneficial response that can protect the host from re-infection (with the same pathogen that elicited TII in the first place), as well as from unrelated pathogens that are encountered for the first time 50,103,116,125127. Consistent with this notion, TII is also present in primitive organisms 103,180. However, in the setting of modern challenges in western-type societies, TII could also promote aberrant immune responses that may potentially aggravate immune-driven pathologies (‘maladaptive’ TII), thus contributing to the prevalence and severity of chronic inflammatory disorders 51,116,129,181,182. Maladaptive TII may be induced not only by infections or purified microbial molecules but also by an obesogenic diet 183. In this regard, oxidized low-density lipoprotein, as well as other unrelated endogenous factors (e.g., uric acid/monosodium urate crystals [inflammasome activators], and the High-Mobility-Group Box 1 protein [activator of Toll-like receptors and the receptor for advanced glycation end products]) can induce TII 184.

5.1. Implications from cardio-metabolic disease

The concept of maladaptive TII is supported by a preclinical study which modelled metabolic syndrome-induced inflammation by feeding low-density lipoprotein receptor-deficient mice a calorie-rich, high-fat western-type diet. The study showed that the western-type diet induces NLRP3 inflammasome– and IL-1–dependent epigenetic reprogramming of bone marrow progenitor cells (including GMPs), which in turn drive trained myelopoiesis that perpetuates systemic inflammation 129 (Figure 5). In line with the long-lasting effects of TII, this maladaptive reprogramming of bone marrow progenitor cells persisted even after the animals were switched back to a healthy (conventional chow) diet 129. From a clinical standpoint, chronic metabolic-inflammatory disorders, including T2D, obesity and atherosclerosis can skew hematopoiesis toward inflammatory myelopoiesis, in turn leading to chronic leukocytosis that may promote propagation and chronification of these inflammatory disorders 51,132,133,152,185.

Figure 5. Western diet leads to NLRP3 Inflammasome activation and induction of maladaptive trained immunity.

Figure 5.

In mice, western-type diet, in great part through oxidized LDL, induces activation of the NLRP3 inflammasome and caspase 1-dependent secretion of IL-1β, which induces expansion and epigenetic reprogramming of bone marrow progenitor cells (including GMPs), which in turn drive trained myelopoiesis (increased numbers of myeloid cells with enhanced immune responsiveness) that can perpetuate systemic inflammation. These maladaptive alterations persist even after the mice are switched back to a healthy diet.

It can be envisioned that the inflammation-induced epigenetic rewiring of HSPCs towards heightened myelopoiesis might perpetuate inflammation leading to the generation of a feed-forward loop between the bone marrow and cardiometabolic disease, such as atherosclerosis, or a chronic inflammatory disorder, in general, such as periodontitis 49,51,132,133,185 (Figure 2). In other words, elevated numbers of circulating inflammatory myeloid cells can augment inflammation in peripheral tissues, which reciprocally can perpetuate HSPC-mediated myelopoiesis 133,185189.

It also follows that the maladaptive training of HSPCs may constitute mechanistic grounds for comorbidities, such as the increased risk of cardiovascular complications in patients with other types of chronic inflammation, such as periodontitis or rheumatoid arthritis 190,191. In the latter regard, experimental arthritis-associated systemic inflammation down-regulates cholesterol export genes (Apoe, Abca1, and Abcg1) and hence elevates the content of membrane cholesterol of HSPCs in the bone marrow of mice; this in turn promotes HSPC expansion and augmented myelopoiesis 192. This is in part because cholesterol accumulation alters the cell membrane properties, thereby leading to enhanced surface abundance of CD131 (the common β-subunit of the IL-3- and GM-CSF-receptor) and increased downstream signaling, which drives myelopoiesis 112,193. Importantly, apolipoprotein E-deficient (Apoe−/−) mice subjected to K/BxN-induced arthritis exhibit increased entry of Ly6Chi monocytes into atherosclerotic lesions (thus elevating the macrophage burden), as compared to non-arthritic Apoe−/− controls 192. This study therefore suggests that the arthritis-associated elevation of circulating myeloid cells fuels the increased abundance of macrophages in atheromas 192. Consistent with these findings, RA patients display systemic inflammation and increased numbers of circulating myeloid cells including CD16+ monocytes as well as decreased mRNA expression of cholesterol efflux genes (ABCA1 and ABCG1) in peripheral blood mononuclear cells, as compared to healthy controls 192,194,195.

In a similar context, a study utilizing positron emission tomography/computed tomography with 2-deoxy-2-[fluorine-18]fluoro-D-glucose (18F-FDG-PET/CT; more on this technique below) showed that a subset of rheumatoid arthritis patients in clinical remission exhibit elevated metabolic activity in the bone marrow and enhanced inflammation in the arterial wall associated with a promigratory phenotype (as assessed by enhanced expression of activation and pro-adhesive markers) of circulating monocytes 196. Thus, although 40% of patients with rheumatoid arthritis display remission of the disease, the remission does not necessarily normalize their CVD risk 196. These findings moreover suggest a common underlying pathophysiological mechanism for RA and CVD that might involve innate immune memory in the bone marrow. Such inflammatory memory may sustain enhanced myelopoiesis in arthritis patients while in remission, increasing their risk for CVD. Interestingly in this regard, rheumatoid arthritis patients have two- to three-fold higher CVD risk, which is only partly attributed to known traditional risk factors for CVD 197,198. Moreover, in these patients, arthritis activity and duration are correlated with enhanced inflammation in their arterial walls 196. Conversely, as alluded to above, cardiometabolic conditions can also affect hematopoiesis and potentially TII 129,131,133,152,199 and could thus affect the development of (or exacerbation of existing) arthritis.

5.2. Inflammatory periodontitis-bone marrow axis and comorbidities

Although there is currently no direct experimental evidence that periodontitis causes maladaptive training of HSPCs in the bone marrow or, conversely, that periodontitis is affected by the state of the bone marrow, clinical observations are consistent with such notions.

Recent studies have attempted to correlate periodontal inflammation with inflammation in extraoral sites using 18F-FDG-PET/CT 200,201, a technique that can identify sites of inflammatory tissue activity 202,203. 18F-FDG, a radiolabeled analogue of glucose, is taken up by the cells via cell membrane glucose transporters and subsequently phosphorylated intracellularly by hexokinase. The ability of 18F-FDG-PET/CT to localize to sites of inflammation is largely attributed to the increased glycolytic activity (high-level expression of glucose transporters and hexokinase activity) of the cells involved in the inflammatory response and particularly neutrophils and monocyte/macrophages 202,203. Importantly, 18F-FDG uptake in periodontal tissue correlates not only with periodontal inflammation (as evaluated by magnetic resonance imaging) but also with the severity of PD as inferred indirectly by the extent of radiographic bone loss in the inflamed area 204.

Using 18F-FDG-PET/CT, another recent study demonstrated a correlation between the metabolic/inflammatory activities of the periodontal tissue and the bone marrow hematopoietic tissue 200. Moreover, by the same technique, periodontal metabolic/inflammatory activity was associated with arterial inflammation, as assessed by 18F-FDG uptake 200,201 or histologically by evaluating macrophage infiltration in excised carotid artery plaques 201. High-dose atorvastatin therapy reduced periodontal inflammation after a 12-week treatment and the changes in periodontal inflammation correlated with changes in carotid inflammation, as evaluated (in both conditions) by 18F-FDG-PET/CT 205. Importantly, a more recent study using the same cohort as the above-discussed report200 showed that 18F-FDG-PET/CT-determined periodontal inflammation was independently associated with both increased arterial inflammation and risk of subsequent cardiovascular events 206. Bone marrow hematopoietic activity was further associated with C-reactive protein, white blood cell count and absolute monocyte count (through not with erythrocyte-related parameters) 200. Together, these studies suggest associations between bone marrow hematopoietic tissue activity and distinct inflammatory comorbidities, although causality and directionality are uncertain regarding these parameters.

It could be argued that sustained chronic inflammation (owing to periodontitis or other systemic disease) may not provide a TII-inducing stimulus for the bone marrow analogous to that induced by defined agonists of TII such as β-glucan or the BCG vaccine, which are administered in an ‘acute’ rather than a ‘chronic’ manner. It should be noted, however, that the chronic nature of periodontitis does not necessarily represent a continual pathologic process but may rather constitute a persistent series of short-lived acute insults (bursts) separated by periods of remission 207210. Similarly, a key characteristic of periodontitis-associated inflammatory disorders, such as rheumatoid arthritis and inflammatory bowel disease, are flares and exacerbations that are also separated by intervals of remission 211213. Therefore, short-term periods of increased inflammation (‘inflammatory attacks’), such as those involved in the induction of TII, are consistent with this burst-like model of chronic inflammatory diseases.

5.3. Trained myeloid cells and periodontitis-associated comorbidities

Monocytes or neutrophils deriving from the blood of periodontal disease patients display an inherent hyper-reactivity in that they elicit higher levels of proinflammatory cytokines when they are activated ex vivo by inflammatory stimuli, such as LPS or whole bacteria, as compared to the same cell types isolated from the peripheral blood of healthy individuals 214,215. Interestingly, this hyper-reactivity can persist even after the patients undergo successful non-surgical periodontal therapy; specifically, the enhanced cytokine-inducing capacity of peripheral blood myeloid cells from periodontitis patients could be observed for at least 2 months (time interval for post-treatment review) 214,215. These findings resemble recent data from individuals treated with TII-inducing agents, such as the BCG vaccine 118,127.

In this regard, BCG vaccination in healthy volunteers resulted not only in an ‘immediate’ (2 weeks post-vaccination) hyper-reactivity of circulating monocytes in terms of proinflammatory cytokine production upon ex vivo stimulation with unrelated bacterial and fungal pathogens, but also in long-term hyper-reactivity. Indeed, the enhanced cytokine-inducing capacity (upon ex vivo stimulation) of circulating monocytes from BCG vaccinated individuals persisted for at least 3 months after vaccination and was accompanied by higher surface expression of activation markers, such as Toll-like receptor 4 and the Mac-1 integrin 127. This enhanced hyper-reactivity could be associated with the fact that BCG vaccination imprinted persistent epigenetic, transcriptional, and functional adaptations in bone marrow HSPCs toward enhanced myelopoiesis 118. Consistent with the notion that the TII phenotype is initiated at the level of progenitors and is passed on to their progeny, similar epigenetic changes in inflammation-associated loci were also observed in peripheral blood CD14+ monocytes of the vaccinated individuals 118. Moreover, peripheral blood mononuclear cells from patients with coronary artery disease exhibit heightened cytokine production (as compared with the same cells from healthy individuals) after ex vivo stimulation; importantly, transcriptome analysis of HSPCs and GMPs from these patients revealed enrichment for neutrophil- and monocyte-related pathways, indicating that the progenitors are skewed to differentiate into the myeloid lineage 216. In general, peripheral blood monocytes isolated from patients with different autoimmune or inflammatory diseases display characteristics consistent with a trained phenotype, such as enhanced responsiveness to ex vivo stimulation, enhanced glycolysis, and epigenetic rewiring 217.

On the premise that periodontal disease may influence hematopoietic tissue inflammation 200, it could be reasoned that, in periodontitis patients, peripheral blood myeloid cells are in a ‘trained’ state, which endows them with hyper-reactivity (compared to cells from healthy controls) to inflammatory stimuli. This trained state of peripheral myeloid cells of periodontitis patients, which may be maintained even after periodontal therapy, might represent a form of long-lasting innate immune memory imprinted in HSPCs in an inflammatory bone marrow environment 50,112,118 (Figure 2). In fact, the recent association of periodontitis with the severity of COVID-19 disease (increased risk for admission to intensive care unit, necessity of assisted ventilation and death) 218 might, in part, be due to a trained/hyper-responsive state of periodontal patients’ myeloid cells. This notion is in keeping with the observation that, in comparison with COVID-19 patients without periodontitis, COVID-19 patients with periodontitis exhibited increased levels of systemic inflammatory markers and white blood cell counts 218. The latter may imply increased myelopoiesis, but further tests would be required to implicate trained myelopoiesis.

In addition to monocytes/macrophages, neutrophils and granulocytes in general are also targets of TII 118,124. Ligature-induced periodontitis in mice increases the neutrophil counts in the bone marrow and enhances the neutrophil response to subsequent acute infectious peritonitis, both quantitatively and qualitatively (enhanced proinflammatory potential), suggesting that periodontitis can enhance the severity of other inflammatory conditions 219. Whether the enhanced responsiveness of the neutrophils involves TII in the bone marrow was not addressed. However, consistent with this preclinical observation, experimental gingivitis in human volunteers for 3 weeks increased the ex vivo inflammatory responsiveness of blood neutrophils 219. By comparing the HSPC transcriptome of individuals before and 3 months after BCG vaccination, a recent study demonstrated upregulation of genes associated with granulocytic lineage priming, thus suggesting that BCG immunization skews the myeloid cell lineage toward enhanced granulopoiesis 118. Interestingly, peripheral blood neutrophils from periodontitis patients display a Type I IFN signature (elevated expression of IFN-stimulated genes relative to neutrophils from healthy individuals), associated with increased plasma concentrations of IFNα, as compared to those of periodontally healthy controls 220. Moreover, the periodontitis-associated peripheral blood neutrophils exhibit a hyper-responsive phenotype ex vivo with regards to ROS production when primed with type-I IFNs (albeit not by LPS), which induce an mRNA expression profile in neutrophils similar to that observed in vivo 220. The authors concluded that type-I IFNs may be a factor determining the hyper-reactive phenotype of periodontitis patients’ peripheral blood neutrophils. It is intriguing to speculate that this type-I IFN-associated hyper-responsiveness of peripheral blood neutrophils from periodontitis patients might arise from trained granulopoiesis in the bone marrow.

The aforementioned notion is supported indirectly by studies in mice. Specifically, mice systemically treated with fungal-derived β-glucan exhibit type I IFN-dependent transcriptomic and epigenetic rewiring of bone marrow granulopoietic progenitors (also designated as trained granulopoiesis), giving rise to neutrophils with augmented ROS-dependent tumor-killing capacity 124. Single-cell epigenetic analysis demonstrated that the neutrophils generated from trained granulopoiesis exhibited both enhanced Type I IFN-signaling and increased chromatin accessibility in the genes encoding the ROS-producing molecules neutrophil cytosolic factors 1 and 2, suggesting that this type I IFN signature is sustained at the epigenetic level and passed on to the progeny 124. Thus, it is possible that trained granulopoiesis induced by different inflammatory TII agonists (as documented in BCG-vaccinated humans 221 or upon β-glucan administration in mice 124) or by systemic inflammation (owing to periodontitis or other inflammatory condition) might promote the generation of a hyper-reactive neutrophil phenotype with Type I IFN signature and ability for enhanced production of ROS upon rechallenge. Such recall responses can be either protective (e.g., in tumor immunity or in acute infections) or destructive (e.g., in periodontitis and inflammatory diseases in general). It is noteworthy that feeding mice with western-type diet also leads to a strong Type I IFN signature in bone marrow granulopoietic progenitors (GMPs) 129. Given that different inflammatory stimuli induce bone marrow progenitor training via Type I IFN signaling, this pathway merits further investigation for its role in mediating maladaptive training of bone marrow progenitors towards perpetuation of inflammation that could contribute to the emergence of periodontitis-associated comorbidities.

From the above, it can be reasoned that systemic inflammation associated with a certain chronic inflammatory disorder, such as periodontitis, may cause maladaptive training of hematopoietic progenitors, resulting in augmented myelopoiesis and production of ‘trained’ inflammatory myeloid cells, which in turn can populate remote organs and tissues, including the periodontium, where they can enhance local inflammation (Figure 2). This adaptation can be epigenetically regulated and thus can be sustained for a long time, during which HSPCs can influence (through their myeloid progeny) the development or course of different inflammatory disorders that emerge as comorbidities.

6. CHIP and chronic inflammation in periodontitis and other conditions

The concept that CHIP-related loss-of-function mutations are associated with human disease is not adequately explored with the exception of myeloid malignancies and cardiometabolic conditions 146,149,175. Conceivably, CHIP may promote increased susceptibility to different chronic inflammatory pathologies, such as periodontitis and rheumatoid arthritis (Figure 4). To substantiate this notion, it would be instructive to first discuss the mechanisms by which CHIP promotes cardiometabolic inflammation.

Most investigations on CHIP-associated cardiovascular risk in humans have primarily focused on mutations in DNMT3A and TET2; however, only TET2 has been examined thoroughly in preclinical animal models. These studies offered causal evidence linking TET2 loss-of-function and exacerbated atherosclerosis 149,155,175,222. Low-density lipoprotein receptor–deficient (Ldlr–/–) mice that were transplanted with bone marrow cells bearing homozygous or heterozygous null mutations in the Tet2 gene exhibited preferential expansion of the mutant clones and accelerated atherosclerotic lesions 149,175. As with human TET2-associated CHIP 168,223, even if only a fraction (10%) of transplanted HSCs bore the TET2 deficiency, the recipient mice still exhibited preferential clonal expansion of Tet2–/– hematopoietic cells and accelerated atherosclerosis 149.

In the aforementioned preclinical studies, the atherosclerotic lesions contained macrophages with increased production of proinflammatory cytokines, including IL-1β and IL-6 (as compared to Ldlr–/– controls receiving wild-type bone marrow cells) 149,175. Consistently, activated TET2-deficient macrophages produce and release increased levels of IL-1β owing to both increased Il1b transcription and augmented expression and activity of the NLRP3 inflammasome 149,224. Similarly, activated TET2-deficient macrophages produce elevated amounts of IL-6. The TET2 deficiency-associated unrestrained IL-1β and IL-6 production are irrespective of the catalytic activity of TET2 but may rather involve histone deacetylation (TET2 associates with certain histone deacetylases and facilitates their function) 149,225. Furthermore, TET2-associated CHIP in mice results in elevated macrophage-derived IL-1β in white adipose tissue and worsens insulin resistance in obesogenic diet-fed mice or in old mice 226, in line with the observational association between CHIP and Type 2 diabetes in humans 151 These findings provide proof-of-concept for a causal role of CHIP in human coronary heart disease and diabetes.

Cancer-free individuals with CHIP (due to mutations in TET2, DNMT3A or ASXL1) have normal total and differential leukocyte counts 158,175. Consistently, Ldlr–/– mice transplanted with TET2-deficient bone marrow cells have normal blood cell counts despite a mild myeloid skewing 149,175. Thus, what appears to cause pathology in CHIP-related atherosclerosis (and likely in other conditions associated with CHIP) is not a quantitative but a qualitative alteration, that is, the dominance of clones; which give rise to myeloid progeny with heightened proinflammatory potential. As these hyper-inflammatory myeloid cells circulate and populate different tissues including the periodontium, CHIP is likely to be associated with different inflammatory conditions, including periodontitis (Figure 4). Therefore, future epidemiologic and mechanistic studies are warranted to explore the full spectrum of pathologies that can be influenced by CHIP.

The therapeutic targeting of inflammatory cytokines, such as IL-6 and IL-1β, may inhibit the detrimental effects of CHIP on cardiometabolic and other chronic inflammatory disorders. IL-1β, in particular, is involved in multiple processes, including the innate immune training of HSPCs in the bone marrow 112,227, augmented myelopoiesis in CVD 152,228,229, and in CHIP-associated exacerbated atherosclerotic inflammation 149,175. The successful application of IL-1β blockade (using canakinumab) in the CANTOS trial for the treatment of atherosclerosis is consistent with the above-mentioned effects of IL-1β 230. Importantly, patients with TET2 mutant clones respond more favorably to the IL-1β-blocking treatment than patients with non-TET2 mutant CHIP clones 231. This is likely because TET2-related CHIP is significantly associated with elevated IL-1β, although in general CHIP is linked with increased IL-6 concentrations 159. That IL-6 may mediate CHIP-related pathology is further suggested by a longitudinal cohort study. Patients with DNMT3A- or TET2- associated CHIP clones were shown to have elevated risk of incident CVD; however, a subset of patients who additionally carry a mutation in the IL-6 receptor (which affects its surface expression; p.Asp358Ala) presented with fewer CVD events 232.

7. Summary and perspective

Periodontitis-associated systemic inflammation may potentially contribute to the inflammatory adaptation of HSPCs, a process that underlies the induction of TII, a non-specific epigenetic memory that promotes amplified responses by HSPCs and their progeny to future infectious or inflammatory challenges 51,114 (Figure 1). In this manner, TII and the generated trained/hyper-reactive myeloid cells could play a role in the chronicity of periodontitis by setting off a vicious cycle of reciprocally reinforced interactions between the periodontium and the bone marrow (Figure 2). As the trained monocytes/neutrophils are recruited to different tissues or organs and have increased propensity for activation in response to local challenges, TII is likely to be a common mechanistic basis for the development of comorbidities. In a similar vein, the ability of CHIP-mutant HSCs to give rise to hyper-inflammatory myeloid cells should elevate systemic and peripheral inflammation that may contribute not only to the pathogenesis of atherosclerotic CVD 149,175 but perhaps also to other inflammatory comorbid disorders, such as periodontitis and arthritis (Figure 2).

Future research should strive for a better patho-mechanistic understanding of the intertwining between periodontitis and linked comorbidities in the context of TII (Figure 3) and CHIP (Figure 4), two non-mutually exclusive states associated with elevated inflammatory potential in innate immune cells. Inhibiting key cytokines involved in these processes, such as IL-1β and IL-6, may mitigate the risk of multiple comorbid pathologies. For instance, blocking IL-1β may potentially interfere with maladaptive training of bone marrow HSPCs and thereby disrupt an inflammatory vicious cycle perpetuating different inflammatory comorbidities 51,112, as well as the detrimental effects of hyperinflammatory myeloid cells derived from CHIP-mutant HSPCs 149,175. Blocking potent inflammatory cytokines requires caution, as such treatments may interfere with the ability of the host to respond effectively to potentially life-threatening infections. However, for treatments that aim to manipulate TII in the bone marrow, the specificity of the treatment and mitigation of potential side effects could be achieved by using nanoparticles with high avidity for the bone marrow. This technology may facilitate targeted delivery of therapeutic compounds (inhibitors or agonists) to the appropriate cell type for modulating TII in a manner that is protective for the patient 233,234.

Acknowledgements

The authors’ research is supported by grants from the U.S. National Institutes of Health (DE024153, DE029436 to GH; DE026152 and DE028561 to GH and TC; U01DE025046 to KD) and the German Research Foundation (SFB-TR 127 and SFB1181 to TC). The figures were created using Biorender.com.

References

  • 1.Lamont RJ, Koo H, Hajishengallis G. The oral microbiota: dynamic communities and host interactions. Nat Rev Microbiol 2018;16(12):745–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hajishengallis G The inflammophilic character of the periodontitis-associated microbiota. Mol Oral Microbiol 2014;29(6):248–257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Tonetti MS, Jepsen S, Jin L, Otomo-Corgel J. Impact of the global burden of periodontal diseases on health, nutrition and wellbeing of mankind: A call for global action. J Clin Periodontol 2017;44(5):456–462. [DOI] [PubMed] [Google Scholar]
  • 4.Flemmig TF, Beikler T. Economics of periodontal care: market trends, competitive forces and incentives. Periodontol 2000 2013;62(1):287–304. [DOI] [PubMed] [Google Scholar]
  • 5.Bernabe E, Marcenes W, Hernandez CR, et al. Global, Regional, and National Levels and Trends in Burden of Oral Conditions from 1990 to 2017: A Systematic Analysis for the Global Burden of Disease 2017 Study. J Dent Res 2020;99(4):362–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Holmstrup P, Damgaard C, Olsen I, et al. Comorbidity of periodontal disease: two sides of the same coin? An introduction for the clinician. J Oral Microbiol 2017;9(1):1332710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Potempa J, Mydel P, Koziel J. The case for periodontitis in the pathogenesis of rheumatoid arthritis. Nat Rev Rheumatol 2017;13(10):606–620. [DOI] [PubMed] [Google Scholar]
  • 8.Ganzetti G, Campanati A, Santarelli A, et al. Involvement of the oral cavity in psoriasis: results of a clinical study. Br J Dermatol 2015;172(1):282–285. [DOI] [PubMed] [Google Scholar]
  • 9.Vavricka SR, Manser CN, Hediger S, et al. Periodontitis and Gingivitis in Inflammatory Bowel Disease: A Case–Control Study. Inflamm Bowel Dis 2013;19(13):2768–2777. [DOI] [PubMed] [Google Scholar]
  • 10.Persson GR, Berglund J, Persson RE, Renvert S. Prediction of hip and hand fractures in older persons with or without a diagnosis of periodontitis. Bone 2011;48(3):552–556. [DOI] [PubMed] [Google Scholar]
  • 11.Huang Y-F, Chang C-T, Liu S-P, et al. The Impact of Oral Hygiene Maintenance on the Association Between Periodontitis and Osteoporosis: A Nationwide Population-Based Cross Sectional Study. Medicine 2016;95(6):e2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Acharya C, Sahingur SE, Bajaj JS. Microbiota, cirrhosis, and the emerging oral-gut-liver axis. JCI Insight 2017;2(19). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Schenkein HA, Papapanou PN, Genco R, Sanz M. Mechanisms underlying the association between periodontitis and atherosclerotic disease. Periodontol 2000 2020;83(1):90–106. [DOI] [PubMed] [Google Scholar]
  • 14.Dutzan N, Kajikawa T, Abusleme L, et al. A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans. Sci Transl Med 2018;10(463):eaat0797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Moutsopoulos NM, Zerbe CS, Wild T, et al. Interleukin-12 and Interleukin-23 Blockade in Leukocyte Adhesion Deficiency Type 1. N Engl J Med 2017;376(12):1141–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat Rev Drug Discov 2012;11(10):763–776. [DOI] [PubMed] [Google Scholar]
  • 17.Abraham C, Cho JH. IL-23 and autoimmunity: new insights into the pathogenesis of inflammatory bowel disease. Annu Rev Med 2009;60:97–110. [DOI] [PubMed] [Google Scholar]
  • 18.Duerr RH, Taylor KD, Brant SR, et al. A Genome-Wide Association Study Identifies L23R as an Inflammatory Bowel Disease Gene. Science 2006;314(5804):1461–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Leonardi CL, Kimball AB, Papp KA, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 2008;371(9625):1665–1674. [DOI] [PubMed] [Google Scholar]
  • 20.Genco RJ, Borgnakke WS. Risk factors for periodontal disease. Periodontol 2000 2013;62(1):59–94. [DOI] [PubMed] [Google Scholar]
  • 21.Schaefer AS, Bochenek G, Jochens A, et al. Genetic Evidence for PLASMINOGEN as a Shared Genetic Risk Factor of Coronary Artery Disease and Periodontitis. Circulation: Cardiovascular Genetics 2015;8(1):159–167. [DOI] [PubMed] [Google Scholar]
  • 22.Geismar K, Enevold C, Sørensen LK, et al. Involvement of Interleukin-1 Genotypes in the Association of Coronary Heart Disease With Periodontitis. J Periodontol 2008;79(12):2322–2330. [DOI] [PubMed] [Google Scholar]
  • 23.Knight ET, Liu J, Seymour GJ, Faggion CM Jr., Cullinan MP. Risk factors that may modify the innate and adaptive immune responses in periodontal diseases. Periodontol 2000 2016;71(1):22–51. [DOI] [PubMed] [Google Scholar]
  • 24.Divaris K, Monda KL, North KE, et al. Exploring the genetic basis of chronic periodontitis: a genome-wide association study. Hum Mol Genet 2013;22(11):2312–2324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Humphrey LL, Fu R, Buckley DI, Freeman M, Helfand M. Periodontal disease and coronary heart disease incidence: a systematic review and meta-analysis. J Gen Intern Med 2008;23(12):2079–2086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rodríguez-Lozano B, González-Febles J, Garnier-Rodríguez JL, et al. Association between severity of periodontitis and clinical activity in rheumatoid arthritis patients: a case–control study. Arthritis Res Ther 2019;21(1):27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Suma S, Naito M, Wakai K, et al. Tooth loss and pneumonia mortality: A cohort study of Japanese dentists. PLoS One 2018;13(4):e0195813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Skudutyte-Rysstad R, Slevolden EM, Hansen BF, Sandvik L, Preus HR. Association between moderate to severe psoriasis and periodontitis in a Scandinavian population. BMC Oral Health 2014;14(1):139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Rydén L, Buhlin K, Ekstrand E, et al. Periodontitis Increases the Risk of a First Myocardial Infarction. Circulation 2016;133(6):576–583. [DOI] [PubMed] [Google Scholar]
  • 30.Hansen GM, Egeberg A, Holmstrup P, Hansen PR. Relation of Periodontitis to Risk of Cardiovascular and All-Cause Mortality (from a Danish Nationwide Cohort Study). Am J Cardiol 2016;118(4):489–493. [DOI] [PubMed] [Google Scholar]
  • 31.Beukers NG, van der Heijden GJ, van Wijk AJ, Loos BG. Periodontitis is an independent risk indicator for atherosclerotic cardiovascular diseases among 60 174 participants in a large dental school in the Netherlands. J Epidemiol Community Health 2017;71(1):37–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Helenius-Hietala J, Suominen AL, Ruokonen H, et al. Periodontitis is associated with incident chronic liver disease—A population-based cohort study. Liver International 2019;39(3):583–591. [DOI] [PubMed] [Google Scholar]
  • 33.Genco RJ, Sanz M. Clinical and public health implications of periodontal and systemic diseases: An overview. Periodontol 2000 2020;83(1):7–13. [DOI] [PubMed] [Google Scholar]
  • 34.D’Aiuto F, Gkranias N, Bhowruth D, et al. Systemic effects of periodontitis treatment in patients with type 2 diabetes: a 12 month, single-centre, investigator-masked, randomised trial. Lancet Diabetes Endocrinol 2018;6(12):954–965. [DOI] [PubMed] [Google Scholar]
  • 35.Tonetti MS, D’Aiuto F, Nibali L, et al. Treatment of periodontitis and endothelial function. N Engl J Med 2007;356(9):911–920. [DOI] [PubMed] [Google Scholar]
  • 36.Desvarieux M, Demmer RT, Jacobs DR, Papapanou PN, Sacco RL, Rundek T. Changes in clinical and microbiological periodontal profiles relate to progression of carotid intima-media thickness: the Oral Infections and Vascular Disease Epidemiology study. J Am Heart Assoc 2013;2(6):e000254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kaur S, Bright R, Proudman SM, Bartold PM. Does periodontal treatment influence clinical and biochemical measures for rheumatoid arthritis? A systematic review and meta-analysis. Sem Arthritis Rheum 2014;44(2):113–122. [DOI] [PubMed] [Google Scholar]
  • 38.Offenbacher S, Beck JD, Moss K, et al. Results from the Periodontitis and Vascular Events (PAVE) Study: a pilot multicentered, randomized, controlled trial to study effects of periodontal therapy in a secondary prevention model of cardiovascular disease. J Periodontol 2009;80(2):190–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Paraskevas S, Huizinga JD, Loos BG. A systematic review and meta-analyses on C-reactive protein in relation to periodontitis. J Clin Periodontol 2008;35(4):277–290. [DOI] [PubMed] [Google Scholar]
  • 40.Al-Katma MK, Bissada NF, Bordeaux JM, Sue J, Askari AD. Control of Periodontal Infection Reduces the Severity of Active Rheumatoid Arthritis. J Clin Rheumatol 2007;13(3):134–137. [DOI] [PubMed] [Google Scholar]
  • 41.Ortiz P, Bissada NF, Palomo L, et al. Periodontal Therapy Reduces the Severity of Active Rheumatoid Arthritis in Patients Treated With or Without Tumor Necrosis Factor Inhibitors. J Periodontol 2009;80(4):535–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Khare N, Vanza B, Sagar D, Saurav K, Chauhan R, Mishra S. Nonsurgical Periodontal Therapy decreases the Severity of Rheumatoid Arthritis: A Case-control Study. J Contemp Dent Pract 2016;17(6):484–488. [DOI] [PubMed] [Google Scholar]
  • 43.D’Aiuto F, Orlandi M, Gunsolley JC. Evidence that periodontal treatment improves biomarkers and CVD outcomes. J Clin Periodontol 2013;40 Suppl 14:S85–105. [DOI] [PubMed] [Google Scholar]
  • 44.Bokhari SAH, Khan AA, Butt AK, et al. Non-surgical periodontal therapy reduces coronary heart disease risk markers: a randomized controlled trial. J Clin Periodontol 2012;39(11):1065–1074. [DOI] [PubMed] [Google Scholar]
  • 45.Tonetti MS. Periodontitis and risk for atherosclerosis: an update on intervention trials. J Clin Periodontol 2009;36 Suppl 10:15–19. [DOI] [PubMed] [Google Scholar]
  • 46.Türer ÇC, Durmuş D, Balli U, Güven B. Effect of Non-Surgical Periodontal Treatment on Gingival Crevicular Fluid and Serum Endocan, Vascular Endothelial Growth Factor-A, and Tumor Necrosis Factor-Alpha Levels. J Periodontol 2017;88(5):493–501. [DOI] [PubMed] [Google Scholar]
  • 47.Bajaj JS, Matin P, White MB, et al. Periodontal therapy favorably modulates the oral-gut-hepatic axis in cirrhosis. Am J Physiol Gastrointest Liver Physiol 2018;315(5):G824–G837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Genco RJ, Graziani F, Hasturk H. Effects of periodontal disease on glycemic control, complications, and incidence of diabetes mellitus. Periodontol 2000 2020;83(1):59–65. [DOI] [PubMed] [Google Scholar]
  • 49.Hajishengallis G, Chavakis T. Local and systemic mechanisms linking periodontal disease and inflammatory comorbidities. Nat Rev Immunol 2021;21(7):426–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Netea MG, Domínguez-Andrés J, Barreiro LB, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol 2020;20:375–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chavakis T, Mitroulis I, Hajishengallis G. Hematopoietic progenitor cells as integrative hubs for adaptation to and fine-tuning of inflammation. Nat Immunol 2019;20(7):802–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Jaiswal S, Ebert BL. Clonal hematopoiesis in human aging and disease. Science 2019;366(6465). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hutter JW, Velden Uvd, Varoufaki A, Huffels RAM, Hoek FJ, Loos BG. Lower numbers of erythrocytes and lower levels of hemoglobin in periodontitis patients compared to control subjects. J ClinPeriodontol 2001;28(10):930–936. [DOI] [PubMed] [Google Scholar]
  • 54.Kweider M, Lowe GDO, Murray GD, Kinane DF, McGowan DA. Dental Disease, Fibrinogen and White Cell Count; Links with Myocardial Infarction? Scott Med J 1993;38(3):73–74. [DOI] [PubMed] [Google Scholar]
  • 55.Genco RJ, Van Dyke TE. Prevention: Reducing the risk of CVD in patients with periodontitis. Nat Rev Cardiol 2010;7(9):479–480. [DOI] [PubMed] [Google Scholar]
  • 56.Monteiro AM, Jardini MAN, Alves S, et al. Cardiovascular Disease Parameters in Periodontitis. J Periodontol 2009;80(3):378–388. [DOI] [PubMed] [Google Scholar]
  • 57.Buhlin K, Hultin M, Norderyd O, et al. Risk factors for atherosclerosis in cases with severe periodontitis. J Clin Periodontol 2009;36(7):541–549. [DOI] [PubMed] [Google Scholar]
  • 58.Loos BG, Craandijk J, Hoek FJ, Dillen PMEW-v, Van Der Velden U. Elevation of Systemic Markers Related to Cardiovascular Diseases in the Peripheral Blood of Periodontitis Patients. J Periodontol 2000;71(10):1528–1534. [DOI] [PubMed] [Google Scholar]
  • 59.Nibali L, Darbar U, Rakmanee T, Donos N. Anemia of inflammation associated with periodontitis: Analysis of two clinical studies. J Periodontol 2019;90(11):1252–1259. [DOI] [PubMed] [Google Scholar]
  • 60.Nibali L, D’Aiuto F, Griffiths G, Patel K, Suvan J, Tonetti MS. Severe periodontitis is associated with systemic inflammation and a dysmetabolic status: a case–control study. J Clin Periodontol 2007;34(11):931–937. [DOI] [PubMed] [Google Scholar]
  • 61.Anand PS, Sagar DK, Ashok S, Kamath KP. Association of aggressive periodontitis with reduced erythrocyte counts and reduced hemoglobin levels. J Periodont Res 2014;49(6):719–728. [DOI] [PubMed] [Google Scholar]
  • 62.Kwon Y-J, Jeon K-J, Chung T-H, Lee Y-J. Elevated leukocyte count is associated with periodontitis in Korean adults: the 2012–2014 KNHANES. Oral Dis 2017;23(2):241–246. [DOI] [PubMed] [Google Scholar]
  • 63.Ebersole JL, Machen RL, Steffen MJ, WIillmann DE. Systemic acute-phase reactants, C-reactive protein and haptoglobin, in adult periodontitis. Clin Exp Immunol 1997;107(2):347–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Slade GD, Offenbacher S, Beck JD, Heiss G, Pankow JS. Acute-phase Inflammatory Response to Periodontal Disease in the US Population. J Dent Res 2000;79(1):49–57. [DOI] [PubMed] [Google Scholar]
  • 65.Andrukhov O, Ulm C, Reischl H, Nguyen PQ, Matejka M, Rausch-Fan X. Serum Cytokine Levels in Periodontitis Patients in Relation to the Bacterial Load. J Periodontol 2011;82(6):885–892. [DOI] [PubMed] [Google Scholar]
  • 66.Duarte PM, da Rocha M, Sampaio E, et al. Serum Levels of Cytokines in Subjects With Generalized Chronic and Aggressive Periodontitis Before and After Non-Surgical Periodontal Therapy: A Pilot Study. J Periodontol 2010;81(7):1056–1063. [DOI] [PubMed] [Google Scholar]
  • 67.Schenkein HA, Koertge TE, Brooks CN, Sabatini R, Purkall DE, Tew JG. IL-17 in Sera from Patients with Aggressive Periodontitis. J Dent Res 2010;89(9):943–947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Thorat M, Pradeep AR, Garg G. Correlation of levels of oncostatin M cytokine in crevicular fluid and serum in periodontal disease. Int J Oral Sci 2010;2(4):198–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zekeridou A, Mombelli A, Cancela J, Courvoisier D, Giannopoulou C. Systemic inflammatory burden and local inflammation in periodontitis: What is the link between inflammatory biomarkers in serum and gingival crevicular fluid? Clin Exp Dent Res 2019;5(2):128–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Georgiou AC, Crielaard W, Armenis I, de Vries R, van der Waal SV. Apical Periodontitis Is Associated with Elevated Concentrations of Inflammatory Mediators in Peripheral Blood: A Systematic Review and Meta-analysis. J Endodont 2019. [DOI] [PubMed] [Google Scholar]
  • 71.de Oliveira C, Watt R, Hamer M. Toothbrushing, inflammation, and risk of cardiovascular disease: results from Scottish Health Survey. BMJ 2010;340:c2451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mendes VS, Cota LOM, Costa AA, Oliveira AMSD, Costa FO. Periodontitis as another comorbidity associated with psoriasis: A case-control study. J Periodontol 2019;90(4):358–366. [DOI] [PubMed] [Google Scholar]
  • 73.Ide M, Harris M, Stevens A, et al. Periodontitis and Cognitive Decline in Alzheimer’s Disease. PLoS One 2016;11(3):e0151081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kamer AR, Pirraglia E, Tsui W, et al. Periodontal disease associates with higher brain amyloid load in normal elderly. Neurobiology of Aging 2015;36(2):627–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Fuggle NR, Smith TO, Kaul A, Sofat N. Hand to Mouth: A Systematic Review and Meta-Analysis of the Association between Rheumatoid Arthritis and Periodontitis. Front Immunol 2016;7:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Grau AJ, Becher H, Ziegler CM, et al. Periodontal Disease as a Risk Factor for Ischemic Stroke. Stroke 2004;35(2):496–501. [DOI] [PubMed] [Google Scholar]
  • 77.Joshipura KJ, Hung H-C, Rimm EB, Willett WC, Ascherio A. Periodontal Disease, Tooth Loss, and Incidence of Ischemic Stroke. Stroke 2003;34(1):47–52. [DOI] [PubMed] [Google Scholar]
  • 78.Zhao D, Zhen Z, Pelekos G, Yiu KH, Jin L. Periodontal disease increases the risk for onset of systemic comorbidities in dental hospital attendees: An 18-year retrospective cohort study. J Periodontol 2019;90(3):225–233. [DOI] [PubMed] [Google Scholar]
  • 79.Lee KH, Choi YY. Rheumatoid arthritis and periodontitis in adults: using the Korean National Health Insurance Service – National Sample Cohort. J Periodontol 2020;91(9):1186–1193. [DOI] [PubMed] [Google Scholar]
  • 80.Lee J-H, Lee J-S, Park J-Y, et al. Association of Lifestyle-Related Comorbidities With Periodontitis: A Nationwide Cohort Study in Korea. Medicine 2015;94(37):e1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Khader Y, Khassawneh B, Obeidat B, et al. Periodontal Status of Patients With Metabolic Syndrome Compared to Those Without Metabolic Syndrome. J Periodontol 2008;79(11):2048–2053. [DOI] [PubMed] [Google Scholar]
  • 82.Yu H-C, Chen T-P, Chang Y-C. Inflammatory bowel disease as a risk factor for periodontitis under Taiwanese National Health Insurance Research database. J Dent Sci 2018;13(3):242–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Papageorgiou SN, Hagner M, Nogueira AVB, Franke A, Jäger A, Deschner J. Inflammatory bowel disease and oral health: systematic review and a meta-analysis. J Clin Periodontol 2017;44(4):382–393. [DOI] [PubMed] [Google Scholar]
  • 84.Mercado FB, Marshall RI, Klestov AC, Bartold PM. Relationship Between Rheumatoid Arthritis and Periodontitis. J Periodontol 2001;72(6):779–787. [DOI] [PubMed] [Google Scholar]
  • 85.Havemose-Poulsen A, Westergaard J, Stoltze K, et al. Periodontal and Hematological Characteristics Associated With Aggressive Periodontitis, Juvenile Idiopathic Arthritis, and Rheumatoid Arthritis. J Periodontol 2006;77(2):280–288. [DOI] [PubMed] [Google Scholar]
  • 86.Mikuls TR, Payne JB, Yu F, et al. Periodontitis and Porphyromonas gingivalis in patients with rheumatoid arthritis. Arthritis Rheumatol 2014;66(5):1090–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pink C, Kocher T, Meisel P, et al. Longitudinal effects of systemic inflammation markers on periodontitis. J Clin Periodontol 2015;42(11):988–997. [DOI] [PubMed] [Google Scholar]
  • 88.Dominy SS, Lynch C, Ermini F, et al. Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv 2019;5(1):eaau3333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Katz J, Chegini N, Shiverick KT, Lamont RJ. Localization of P. gingivalis in preterm delivery placenta. J Dent Res 2009;88(6):575–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Vander Haar EL, So J, Gyamfi-Bannerman C, Han YW. Fusobacterium nucleatum and adverse pregnancy outcomes: Epidemiological and mechanistic evidence. Anaerobe 2018;50:55–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Haraszthy VI, Zambon JJ, Trevisan M, Zeid M, Genco RJ. Identification of periodontal pathogens in atheromatous plaques. J Periodontol 2000;71(10):1554–1560. [DOI] [PubMed] [Google Scholar]
  • 92.Kozarov EV, Dorn BR, Shelburne CE, Dunn WA Jr., Progulske-Fox A Human atherosclerotic plaque contains viable invasive Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis. Arterioscler Thromb Vasc Biol 2005;25(3):e17–e18. [DOI] [PubMed] [Google Scholar]
  • 93.Hujoel PP, White BA, García RI, Listgarten MA. The dentogingival epithelial surface area revisited. J Periodont Res 2001;36(1):48–55. [DOI] [PubMed] [Google Scholar]
  • 94.Friedewald VE, Kornman KS, Beck JD, et al. The American Journal of Cardiology and Journal of Periodontology Editors’ Consensus: periodontitis and atherosclerotic cardiovascular disease. Am J Cardiol 2009;104(1):59–68. [DOI] [PubMed] [Google Scholar]
  • 95.Silver JG, Martin AW, McBride BC. Experimental transient bacteraemias in human subjects with varying degrees of plaque accumulation and gingival inflammation. J Clin Periodontol 1977;4(2):92–99. [DOI] [PubMed] [Google Scholar]
  • 96.Scannapieco FA. Position paper of The American Academy of Periodontology: periodontal disease as a potential risk factor for systemic diseases. J Periodontol 1998;69(7):841–850. [PubMed] [Google Scholar]
  • 97.Lafaurie GI, Mayorga-Fayad I, Torres MF, et al. Periodontopathic microorganisms in peripheric blood after scaling and root planing. J Clin Periodontol 2007;34(10):873–879. [DOI] [PubMed] [Google Scholar]
  • 98.Horliana AC, Chambrone L, Foz AM, et al. Dissemination of periodontal pathogens in the bloodstream after periodontal procedures: a systematic review. PLoS One 2014;9(5):e98271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Garlet GP. Destructive and protective roles of cytokines in periodontitis: A re-appraisal from host defense and tissue destruction viewpoints. J Dent Res 2010;89(12):1349–1363. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang Z, Yuan W, Deng J, et al. Granulocyte colony stimulating factor (G-CSF) regulates neutrophils infiltration and periodontal tissue destruction in an experimental periodontitis. Molecular Immunology 2020;117:110–121. [DOI] [PubMed] [Google Scholar]
  • 101.Silva N, Abusleme L, Bravo D, et al. Host response mechanisms in periodontal diseases. J Appl Oral Sci 2015;23(3):329–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Slots J Human viruses in periodontitis. Periodontol 2000 2010;53:89–110. [DOI] [PubMed] [Google Scholar]
  • 103.Penkov S, Mitroulis I, Hajishengallis G, Chavakis T. Immunometabolic Crosstalk: An Ancestral Principle of Trained Immunity? Trends Immunol 2019;40(1):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kovtonyuk LV, Fritsch K, Feng X, Manz MG, Takizawa H. Inflamm-Aging of Hematopoiesis, Hematopoietic Stem Cells, and the Bone Marrow Microenvironment. Front Immunol 2016;7:502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Jaiswal S, Libby P. Clonal haematopoiesis: connecting ageing and inflammation in cardiovascular disease. Nat Rev Cardiol 2020;17(3):137–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Steensma DP. Clinical Implications of Clonal Hematopoiesis. Mayo Clinic Proceedings 2018;93(8):1122–1130. [DOI] [PubMed] [Google Scholar]
  • 107.King KY, Goodell MA. Inflammatory modulation of HSCs: viewing the HSC as a foundation for the immune response. Nat Rev Immunol 2011;11:685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Oguro H, Ding L, Morrison SJ. SLAM family markers resolve functionally distinct subpopulations of hematopoietic stem cells and multipotent progenitors. Cell Stem Cell 2013;13(1):102–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Manz MG, Boettcher S. Emergency granulopoiesis. Nat Rev Immunol 2014;14(5):302–314. [DOI] [PubMed] [Google Scholar]
  • 110.Yamamoto R, Morita Y, Ooehara J, et al. Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells. Cell 2013;154(5):1112–1126. [DOI] [PubMed] [Google Scholar]
  • 111.Kaufmann E, Sanz J, Dunn JL, et al. BCG Educates Hematopoietic Stem Cells to Generate Protective Innate Immunity against Tuberculosis. Cell 2018;172(1–2):176–190 e119. [DOI] [PubMed] [Google Scholar]
  • 112.Mitroulis I, Ruppova K, Wang B, et al. Modulation of Myelopoiesis Progenitors Is an Integral Component of Trained Immunity. Cell 2018;172(1–2):147–161 e112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.de Laval B, Maurizio J, Kandalla PK, et al. C/EBPβ-Dependent Epigenetic Memory Induces Trained Immunity in Hematopoietic Stem Cells. Cell Stem Cell 2020. [DOI] [PubMed] [Google Scholar]
  • 114.Netea MG, Joosten LA, Latz E, et al. Trained immunity: A program of innate immune memory in health and disease. Science 2016;352(6284):aaf1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Goodridge HS, Ahmed SS, Curtis N, et al. Harnessing the beneficial heterologous effects of vaccination. Nat Rev Immunol 2016;16(6):392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hajishengallis G, Li X, Mitroulis I, Chavakis T. Trained Innate Immunity and Its Implications for Mucosal Immunity and Inflammation. Adv Exp Med Biol 2019;1197:11–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Yona S, Kim KW, Wolf Y, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 2013;38(1):79–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Cirovic B, de Bree LCJ, Groh L, et al. BCG Vaccination in Humans Elicits Trained Immunity via the Hematopoietic Progenitor Compartment. Cell Host & Microbe 2020;28(2):322–334.e325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bekkering S, Arts RJW, Novakovic B, et al. Metabolic Induction of Trained Immunity through the Mevalonate Pathway. Cell 2018;172(1–2):135–146 e139. [DOI] [PubMed] [Google Scholar]
  • 120.Arts RJ, Joosten LA, Netea MG. Immunometabolic circuits in trained immunity. Semin Immunol 2016;28(5):425–430. [DOI] [PubMed] [Google Scholar]
  • 121.Saeed S, Quintin J, Kerstens HH, et al. Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity. Science 2014;345(6204):1251086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Cheng SC, Quintin J, Cramer RA, et al. mTOR- and HIF-1alpha-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 2014;345(6204):1250684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Divangahi M, Aaby P, Khader SA, et al. Trained immunity, tolerance, priming and differentiation: distinct immunological processes. Nat Immunol 2021;22(1):2–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kalafati L, Kourtzelis I, Schulte-Schrepping J, et al. Innate Immune Training of Granulopoiesis Promotes Anti-tumor Activity. Cell 2020;183(3):771–785.e712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Quintin J, Saeed S, Martens JHA, et al. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe 2012;12(2):223–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Arts RJW, Moorlag S, Novakovic B, et al. BCG Vaccination Protects against Experimental Viral Infection in Humans through the Induction of Cytokines Associated with Trained Immunity. Cell Host Microbe 2018;23(1):89–100 e105. [DOI] [PubMed] [Google Scholar]
  • 127.Kleinnijenhuis J, Quintin J, Preijers F, et al. Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc Natl Acad Sci U S A 2012;109(43):17537–17542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Netea MG, Schlitzer A, Placek K, Joosten LAB, Schultze JL. Innate and Adaptive Immune Memory: an Evolutionary Continuum in the Host’s Response to Pathogens. Cell Host Microbe 2019;25(1):13–26. [DOI] [PubMed] [Google Scholar]
  • 129.Christ A, Gunther P, Lauterbach MAR, et al. Western Diet Triggers NLRP3-Dependent Innate Immune Reprogramming. Cell 2018;172(1–2):162–175 e114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Leentjens J, Bekkering S, Joosten LAB, Netea MG, Burgner DP, Riksen NP. Trained Innate Immunity as a Novel Mechanism Linking Infection and the Development of Atherosclerosis. Circ Res 2018;122(5):664–669. [DOI] [PubMed] [Google Scholar]
  • 131.Bekkering S, Saner C, Riksen NP, et al. Trained Immunity: Linking Obesity and Cardiovascular Disease across the Life-Course? Trends Endocrinol Metab 2020;31(5):378–389. [DOI] [PubMed] [Google Scholar]
  • 132.Barrett TJ, Murphy AJ, Goldberg IJ, Fisher EA. Diabetes-mediated myelopoiesis and the relationship to cardiovascular risk. Ann N Y Acad Sci 2017;1402(1):31–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Nahrendorf M Myeloid cell contributions to cardiovascular health and disease. Nat Med 2018;24(6):711–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Aging Hajishengallis G. and its impact on innate immunity and inflammation: Implications for periodontitis. J Oral Biosci 2014;56:30–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Ebersole JL, Graves CL, Gonzalez OA, et al. Aging, inflammation, immunity and periodontal disease. Periodontol 2000 2016;72(1):54–75. [DOI] [PubMed] [Google Scholar]
  • 136.Geiger H, de Haan G, Florian MC. The ageing haematopoietic stem cell compartment. Nat Rev Immunol 2013;13(5):376–389. [DOI] [PubMed] [Google Scholar]
  • 137.Dorshkind K, Montecino-Rodriguez E, Signer RA. The ageing immune system: is it ever too old to become young again? Nat Rev Immunol 2009;9(1):57–62. [DOI] [PubMed] [Google Scholar]
  • 138.Weyand CM, Goronzy JJ. Aging of the Immune System. Mechanisms and Therapeutic Targets. Annals of the American Thoracic Society 2016;13(Supplement_5):S422–S428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Franceschi C, Bonafe M, Valensin S, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci 2000;908:244–254. [DOI] [PubMed] [Google Scholar]
  • 140.Cevenini E, Monti D, Franceschi C. Inflamm-ageing. Curr Opin Clin Nutr Metab Care 2013;16(1):14–20. [DOI] [PubMed] [Google Scholar]
  • 141.Liberale L, Montecucco F, Tardif JC, Libby P, Camici GG. Inflamm-ageing: the role of inflammation in age-dependent cardiovascular disease. Eur Heart J 2020;41(31):2974–2982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Wikby A, Nilsson BO, Forsey R, et al. The immune risk phenotype is associated with IL-6 in the terminal decline stage: findings from the Swedish NONA immune longitudinal study of very late life functioning. Mech Ageing Dev 2006;127(8):695–704. [DOI] [PubMed] [Google Scholar]
  • 143.Ferrucci L, Corsi A, Lauretani F, et al. The origins of age-related proinflammatory state. Blood 2005;105(6):2294–2299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Furman D, Chang J, Lartigue L, et al. Expression of specific inflammasome gene modules stratifies older individuals into two extreme clinical and immunological states. Nat Med 2017;23(2):174–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci 2014;69 Suppl 1:S4–9. [DOI] [PubMed] [Google Scholar]
  • 146.Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014;371(26):2488–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Gutierrez-Martinez P, Hogdal L, Nagai M, et al. Diminished apoptotic priming and ATM signalling confer a survival advantage onto aged haematopoietic stem cells in response to DNA damage. Nat Cell Biol 2018;20(4):413–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Genovese G, Kähler AK, Handsaker RE, et al. Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence. N Engl J Med 2014;371(26):2477–2487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Fuster JJ, MacLauchlan S, Zuriaga MA, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science 2017;355(6327):842–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Dorsheimer L, Assmus B, Rasper T, et al. Association of Mutations Contributing to Clonal Hematopoiesis With Prognosis in Chronic Ischemic Heart Failure. JAMA Cardiol 2019;4(1):25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Lee MKS, Dragoljevic D, Bertuzzo Veiga C, Wang N, Yvan-Charvet L, Murphy AJ. Interplay between Clonal Hematopoiesis of Indeterminate Potential and Metabolism. Trends Endocrinol Metab 2020. [DOI] [PubMed] [Google Scholar]
  • 152.Mitroulis I, Hajishengallis G, Chavakis T. Trained Immunity and Cardiometabolic Disease: The Role of Bone Marrow. Arterioscler Thromb Vasc Biol 2021;41(1):48–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Honigberg MC, Zekavat SM, Niroula A, et al. Premature Menopause, Clonal Hematopoiesis, and Coronary Artery Disease in Postmenopausal Women. Circulation 2021;143(5):410–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Zhang CRC, Nix D, Gregory M, et al. Inflammatory cytokines promote clonal hematopoiesis with specific mutations in ulcerative colitis patients. Experimental Hematology 2019;80:36–41.e33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Cook EK, Luo M, Rauh MJ. Clonal hematopoiesis and inflammation: Partners in leukemogenesis and comorbidity. Exp Hematol 2020. [DOI] [PubMed] [Google Scholar]
  • 156.Meisel M, Hinterleitner R, Pacis A, et al. Microbial signals drive pre-leukaemic myeloproliferation in a Tet2-deficient host. Nature 2018;557(7706):580–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Acuna-Hidalgo R, Sengul H, Steehouwer M, et al. Ultra-sensitive Sequencing Identifies High Prevalence of Clonal Hematopoiesis-Associated Mutations throughout Adult Life. Am J Hum Genet 2017;101(1):50–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Busque L, Patel JP, Figueroa ME, et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat Genet 2012;44:1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Bick AG, Weinstock JS, Nandakumar SK, et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 2020;586(7831):763–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Rasmussen KD, Jia G, Johansen JV, et al. Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis. Genes Dev 2015;29(9):910–922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Yamazaki J, Jelinek J, Lu Y, et al. TET2 Mutations Affect Non-CpG Island DNA Methylation at Enhancers and Transcription Factor–Binding Sites in Chronic Myelomonocytic Leukemia. Cancer Res 2015;75(14):2833–2843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Nakajima H, Kunimoto H. TET2 as an epigenetic master regulator for normal and malignant hematopoiesis. Cancer Sci 2014;105(9):1093–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Delhommeau F, Dupont S, Valle VD, et al. Mutation in TET2 in Myeloid Cancers. N Engl J Med 2009;360(22):2289–2301. [DOI] [PubMed] [Google Scholar]
  • 164.Quivoron C, Couronné L, Della Valle V, et al. TET2 Inactivation Results in Pleiotropic Hematopoietic Abnormalities in Mouse and Is a Recurrent Event during Human Lymphomagenesis. Cancer Cell 2011;20(1):25–38. [DOI] [PubMed] [Google Scholar]
  • 165.Moran-Crusio K, Reavie L, Shih A, et al. Tet2 Loss Leads to Increased Hematopoietic Stem Cell Self-Renewal and Myeloid Transformation. Cancer Cell 2011;20(1):11–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ko M, Bandukwala HS, An J, et al. Ten-Eleven-Translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice. Proc Natl Acad Sci U S A 2011;108(35):14566–14571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Mas-Peiro S, Hoffmann J, Fichtlscherer S, et al. Clonal haematopoiesis in patients with degenerative aortic valve stenosis undergoing transcatheter aortic valve implantation. Eur Heart J 2019;41(8):933–939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Buscarlet M, Provost S, Zada YF, et al. Lineage restriction analyses in CHIP indicate myeloid bias for TET2 and multipotent stem cell origin for DNMT3A. Blood 2018;132(3):277–280. [DOI] [PubMed] [Google Scholar]
  • 169.Guryanova OA, Shank K, Spitzer B, et al. DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling. Nat Med 2016;22(12):1488–1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Cole CB, Russler-Germain DA, Ketkar S, et al. Haploinsufficiency for DNA methyltransferase 3A predisposes hematopoietic cells to myeloid malignancies. J Clin Invest 2017;127(10):3657–3674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Celik H, Mallaney C, Kothari A, et al. Enforced differentiation of Dnmt3a-null bone marrow leads to failure with c-Kit mutations driving leukemic transformation. Blood 2015;125(4):619–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Mayle A, Yang L, Rodriguez B, et al. Dnmt3a loss predisposes murine hematopoietic stem cells to malignant transformation. Blood 2015;125(4):629–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Abdel-Wahab O, Gao J, Adli M, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med 2013;210(12):2641–2659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Abdel-Wahab O, Adli M, LaFave LM, et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell 2012;22(2):180–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Jaiswal S, Natarajan P, Silver AJ, et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N Engl J Med 2017;377(2):111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Cai Z, Kotzin JJ, Ramdas B, et al. Inhibition of Inflammatory Signaling in Tet2 Mutant Preleukemic Cells Mitigates Stress-Induced Abnormalities and Clonal Hematopoiesis. Cell Stem Cell 2018;23(6):833–849.e835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Abegunde SO, Buckstein R, Wells RA, Rauh MJ. An inflammatory environment containing TNFα favors Tet2-mutant clonal hematopoiesis. Exp Hematol 2018;59:60–65. [DOI] [PubMed] [Google Scholar]
  • 178.Craver BM, El Alaoui K, Scherber RM, Fleischman AG. The Critical Role of Inflammation in the Pathogenesis and Progression of Myeloid Malignancies. Cancers 2018;10(4):104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Heyde A, Rohde D, McAlpine CS, et al. Increased stem cell proliferation in atherosclerosis accelerates clonal hematopoiesis. Cell 2021;184(5):1348–1361.e1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Netea MG, van der Meer JW. Trained Immunity: An Ancient Way of Remembering. Cell Host Microbe 2017;21(3):297–300. [DOI] [PubMed] [Google Scholar]
  • 181.Wendeln AC, Degenhardt K, Kaurani L, et al. Innate immune memory in the brain shapes neurological disease hallmarks. Nature 2018;556(7701):332–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Norden DM, Muccigrosso MM, Godbout JP. Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease. Neuropharmacology 2015;96(Pt A):29–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Charles-Messance H, Sheedy FJ. Train to Lose: Innate Immune Memory in Metaflammation. Molecular Nutrition & Food Research 2020;n/a(n/a):1900480. [DOI] [PubMed] [Google Scholar]
  • 184.Crișan TO, Netea MG, Joosten LAB. Innate immune memory: Implications for host responses to damage-associated molecular patterns. Eur J Immunol 2016;46(4):817–828. [DOI] [PubMed] [Google Scholar]
  • 185.Murphy AJ, Tall AR. Disordered haematopoiesis and athero-thrombosis. Eur Heart J 2016;37(14):1113–1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Dutta P, Courties G, Wei Y, et al. Myocardial infarction accelerates atherosclerosis. Nature 2012;487(7407):325–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Dutta P, Sager HB, Stengel KR, et al. Myocardial Infarction Activates CCR2(+) Hematopoietic Stem and Progenitor Cells. Cell Stem Cell 2015;16(5):477–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Leuschner F, Rauch PJ, Ueno T, et al. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J Exp Med 2012;209(1):123–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Tall AR, Yvan-Charvet L. Cholesterol, inflammation and innate immunity. Nat Rev Immunol 2015;15(2):104–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Kaplan MJ. Cardiovascular Complications of Rheumatoid Arthritis: Assessment, Prevention, and Treatment. Rheumatic Disease Clinics 2010;36(2):405–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Hajishengallis G Periodontitis: from microbial immune subversion to systemic inflammation. Nat Rev Immunol 2015;15(1):30–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Dragoljevic D, Kraakman MJ, Nagareddy PR, et al. Defective cholesterol metabolism in haematopoietic stem cells promotes monocyte-driven atherosclerosis in rheumatoid arthritis. Eur Heart J 2018;39(23):2158–2167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Yvan-Charvet L, Pagler T, Gautier EL, et al. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science 2010;328(5986):1689–1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Coulthard LR, Geiler J, Mathews RJ, et al. Differential effects of infliximab on absolute circulating blood leucocyte counts of innate immune cells in early and late rheumatoid arthritis patients. Clin Exp Immunol 2012;170(1):36–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Klimek E, Mikołajczyk T, Sulicka J, et al. Blood monocyte subsets and selected cardiovascular risk markers in rheumatoid arthritis of short duration in relation to disease activity. Biomed Res Int 2014;2014:736853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Bernelot Moens SJ, van der Valk FM, Strang AC, et al. Unexpected arterial wall and cellular inflammation in patients with rheumatoid arthritis in remission using biological therapy: a cross-sectional study. Arthritis Res Ther 2016;18(1):115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Baghdadi LR, Woodman RJ, Shanahan EM, Mangoni AA. The impact of traditional cardiovascular risk factors on cardiovascular outcomes in patients with rheumatoid arthritis: a systematic review and meta-analysis. PLoS One 2015;10(2):e0117952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Hoogeveen RM, Nahrendorf M, Riksen NP, et al. Monocyte and haematopoietic progenitor reprogramming as common mechanism underlying chronic inflammatory and cardiovascular diseases. Eur Heart J 2018;39(38):3521–3527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Schloss MJ, Swirski FK, Nahrendorf M. Modifiable Cardiovascular Risk, Hematopoiesis, and Innate Immunity. Circ Res 2020;126(9):1242–1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Ishai A, Osborne MT, El Kholy K, et al. Periodontal Disease Associates With Arterial Inflammation Via Potentiation of a Hematopoietic-Arterial Axis. JACC Cardiovasc Imaging 2019;12((11 Pt 1)):2271–2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Fifer KM, Qadir S, Subramanian S, et al. Positron Emission Tomography Measurement of Periodontal 18F-Fluorodeoxyglucose Uptake Is Associated With Histologically Determined Carotid Plaque Inflammation. J Amer Coll Cardiol 2011;57(8):971–976. [DOI] [PubMed] [Google Scholar]
  • 202.Glaudemans AW, de Vries EF, Galli F, Dierckx RA, Slart RH, Signore A. The use of (18)F-FDG-PET/CT for diagnosis and treatment monitoring of inflammatory and infectious diseases. Clin Dev Immunol 2013;2013:623036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Treglia G Diagnostic Performance of (18)F-FDG PET/CT in Infectious and Inflammatory Diseases according to Published Meta-Analyses. Contrast Media Mol Imaging 2019;2019:3018349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Kito S, Koga H, Kodama M, et al. Reflection of ¹⁸F-FDG accumulation in the evaluation of the extent of periapical or periodontal inflammation. Oral Surg Oral Med Oral Pathol Oral Radiol 2012;114(6):e62–69. [DOI] [PubMed] [Google Scholar]
  • 205.Subramanian S, Emami H, Vucic E, et al. High-Dose Atorvastatin Reduces Periodontal Inflammation: A Novel Pleiotropic Effect of Statins. J Am Coll Cardiol 2013;62(25):2382–2391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Van Dyke TE, Kholy KE, Ishai A, et al. Inflammation of the periodontium associates with risk of future cardiovascular events. J Periodontol 2021;92(3):348–358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Goodson JM, Tanner AC, Haffajee AD, Sornberger GC, Socransky SS. Patterns of progression and regression of advanced destructive periodontal disease. J Clin Periodontol 1982;9(6):472–481. [DOI] [PubMed] [Google Scholar]
  • 208.Graves DT, Oates T, Garlet GP. Review of osteoimmunology and the host response in endodontic and periodontal lesions. J Oral Microbiol 2011;3: 10.3402/jom.v3403i3400.5304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Gilthorpe MS, Zamzuri AT, Griffiths GS, Maddick IH, Eaton KA, Johnson NW. Unification of the “Burst” and “Linear” Theories of Periodontal Disease Progression: A Multilevel Manifestation of the Same Phenomenon. J Dent Res 2003;82(3):200–205. [DOI] [PubMed] [Google Scholar]
  • 210.Teles R, Moss K, Preisser JS, et al. Patterns of periodontal disease progression based on linear mixed models of clinical attachment loss. J Clin Periodontol 2018;45(1):15–25. [DOI] [PubMed] [Google Scholar]
  • 211.Kristensen SL, Ahlehoff O, Lindhardsen J, et al. Disease activity in inflammatory bowel disease is associated with increased risk of myocardial infarction, stroke and cardiovascular death--a Danish nationwide cohort study. PLoS One 2013;8(2):e56944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Kristensen SL, Lindhardsen J, Ahlehoff O, et al. Increased risk of atrial fibrillation and stroke during active stages of inflammatory bowel disease: a nationwide study. Europace 2014;16(4):477–484. [DOI] [PubMed] [Google Scholar]
  • 213.Myasoedova E, Chandran A, Ilhan B, et al. The role of rheumatoid arthritis (RA) flare and cumulative burden of RA severity in the risk of cardiovascular disease. Ann Rheum Dis 2016;75(3):560–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Ling MR, Chapple IL, Matthews JB. Peripheral blood neutrophil cytokine hyper-reactivity in chronic periodontitis. Innate Immun 2015;21(7):714–725. [DOI] [PubMed] [Google Scholar]
  • 215.Radvar M, Tavakkol-Afshari J, Bajestan MN, Naseh MR, Arab HR. The effect of periodontal treatment on IL-6 production of peripheral blood monocytes in aggressive periodontitis and chronic periodontitis patients. Iran J Immunol 2008;5(2):100–106. [PubMed] [Google Scholar]
  • 216.Noz MP, Bekkering S, Groh L, et al. Reprogramming of bone marrow myeloid progenitor cells in patients with severe coronary artery disease. eLife 2020;9:e60939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Arts RJW, Joosten LAB, Netea MG. The Potential Role of Trained Immunity in Autoimmune and Autoinflammatory Disorders. Front Immunol 2018;9(298). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Marouf N, Cai W, Said KN, et al. Association between periodontitis and severity of COVID-19 infection: A case–control study. J Clin Periodontol 2021;48(4): 483–491 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Fine N, Chadwick JW, Sun C, et al. Periodontal Inflammation Primes the Systemic Innate Immune Response. J Dent Res 2020:22034520963710. [DOI] [PubMed] [Google Scholar]
  • 220.Wright HJ, Matthews JB, Chapple IL, Ling-Mountford N, Cooper PR. Periodontitis associates with a type 1 IFN signature in peripheral blood neutrophils. J Immunol 2008;181(8):5775–5784. [DOI] [PubMed] [Google Scholar]
  • 221.Moorlag SJCFM, Rodriguez-Rosales YA, Gillard J, et al. BCG Vaccination Induces Long-Term Functional Reprogramming of Human Neutrophils. Cell Rep 2020;33(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Amorós-Pérez M, Fuster JJ. Clonal hematopoiesis driven by somatic mutations: A new player in atherosclerotic cardiovascular disease. Atherosclerosis 2020;297:120–126. [DOI] [PubMed] [Google Scholar]
  • 223.Arends CM, Galan-Sousa J, Hoyer K, et al. Hematopoietic lineage distribution and evolutionary dynamics of clonal hematopoiesis. Leukemia 2018;32(9):1908–1919. [DOI] [PubMed] [Google Scholar]
  • 224.Sano S, Oshima K, Wang Y, et al. Tet2-Mediated Clonal Hematopoiesis Accelerates Heart Failure Through a Mechanism Involving the IL-1beta/NLRP3 Inflammasome. J Am Coll Cardiol 2018;71(8):875–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Zhang Q, Zhao K, Shen Q, et al. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature 2015;525:389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Fuster JJ, Zuriaga MA, Zorita V, et al. TET2-Loss-of-Function-Driven Clonal Hematopoiesis Exacerbates Experimental Insulin Resistance in Aging and Obesity. Cell Rep 2020;33(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Hajishengallis G, Li X, Chavakis T. Immunometabolic control of hematopoiesis. Mol Aspects Med 2021;77:100923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Poller WC, Nahrendorf M, Swirski FK. Hematopoiesis and Cardiovascular Disease. Circ Res 2020;126(8):1061–1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Mitroulis I, Kalafati L, Bornhäuser M, Hajishengallis G, Chavakis T. Regulation of the Bone Marrow Niche by Inflammation. Front Immunol 2020;11(1540). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med 2017;377(12):1119–1131. [DOI] [PubMed] [Google Scholar]
  • 231.Svensson EC, Madar A, Campbell CD, et al. TET2-Driven Clonal Hematopoiesis Predicts Enhanced Response to Canakinumab in the CANTOS Trial: An Exploratory Analysis. Circulation 2018;138(Suppl_1):A15111–A15111. [Google Scholar]
  • 232.Bick AG, Pirruccello JP, Griffin GK, et al. Genetic Interleukin 6 Signaling Deficiency Attenuates Cardiovascular Risk in Clonal Hematopoiesis. Circulation 2020;141(2):124–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Priem B, van Leent MMT, Teunissen AJP, et al. Trained Immunity-Promoting Nanobiologic Therapy Suppresses Tumor Growth and Potentiates Checkpoint Inhibition. Cell 2020;183(3):786–801.e719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Mulder WJM, Ochando J, Joosten LAB, Fayad ZA, Netea MG. Therapeutic targeting of trained immunity. Nat Rev Drug Disc 2019;18:553–566. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES